Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
FASEB J ; 35(1): e21092, 2021 01.
Article in English | MEDLINE | ID: mdl-33378124

ABSTRACT

Myosin 18Aα is a myosin 2-like protein containing unique N- and C-terminal protein interaction domains that co-assembles with myosin 2. One protein known to bind to myosin 18Aα is ß-Pix, a guanine nucleotide exchange factor (GEF) for Rac1 and Cdc42 that has been shown to promote dendritic spine maturation by activating the assembly of actin and myosin filaments in spines. Here, we show that myosin 18A⍺ concentrates in the spines of cerebellar Purkinje neurons via co-assembly with myosin 2 and through an actin binding site in its N-terminal extension. miRNA-mediated knockdown of myosin 18A⍺ results in a significant defect in spine maturation that is rescued by an RNAi-immune version of myosin 18A⍺. Importantly, ß-Pix co-localizes with myosin 18A⍺ in spines, and its spine localization is lost upon myosin 18A⍺ knockdown or when its myosin 18A⍺ binding site is deleted. Finally, we show that the spines of myosin 18A⍺ knockdown Purkinje neurons contain significantly less F-actin and myosin 2. Together, these data argue that mixed filaments of myosin 2 and myosin 18A⍺ form a complex with ß-Pix in Purkinje neuron spines that promotes spine maturation by enhancing the assembly of actin and myosin filaments downstream of ß-Pix's GEF activity.


Subject(s)
Dendritic Spines/metabolism , Myosins/metabolism , Purkinje Cells/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Dendritic Spines/genetics , Gene Deletion , Mice , Myosin Type II/genetics , Myosin Type II/metabolism , Myosins/genetics , Rho Guanine Nucleotide Exchange Factors/genetics
2.
Cytoskeleton (Hoboken) ; 74(5): 205-218, 2017 May.
Article in English | MEDLINE | ID: mdl-28329908

ABSTRACT

The peri-centrosomal localization and morphology of the Golgi apparatus depends largely on the microtubule cytoskeleton and the microtubule motor protein dynein. Recent studies proposed that myosin 18Aα (M18Aα) also contributes to Golgi morphology by binding the Golgi protein GOLPH3 and walking along adjacent actin filaments to stretch the Golgi into its classic ribbon structure. Biochemical analyses have shown, however, that M18A is not an actin-activated ATPase and lacks motor activity. Our goal, therefore, was to define the precise molecular mechanism by which M18Aα determines Golgi morphology. We show that purified M18Aα remains inactive in the presence of GOLPH3, arguing against the Golgi-specific activation of the myosin. Using M18A-specific antibodies and expression of GFP-tagged M18Aα, we find no evidence that it localizes to the Golgi. Moreover, several cell lines with reduced or eliminated M18Aα expression exhibited normal Golgi morphology. Interestingly, actin filament disassembly resulted in a marked reduction in lateral stretching of the Golgi in both control and M18Aα-deficient cells. Importantly, this reduction was accompanied by an expansion of the Golgi in the vertical direction, vertical movement of the centrosome, and increases in the height of both the nucleus and the cell. Collectively, our data indicate that M18Aα does not localize to the Golgi or play a significant role in determining its morphology, and suggest that global F-actin disassembly alters Golgi morphology indirectly by altering cell shape.


Subject(s)
Actins/metabolism , Golgi Apparatus/metabolism , Myosins/metabolism , Humans
3.
Nat Cell Biol ; 19(2): 85-93, 2017 02.
Article in English | MEDLINE | ID: mdl-28114272

ABSTRACT

The cellular mechanisms governing non-muscle myosin II (NM2) filament assembly are largely unknown. Using EGFP-NM2A knock-in fibroblasts and multiple super-resolution imaging modalities, we characterized and quantified the sequential amplification of NM2 filaments within lamellae, wherein filaments emanating from single nucleation events continuously partition, forming filament clusters that populate large-scale actomyosin structures deeper in the cell. Individual partitioning events coincide spatially and temporally with the movements of diverging actin fibres, suppression of which inhibits partitioning. These and other data indicate that NM2A filaments are partitioned by the dynamic movements of actin fibres to which they are bound. Finally, we showed that partition frequency and filament growth rate in the lamella depend on MLCK, and that MLCK is competing with centrally active ROCK for a limiting pool of monomer with which to drive lamellar filament assembly. Together, our results provide new insights into the mechanism and spatio-temporal regulation of NM2 filament assembly in cells.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Myosin Light Chains/genetics , Myosin-Light-Chain Kinase/metabolism , Myosins/metabolism , Peptide Fragments/metabolism , Actomyosin/metabolism , Animals , Gene Knock-In Techniques , Mice
4.
Curr Biol ; 25(7): 942-8, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25754640

