Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 263
Filter
1.
PLoS One ; 16(12): e0252845, 2021.
Article in English | MEDLINE | ID: mdl-34962917

ABSTRACT

The cytokinetic contractile ring (CR) was first described some 50 years ago, however our understanding of the assembly and structure of the animal cell CR remains incomplete. We recently reported that mature CRs in sea urchin embryos contain myosin II mini-filaments organized into aligned concatenated arrays, and that in early CRs myosin II formed discrete clusters that transformed into the linearized structure over time. The present study extends our previous work by addressing the hypothesis that these myosin II clusters also contain the crucial scaffolding proteins anillin and septin, known to help link actin, myosin II, RhoA, and the membrane during cytokinesis. Super-resolution imaging of cortices from dividing embryos indicates that within each cluster, anillin and septin2 occupy a centralized position relative to the myosin II mini-filaments. As CR formation progresses, the myosin II, septin and anillin containing clusters enlarge and coalesce into patchy and faintly linear patterns. Our super-resolution images provide the initial visualization of anillin and septin nanostructure within an animal cell CR, including evidence of a septin filament-like network. Furthermore, Latrunculin-treated embryos indicated that the localization of septin or anillin to the myosin II clusters in the early CR was not dependent on actin filaments. These results highlight the structural progression of the CR in sea urchin embryos from an array of clusters to a linearized purse string, the association of anillin and septin with this process, and provide the visualization of an apparent septin filament network with the CR structure of an animal cell.


Subject(s)
Contractile Proteins/metabolism , Cytokinesis , Embryo, Nonmammalian/metabolism , Myosin Type II/metabolism , Sea Urchins/embryology , Sea Urchins/metabolism , Septins/metabolism , Actin Cytoskeleton/metabolism , Animals , Antibodies/metabolism , Contractile Proteins/chemistry , Contractile Proteins/immunology , Imaging, Three-Dimensional , Protein Domains , Septins/immunology , rho GTP-Binding Proteins/metabolism
2.
Biochemistry ; 58(27): 3031-3041, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31243991

ABSTRACT

Division of fungal and animal cells depends on scaffold proteins called anillins. Cytokinesis by the fission yeast Schizosaccharomyces pombe is compromised by the loss of anillin Mid1p (Mid1, UniProtKB P78953 ), because cytokinesis organizing centers, called nodes, are misplaced and fail to acquire myosin-II, so they assemble slowly into abnormal contractile rings. The C-terminal half of Mid1p consists of lipid binding C2 and PH domains, but the N-terminal half (Mid1p-N452) performs most of the functions of the full-length protein. Little is known about the structure of the N-terminal half of Mid1p, so we investigated its physical properties using structure prediction tools, spectroscopic techniques, and hydrodynamic measurements. The data indicate that Mid1p-N452 is intrinsically disordered but moderately compact. Recombinant Mid1p-N452 purified from insect cells was phosphorylated, which weakens its tendency to aggregate. Purified Mid1p-N452 demixes into liquid droplets at concentrations far below its concentration in nodes. These physical properties are appropriate for scaffolding other proteins in nodes.


Subject(s)
Contractile Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/chemistry , Amino Acid Sequence , Contractile Proteins/metabolism , Contractile Proteins/ultrastructure , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/ultrastructure , Models, Molecular , Phase Transition , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/ultrastructure , Solubility
3.
Elife ; 82019 01 31.
Article in English | MEDLINE | ID: mdl-30702429

ABSTRACT

Cellular forces sculpt organisms during development, while misregulation of cellular mechanics can promote disease. Here, we investigate how the actomyosin scaffold protein anillin contributes to epithelial mechanics in Xenopus laevis embryos. Increased mechanosensitive recruitment of vinculin to cell-cell junctions when anillin is overexpressed suggested that anillin promotes junctional tension. However, junctional laser ablation unexpectedly showed that junctions recoil faster when anillin is depleted and slower when anillin is overexpressed. Unifying these findings, we demonstrate that anillin regulates medial-apical actomyosin. Medial-apical laser ablation supports the conclusion that that tensile forces are stored across the apical surface of epithelial cells, and anillin promotes the tensile forces stored in this network. Finally, we show that anillin's effects on cellular mechanics impact tissue-wide mechanics. These results reveal anillin as a key regulator of epithelial mechanics and lay the groundwork for future studies on how anillin may contribute to mechanical events in development and disease.