ABSTRACT

Class-18 myosins are most closely related to conventional class-2 nonmuscle myosins (NM2). Surprisingly, the purified head domains of Drosophila, mouse, and human myosin 18A (M18A) lack actin-activated ATPase activity and the ability to translocate actin filaments, suggesting that the functions of M18A in vivo do not depend on intrinsic motor activity. M18A has the longest coiled coil of any myosin outside of the class-2 myosins, suggesting that it might form bipolar filaments similar to conventional myosins. To address this possibility, we expressed and purified full-length mouse M18A using the baculovirus/Sf9 system. M18A did not form large bipolar filaments under any of the conditions tested. Instead, M18A formed an ∼ 65-nm-long bipolar structure with two heads at each end. Importantly, when NM2 was polymerized in the presence of M18A, the two myosins formed mixed bipolar filaments, as evidenced by cosedimentation, electron microscopy, and single-molecule imaging. Moreover, super-resolution imaging of NM2 and M18A using fluorescently tagged proteins and immunostaining of endogenous proteins showed that NM2 and M18A are present together within individual filaments inside living cells. Together, our in vitro and live-cell imaging data argue strongly that M18A coassembles with NM2 into mixed bipolar filaments. M18A could regulate the biophysical properties of these filaments and, by virtue of its extra N- and C-terminal domains, determine the localization and/or molecular interactions of the filaments. Given the numerous, fundamental cellular and developmental roles attributed to NM2, our results have far-reaching biological implications.


Subject(s)
Cytoskeleton/chemistry , Myosins/metabolism , Animals , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Mice , Microscopy, Electron/methods , Microscopy, Fluorescence/methods , Molecular Imaging/methods , Myosin Type II/metabolism , Protein Isoforms/metabolism
6.
Curr Biol ; 24(10): 1160-6, 2014 May 19.
Article in English | MEDLINE | ID: mdl-24814144

ABSTRACT

Nonmuscle myosin II (NM II) powers myriad developmental and cellular processes, including embryogenesis, cell migration, and cytokinesis [1]. To exert its functions, monomers of NM II assemble into bipolar filaments that produce a contractile force on the actin cytoskeleton. Mammalian cells express up to three isoforms of NM II (NM IIA, IIB, and IIC), each of which possesses distinct biophysical properties and supports unique as well as redundant cellular functions [2-8]. Despite previous efforts [9-13], it remains unclear whether NM II isoforms assemble in living cells to produce mixed (heterotypic) bipolar filaments or whether filaments consist entirely of a single isoform (homotypic). We addressed this question using fluorescently tagged versions of NM IIA, IIB, and IIC, isoform-specific immunostaining of the endogenous proteins, and two-color total internal reflection fluorescence structured-illumination microscopy, or TIRF-SIM, to visualize individual myosin II bipolar filaments inside cells. We show that NM II isoforms coassemble into heterotypic filaments in a variety of settings, including various types of stress fibers, individual filaments throughout the cell, and the contractile ring. We also show that the differential distribution of NM IIA and NM IIB typically seen in confocal micrographs of well-polarized cells is reflected in the composition of individual bipolar filaments. Interestingly, this differential distribution is less pronounced in freshly spread cells, arguing for the existence of a sorting mechanism acting over time. Together, our work argues that individual NM II isoforms are potentially performing both isoform-specific and isoform-redundant functions while coassembled with other NM II isoforms.


Subject(s)
Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Myosin Type II/metabolism , Blotting, Western , Cell Line, Tumor , Cell Movement , Electrophoresis, Polyacrylamide Gel , Female , Humans , Microscopy, Fluorescence , Muscle Contraction , Protein Isoforms/metabolism
7.
Proc Natl Acad Sci U S A ; 111(19): E1970-9, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778263

ABSTRACT

Although capping protein (CP) terminates actin filament elongation, it promotes Arp2/3-dependent actin network assembly and accelerates actin-based motility both in vitro and in vivo. In vitro, capping protein Arp2/3 myosin I linker (CARMIL) antagonizes CP by reducing its affinity for the barbed end and by uncapping CP-capped filaments, whereas the protein V-1/myotrophin sequesters CP in an inactive complex. Previous work showed that CARMIL can readily retrieve CP from the CP:V-1 complex, thereby converting inactive CP into a version with moderate affinity for the barbed end. Here we further clarify the mechanism of this exchange reaction, and we demonstrate that the CP:CARMIL complex created by complex exchange slows the rate of barbed-end elongation by rapidly associating with, and dissociating from, the barbed end. Importantly, the cellular concentrations of V-1 and CP determined here argue that most CP is sequestered by V-1 at steady state in vivo. Finally, we show that CARMIL is recruited to the plasma membrane and only at cell edges undergoing active protrusion. Assuming that CARMIL is active only at this location, our data argue that a large pool of freely diffusing, inactive CP (CP:V-1) feeds, via CARMIL-driven complex exchange, the formation of weak-capping complexes (CP:CARMIL) at the plasma membrane of protruding edges. In vivo, therefore, CARMIL should promote Arp2/3-dependent actin network assembly at the leading edge by promoting barbed-end capping there.