Subject(s)
Actomyosin/metabolism , Contractile Proteins/metabolism , Epithelial Cells/metabolism , Actins , Adenosine Triphosphate/pharmacology , Animals , Biomechanical Phenomena , Cell Polarity , Contractile Proteins/chemistry , Embryo, Nonmammalian/metabolism , Myosin Type II/metabolism , Protein Binding , Protein Domains , Protein Stability , Vinculin/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism
4.
Proc Natl Acad Sci U S A ; 116(9): 3594-3603, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808751

ABSTRACT

The contractile actin cortex is a thin layer of filamentous actin, myosin motors, and regulatory proteins beneath the plasma membrane crucial to cytokinesis, morphogenesis, and cell migration. However, the factors regulating actin assembly in this compartment are not well understood. Using the Dictyostelium model system, we show that the three Diaphanous-related formins (DRFs) ForA, ForE, and ForH are regulated by the RhoA-like GTPase RacE and synergize in the assembly of filaments in the actin cortex. Single or double formin-null mutants displayed only moderate defects in cortex function whereas the concurrent elimination of all three formins or of RacE caused massive defects in cortical rigidity and architecture as assessed by aspiration assays and electron microscopy. Consistently, the triple formin and RacE mutants encompassed large peripheral patches devoid of cortical F-actin and exhibited severe defects in cytokinesis and multicellular development. Unexpectedly, many forA- /E-/H- and racE- mutants protruded efficiently, formed multiple exaggerated fronts, and migrated with morphologies reminiscent of rapidly moving fish keratocytes. In 2D-confinement, however, these mutants failed to properly polarize and recruit myosin II to the cell rear essential for migration. Cells arrested in these conditions displayed dramatically amplified flow of cortical actin filaments, as revealed by total internal reflection fluorescence (TIRF) imaging and iterative particle image velocimetry (PIV). Consistently, individual and combined, CRISPR/Cas9-mediated disruption of genes encoding mDia1 and -3 formins in B16-F1 mouse melanoma cells revealed enhanced frequency of cells displaying multiple fronts, again accompanied by defects in cell polarization and migration. These results suggest evolutionarily conserved functions for formin-mediated actin assembly in actin cortex mechanics.


Subject(s)
Actin Cytoskeleton/genetics , Carrier Proteins/genetics , Contractile Proteins/genetics , Melanoma, Experimental/genetics , Actin Cytoskeleton/chemistry , Actins/genetics , Animals , CRISPR-Cas Systems , Cell Movement/genetics , Cell Polarity/genetics , Contractile Proteins/chemistry , Dictyostelium/genetics , Disease Models, Animal , Formins , Humans , Melanoma, Experimental/pathology , Mice , Microscopy, Electron , Muscle Contraction/genetics , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
5.
Methods Cell Biol ; 137: 387-394, 2017.
Article in English | MEDLINE | ID: mdl-28065318

ABSTRACT

Cytokinesis is a process by which a mother cell is divided into two daughter cells after chromosome segregation. In both animal and fungal cells, cytokinesis is carried out by the constriction of the contractile ring made up of actin, myosin-II, and other conserved proteins. Detailed genetic and cell biological analysis of cytokinesis has led to the identification of various genes involved in the process of cytokinesis including the cytological description of the process. However, detailed biochemical analysis of the process is lacking. Critical questions that aim to understand aspects, such as the organization of actin and myosin in the contractile ring, the architecture of the ring, and the molecular process of ring contraction, remain unanswered. We have developed a method to address these aspects of cytokinesis. Using the fission yeast Schizosaccharomyces pombe, we present a method whereby cell-ghosts containing functional contractile rings can be isolated and used to perform various biochemical analysis as well as detailed electron microscopy studies.