Subject(s)
Actin Capping Proteins/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Kidney/cytology , Mice , Microfilament Proteins , Myosin Type I/metabolism , Polymerization , Pseudopodia/metabolism , Rabbits , Rats
8.
J Biol Chem ; 285(4): 2707-20, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19926785

ABSTRACT

Bulk solution assays have shown that the isolated CARMIL homology 3 (CAH3) domain from mouse and Acanthamoeba CARMIL rapidly and potently restores actin polymerization when added to actin filaments previously capped with capping protein (CP). To demonstrate this putative uncapping activity directly, we used total internal reflection microscopy to observe single, CP-capped actin filaments before and after the addition of the CAH3 domain from mouse CARMIL-1 (mCAH3). The addition of mCAH3 rapidly restored the polymerization of individual capped filaments, consistent with uncapping. To verify uncapping, filaments were capped with recombinant mouse CP tagged with monomeric green fluorescent protein (mGFP-CP). Restoration of polymerization upon the addition of mCAH3 was immediately preceded by the complete dissociation of mGFP-CP from the filament end, confirming the CAH3-driven uncapping mechanism. Quantitative analyses showed that the percentage of capped filaments that uncapped increased as the concentration of mCAH3 was increased, reaching a maximum of approximately 90% at approximately 250 nm mCAH3. Moreover, the time interval between mCAH3 addition and uncapping decreased as the concentration of mCAH3 increased, with the half-time of CP at the barbed end decreasing from approximately 30 min without mCAH3 to approximately 10 s with a saturating amount of mCAH3. Finally, using mCAH3 tagged with mGFP, we obtained direct evidence that the complex of CP and mCAH3 has a small but measurable affinity for the barbed end, as inferred from previous studies and kinetic modeling. We conclude that the isolated CAH3 domain of CARMIL (and presumably the intact molecule as well) possesses the ability to uncap CP-capped actin filaments.


Subject(s)
Acanthamoeba/metabolism , Actin Cytoskeleton/metabolism , Carrier Proteins/metabolism , Protozoan Proteins/metabolism , Acanthamoeba/genetics , Actin Capping Proteins/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Green Fluorescent Proteins/genetics , Mice , Microfilament Proteins , Microscopy/methods , Polymers/metabolism , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
J Biol Chem ; 285(8): 5738-47, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20018884

ABSTRACT

Programs exist for searching protein sequences for potential membrane-penetrating segments (hydrophobic regions) and for lipid-binding sites with highly defined tertiary structures, such as PH, FERM, C2, ENTH, and other domains. However, a rapidly growing number of membrane-associated proteins (including cytoskeletal proteins, kinases, GTP-binding proteins, and their effectors) bind lipids through less structured regions. Here, we describe the development and testing of a simple computer search program that identifies unstructured potential membrane-binding sites. Initially, we found that both basic and hydrophobic amino acids, irrespective of sequence, contribute to the binding to acidic phospholipid vesicles of synthetic peptides that correspond to the putative membrane-binding domains of Acanthamoeba class I myosins. Based on these results, we modified a hydrophobicity scale giving Arg- and Lys-positive, rather than negative, values. Using this basic and hydrophobic scale with a standard search algorithm, we successfully identified previously determined unstructured membrane-binding sites in all 16 proteins tested. Importantly, basic and hydrophobic searches identified previously unknown potential membrane-binding sites in class I myosins, PAKs and CARMIL (capping protein, Arp2/3, myosin I linker; a membrane-associated cytoskeletal scaffold protein), and synthetic peptides and protein domains containing these newly identified sites bound to acidic phospholipids in vitro.


Subject(s)
Acanthamoeba/chemistry , Algorithms , Carrier Proteins/chemistry , Cell Membrane/chemistry , Myosins/metabolism , Protozoan Proteins/chemistry , Sequence Analysis, Protein , Acanthamoeba/genetics , Acanthamoeba/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Hydrophobic and Hydrophilic Interactions , Mice , Myosins/chemistry , Myosins/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
10.
Protein Expr Purif ; 67(2): 113-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19427903

ABSTRACT

Capping protein (CP) is a ubiquitously expressed, heterodimeric actin binding protein that is essential for normal actin dynamics in cells. The existing methods for purifying native CP from tissues and recombinant CP from bacteria are time-consuming processes that involve numerous conventional chromatographic steps and functional assays to achieve a homogeneous preparation of the protein. Here, we report the rapid purification of Acanthamoeba CP from amoeba extracts and recombinant mouse CP from E. coli extracts using as an affinity matrix GST-fusion proteins containing the CP binding site from Acanthamoeba CARMIL and mouse CARMIL-1, respectively. This improved method for CP purification should facilitate the in vitro analysis of CP structure, function, and regulation.