Subject(s)
Contractile Proteins/isolation & purification , Cytokinesis/genetics , Molecular Biology/methods , Myosin Type II/isolation & purification , Actin Cytoskeleton/chemistry , Actins/chemistry , Actins/isolation & purification , Cell Division/genetics , Contractile Proteins/chemistry , Myosin Type II/chemistry , Myosin Type II/genetics , Schizosaccharomyces/chemistry , Schizosaccharomyces/genetics
6.
Structure ; 24(11): 1972-1983, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27773687

ABSTRACT

The spliceosomal B complex-specific protein Prp38 forms a complex with the intrinsically unstructured proteins MFAP1 and Snu23. Our binding and crystal structure analyses show that MFAP1 and Snu23 contact Prp38 via ER/K motif-stabilized single α helices, which have previously been recognized only as rigid connectors or force springs between protein domains. A variant of the Prp38-binding single α helix of MFAP1, in which ER/K motifs not involved in Prp38 binding were mutated, was less α-helical in isolation and showed a reduced Prp38 affinity, with opposing tendencies in interaction enthalpy and entropy. Our results indicate that the strengths of single α helix-based interactions can be tuned by the degree of helix stabilization in the unbound state. MFAP1, Snu23, and several other spliceosomal proteins contain multiple regions that likely form single α helices via which they might tether several binding partners and act as intermittent scaffolds that facilitate remodeling steps during assembly of an active spliceosome.


Subject(s)
Contractile Proteins/chemistry , Extracellular Matrix Proteins/chemistry , RNA Splicing Factors/chemistry , Ribonucleoproteins, Small Nuclear/chemistry , Spliceosomes/chemistry , Binding Sites , Circular Dichroism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Structure, Secondary , Thermodynamics
7.
Nat Struct Mol Biol ; 22(5): 377-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25822993

ABSTRACT

R-type pyocins are representatives of contractile ejection systems, a class of biological nanomachines that includes, among others, the bacterial type VI secretion system (T6SS) and contractile bacteriophage tails. We report atomic models of the Pseudomonas aeruginosa precontraction pyocin sheath and tube, and the postcontraction sheath, obtained by cryo-EM at 3.5-Å and 3.9-Å resolutions, respectively. The central channel of the tube is negatively charged, in contrast to the neutral and positive counterparts in T6SSs and phage tails. The sheath is interwoven by long N- and C-terminal extension arms emanating from each subunit, which create an extensive two-dimensional mesh that has the same connectivity in the extended and contracted state of the sheath. We propose that the contraction process draws energy from electrostatic and shape complementarities to insert the inner tube through bacterial cell membranes to eventually kill the bacteria.


Subject(s)
Anti-Bacterial Agents/chemistry , Contractile Proteins/ultrastructure , Nanotubes/chemistry , Pseudomonas aeruginosa/pathogenicity , Pyocins/chemistry , Bacterial Proteins/chemistry , Bacterial Secretion Systems , Bacteriophages/chemistry , Cell Membrane/metabolism , Contractile Proteins/chemistry , Crystallography, X-Ray , Microscopy, Electron , Models, Molecular , Protein Structure, Secondary
8.
J Biol Chem ; 288(40): 28869-80, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23963447

ABSTRACT

Microfibril-associated glycoprotein (MAGP) 1 and 2 are evolutionarily related but structurally divergent proteins that are components of microfibrils of the extracellular matrix. Using mice with a targeted inactivation of Mfap5, the gene for MAGP2 protein, we demonstrate that MAGPs have shared as well as unique functions in vivo. Mfap5(-/-) mice appear grossly normal, are fertile, and have no reduction in life span. Cardiopulmonary development is typical. The animals are normotensive and have vascular compliance comparable with age-matched wild-type mice, which is indicative of normal, functional elastic fibers. Loss of MAGP2 alone does not significantly alter bone mass or architecture, and loss of MAGP2 in tandem with loss of MAGP1 does not exacerbate MAGP1-dependent osteopenia. MAGP2-deficient mice are neutropenic, which contrasts with monocytopenia described in MAGP1-deficient animals. This suggests that MAGP1 and MAGP2 have discrete functions in hematopoiesis. In the cardiovascular system, MAGP1;MAGP2 double knockout mice (Mfap2(-/-);Mfap5(-/-)) show age-dependent aortic dilation. These findings indicate that MAGPs have shared primary functions in maintaining large vessel integrity. In solid phase binding assays, MAGP2 binds active TGFß1, TGFß2, and BMP2. Together, these data demonstrate that loss of MAGP2 expression in vivo has pleiotropic effects potentially related to the ability of MAGP2 to regulate growth factors or participate in cell signaling.


Subject(s)
Contractile Proteins/deficiency , Contractile Proteins/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/metabolism , Genetic Pleiotropy , Alleles , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Bone Density , Bone Morphogenetic Proteins/metabolism , Bone and Bones/pathology , Bone and Bones/physiopathology , Cell Movement , Contractile Proteins/chemistry , Exons/genetics , Extracellular Matrix Proteins/chemistry , Gene Targeting , Leukocyte Count , Male , Mice , Mice, Knockout , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neutropenia/metabolism , Neutropenia/pathology , Neutrophils/metabolism , Neutrophils/pathology , Organ Size , Protein Binding , RNA Splicing Factors , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism
9.
Platelets ; 24(1): 1-5, 2013.
Article in English | MEDLINE | ID: mdl-22372530

ABSTRACT

Recent findings have identified critical roles for the actin filament-crosslinking protein filamin A (FlnA) in platelets and megakaryocytes. This short review focuses on the structure of FlnA and its interaction with the Von Willebrand Factor receptor GPIb-IX-V complex and the fibrinogen receptor, the integrin αIIbß3 in platelets.


Subject(s)
Blood Platelets/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Carrier Proteins , Contractile Proteins/chemistry , Contractile Proteins/genetics , Filamins , Humans , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Mutation , Protein Binding , Thrombocytopenia/genetics , Thrombocytopenia/metabolism
10.
J Biol Chem ; 288(1): 264-73, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23161538

ABSTRACT

Epithelial sodium channel (ENaC) in the kidneys is critical for Na(+) balance, extracellular volume, and blood pressure. Altered ENaC function is associated with respiratory disorders, pseudohypoaldosteronism type 1, and Liddle syndrome. ENaC is known to interact with components of the cytoskeleton, but the functional roles remain largely unclear. Here, we examined the interaction between ENaC and filamins, important actin filament components. We first discovered by yeast two-hybrid screening that the C termini of ENaC α and ß subunits bind filamin A, B, and C, and we then confirmed the binding by in vitro biochemical assays. We demonstrated by co-immunoprecipitation that ENaC, either overexpressed in HEK, HeLa, and melanoma A7 cells or natively expressed in LLC-PK1 and IMCD cells, is in the same complex with native filamin. Furthermore, the biotinylation and co-immunoprecipitation combined assays showed the ENaC-filamin interaction on the cell surface. Using Xenopus oocyte expression and two-electrode voltage clamp electrophysiology, we found that co-expression of an ENaC-binding domain of filamin substantially reduces ENaC channel function. Western blot and immunohistochemistry experiments revealed that the filamin A C terminus (FLNAC) modestly reduces the expression of the ENaC α subunit in oocytes and A7 cells. After normalizing the current by plasma membrane expression, we found that FLNAC results in ~50% reduction in the ENaC channel activity. The inhibitory effect of FLNAC was confirmed by lipid bilayer electrophysiology experiments using purified ENaC and FLNAC proteins, which showed that FLNAC substantially reduces ENaC single channel open probability. Taken together, our study demonstrated that filamin reduces ENaC channel function through direct interaction on the cell surface.


Subject(s)
Contractile Proteins/chemistry , Epithelial Sodium Channels/chemistry , Gene Expression Regulation , Microfilament Proteins/chemistry , Sodium/metabolism , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cytoskeleton/metabolism , Dogs , Filamins , Glutathione Transferase/metabolism , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Kidney/metabolism , Mice , Oocytes/metabolism , Protein Interaction Mapping/methods , Sodium Channels/metabolism , Swine , Two-Hybrid System Techniques , Xenopus
11.
Proc Natl Acad Sci U S A ; 109(48): 19679-84, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23150587

ABSTRACT

Mechanical forces are important signals for cell response and development, but detailed molecular mechanisms of force sensing are largely unexplored. The cytoskeletal protein filamin is a key connecting element between the cytoskeleton and transmembrane complexes such as integrins or the von Willebrand receptor glycoprotein Ib. Here, we show using single-molecule mechanical measurements that the recently reported Ig domain pair 20-21 of human filamin A acts as an autoinhibited force-activatable mechanosensor. We developed a mechanical single-molecule competition assay that allows online observation of binding events of target peptides in solution to the strained domain pair. We find that filamin force sensing is a highly dynamic process occurring in rapid equilibrium that increases the affinity to the target peptides by up to a factor of 17 between 2 and 5 pN. The equilibrium mechanism we find here can offer a general scheme for cellular force sensing.