Subject(s)
Actin Capping Proteins/isolation & purification , Carrier Proteins/chemistry , Protozoan Proteins/isolation & purification , Acanthamoeba/chemistry , Actin Capping Proteins/genetics , Actin Capping Proteins/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Extracts/chemistry , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Immunoblotting , Mice , Microfilament Proteins , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sepharose/chemistry
11.
J Biol Chem ; 281(15): 10635-50, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16434392

ABSTRACT

Acanthamoeba CARMIL was previously shown to co-purify with capping protein (CP) and to bind pure CP. Here we show that this interaction inhibits the barbed end-capping activity of CP. Even more strikingly, this interaction drives the uncapping of actin filaments previously capped with CP. These activities are CP-specific; CARMIL does not inhibit the capping activities of either gelsolin or CapG and does not uncap gelsolin-capped filaments. Although full-length (FL) CARMIL (residues 1-1121) possesses both anti-CP activities, C-terminal fragments like glutathione S-transferase (GST)-P (940-1121) that contain the CARMIL CP binding site are at least 10 times more active. We localized the full activities of GST-P to its C-terminal 51 residues (1071-1121). This sequence contains a stretch of 25 residues that is highly conserved in CARMIL proteins from protozoa, flies, worms, and vertebrates (CARMIL Homology domain 3; CAH3). Point mutations showed that the majority of the most highly conserved residues within CAH3 are critical for the anti-CP activity of GST-AP (862-1121). Finally, we found that GST-AP binds CP approximately 20-fold more tightly than does FL-CARMIL. This observation together with the elevated activities of C-terminal fragments relative to FL-CARMIL suggests that FL-CARMIL might exist primarily in an autoinhibited state. Consistent with this idea, proteolytic cleavage of FL-CARMIL with thrombin generated an approximately 14-kDa C-terminal fragment that expresses full anti-CP activities. We propose that, after some type of physiological activation event, FL-CARMIL could function in vivo as a potent CP antagonist. Given the pivotal role that CP plays in determining the global actin phenotype of cells, our results suggest that CARMIL may play an important role in the physiological regulation of actin assembly.


Subject(s)
Actins/chemistry , Carrier Proteins/chemistry , Carrier Proteins/physiology , Acanthamoeba/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/metabolism , Catalysis , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Gelsolin/chemistry , Glutathione Transferase/metabolism , Humans , Microfilament Proteins , Molecular Sequence Data , Myosins/chemistry , Point Mutation , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Thrombin/chemistry , Time Factors
12.
J Biol Chem ; 279(4): 3068-77, 2004 Jan 23.
Article in English | MEDLINE | ID: mdl-14594951

ABSTRACT

CARMIL, also known as Acan 125, is a multidomain protein that was originally identified on the basis of its interaction with the Src homology 3 (SH3) domain of type I myosins from Acanthamoeba. In a subsequent study of CARMIL from Dictyostelium, pull-down assays indicated that the protein also bound capping protein and the Arp2/3 complex. Here we present biochemical evidence that Acanthamoeba CARMIL interacts tightly with capping protein. In biochemical preparations, CARMIL copurified extensively with two polypeptides that were shown by microsequencing to be the alpha- and beta-subunits of Acanthamoeba capping protein. The complex between CARMIL and capping protein, which is readily demonstratable by chemical cross-linking, can be completely dissociated by size exclusion chromatography at pH 5.4. Analytical ultracentrifugation, surface plasmon resonance and SH3 domain pull-down assays indicate that the dissociation constant of capping protein for CARMIL is approximately 0.4 microm or lower. Using CARMIL fusion proteins, the binding site for capping protein was shown to reside within the carboxyl-terminal, approximately 200 residue, proline-rich domain of CARMIL. Finally, chemical cross-linking, analytical ultracentrifugation, and rotary shadowed electron microscopy revealed that CARMIL is asymmetric and that it exists in a monomer <--> dimer equilibrium with an association constant of 1.0 x 10(6) m(-1). Together, these results indicate that CARMIL self-associates and interacts with capping protein with affinities that, given the cellular concentrations of the proteins ( approximately 1 and 2 microm for capping protein and CARMIL, respectively), indicate that both activities should be physiologically relevant.


Subject(s)
Acanthamoeba/metabolism , Carrier Proteins/analysis , Protozoan Proteins/analysis , Animals , Carrier Proteins/chemistry , Carrier Proteins/isolation & purification , Microfilament Proteins/analysis , Microfilament Proteins/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...