Subject(s)
Contractile Proteins/chemistry , Microfilament Proteins/chemistry , Contractile Proteins/metabolism , Filamins , Humans , Ligands , Microfilament Proteins/metabolism , Protein Binding
12.
J Mol Biol ; 424(5): 240-7, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23036857

ABSTRACT

The filamin proteins cross-link F-actin and interact with protein partners to integrate both extracellular and intracellular signalling events with the cytoskeleton and to provide mechanoprotection and sensing to cells. The filamins are large, flexible, multi-domain homodimers with the interactions between domains important for protein function. The crystal structure of the N-terminal region of filamin B, containing the actin binding domain (ABD) and the first filamin repeat (FR1) domain, reveals an extended two-domain conformation with no interaction between the ABD and FR1 other than the connecting linker region. The two FLNB347 structures in the crystallographic asymmetric unit exhibit differing relative domain orientations providing the first high-resolution structural characterisation of a filamin inter-domain conformational change. The structure reveals a new hinge in the linker region between ABD and FR1 that is ideally positioned to orient the ABD for actin binding and adds to the previously described hinge regions, hinge 1 (between repeats 15 and 16) and hinge 2 (repeats 23 and 24), providing an additional mechanism by which filamin can exhibit inter-domain flexibility. The extended structure, with the absence of interactions between the domains, implies that any conformational rearrangements required for actin binding by the ABD, as observed for homologous proteins, can freely occur without being influenced by FR1. The ABD retains its previously observed compact conformation. FR1 exhibits a filamin immunoglobulin-like domain fold with a closed C-D ß-strand groove, in contrast to filamin repeats that bind protein partners with this region of the domain surface.


Subject(s)
Contractile Proteins/chemistry , Microfilament Proteins/chemistry , Actins/metabolism , Binding Sites , Contractile Proteins/metabolism , Crystallography, X-Ray , Filamins , Humans , Microfilament Proteins/metabolism , Protein Conformation
13.
J Mol Biol ; 424(5): 248-56, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23041423

ABSTRACT

Filamin A (FLNa) is an actin-binding protein that cross-links F-actin into networks of orthogonally branched filaments. FLNa also directs the networks to integrins while responding to mechanochemical signaling pathways. Flexible, 160-nm-long FLNa molecules are tail-to-tail dimers, each subunit of which contains an N-terminal calponin homology (CH)/actin-binding domain connected by a series of 24 immunoglobulin (Ig) repeats to a dimerization site at their C-terminal end. Whereas the contribution of the CH domains to F-actin affinity is weak (apparent K(a)~10(5)), the binding of the intact protein to F-actin is strong (apparent K(a)~10(8)), suggesting involvement of additional parts of the molecule in this association. Indeed, previous results indicate that Ig repeats along FLNa contribute significantly to the strength of the actin filament interaction. In the current study, we used electron microscopy and three-dimensional reconstruction to elucidate the structural basis of the Ig repeat-F-actin binding. We find that FLNa density is clearly delineated in reconstructions of F-actin complexed either with a four-Ig-repeat segment of FLNa containing Ig repeat 10 or with immunoglobulin-like filamin A repeat (IgFLNa)10 alone. The mass attributable to IgFLNa10 lies peripherally along the actin helix over the N-terminus of actin subdomain 1. The IgFLNa10 interaction appears to be specific, since no other individual Ig repeat or fragment of the FLNa molecule examined, besides ones with IgFLNa10 or CH domains, decorated F-actin filaments or were detected in reconstructions. We conclude that the combined interactions of CH domains and the IgFLNa10 repeat provide the binding strength of the whole FLNa molecule and propose a model for the association of IgFLNa10 on actin filaments.


Subject(s)
Contractile Proteins/chemistry , Contractile Proteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Actins/metabolism , Filamins , Humans , Imaging, Three-Dimensional , Microscopy, Electron , Models, Biological , Models, Molecular , Protein Binding
14.
J Biol Chem ; 287(48): 40371-80, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23007402

ABSTRACT

BACKGROUND: Regulation of integrin activation has important implications for tumor cell invasion and metastasis. RESULTS: EGF activates ERK/p90RSK and Rho/Rho kinase signaling in A431 and DiFi colon cancer cells, leading to phosphorylation of filamin A (FLNa) and inactivation of the α5ß1 integrin receptor. CONCLUSION: EGF promotes α5ß1 inactivation through the p90RSK-dependent phosphorylation of FLNa. SIGNIFICANCE: We have identified a novel EGF-dependent mechanism controlling the α5ß1 integrin activation state. Cell adhesion, motility, and invasion are regulated by the ligand-binding activity of integrin receptors, transmembrane proteins that bind to the extracellular matrix. Integrins whose conformation allows for ligand binding and appropriate functional activity are said to be in an active state. Integrin activation and subsequent ligand binding are dynamically regulated by the association of cytoplasmic proteins with integrin intracellular domains. In this study, we evaluated the role of EGF in the regulation of the activation state of the α5ß1 integrin receptor for fibronectin. The addition of EGF to either A431 squamous carcinoma cells or DiFi colon cancer cells resulted in loss of α5ß1-dependent adhesion to fibronectin but no loss of integrin from the cell surface. EGF activated the EGF receptor/ERK/p90RSK and Rho/Rho kinase signaling pathways. Blocking either pathway inhibited EGF-mediated loss of adhesion, suggesting that they work in parallel to regulate integrin function. EGF treatment also resulted in phosphorylation of filamin A (FLNa), which binds and inactivates ß1 integrins. EGF-mediated FLNa phosphorylation was completely blocked by an inhibitor of p90RSK and partially attenuated by an inhibitor of Rho kinase, suggesting that both pathways converge on FLNa to regulate integrin function. A431 clonal cell lines expressing non-phosphorylated dominant-negative FLNa were resistant to the inhibitory effects of EGF on integrin function, whereas clonal cell lines overexpressing wild-type FLNa were more sensitive to the inhibitory effect of EGF. These data suggest that EGF-dependent inactivation of α5ß1 integrin is regulated through FLNa phosphorylation and cellular contractility.


Subject(s)
Contractile Proteins/metabolism , Epidermal Growth Factor/metabolism , Integrin alpha5beta1/metabolism , Microfilament Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Amino Acid Motifs , Cell Line, Tumor , Contractile Proteins/chemistry , Contractile Proteins/genetics , Filamins , Humans , Integrin alpha5beta1/genetics , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics
15.
Biochim Biophys Acta ; 1824(12): 1334-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22850197

ABSTRACT

The role of filamin in molluscan catch muscles is unknown. In this work three proteins isolated from the posterior adductor muscle of the sea mussel Mytilus galloprovincialis were identified by MALDI-TOF/TOF MS as homologous to mammalian filamin. They were named FLN-270, FLN-230 and FLN-105, according to their apparent molecular weight determined by SDS-PAGE: 270kDa, 230kDa and 105kDa, respectively. Both FLN-270 and FLN-230 contain the C-terminal dimerization domain and the N-terminal actin-binding domain typical of filamins. These findings, together with the data from peptide mass fingerprints, indicate that FLN-270 and FLN-230 are different isoforms of mussel filamin, with FLN-230 being the predominant isoform in the mussel catch muscle. De novo sequencing data revealed structural differences between both filamin isoforms at the rod 2 segment, the one responsible for the interaction of filamin with the most of its binding partners. FLN270 but not FLN230 was phosphorylated in vitro by cAMP-dependent protein kinase. As for the FLN-105, it would be an N-terminal proteolytic fragment generated from the FLN-270 isoform or a C-terminally truncated variant of filamin. On the other hand, a 45-kDa protein that copurifies with mussel catch muscle filamins was identified as the mussel calponin-like protein. The fact that this protein coelutes with the FLN-270 isoform from a gel filtration chromatography suggests a specific interaction between both proteins.


Subject(s)
Contractile Proteins/analysis , Microfilament Proteins/analysis , Muscle, Smooth/chemistry , Mytilus/chemistry , Actins/metabolism , Amino Acid Sequence , Animals , Contractile Proteins/chemistry , Filamins , Microfilament Proteins/chemistry , Molecular Sequence Data , Molecular Weight , Protein Isoforms , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
Cytoskeleton (Hoboken) ; 69(10): 764-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22888038

ABSTRACT

Cell division is a critical and irreversible step in the cell cycle. The strategies that cells follow to regulate the position of the division plane must take into account the global geometry of the cell as well as position of the genetic material to ensure its accurate segregation into daughter cells of a given cell shape and size. Along the years, research on Schizosaccharomyces pombe, a well-recognized model organism for cell division studies has allowed a detailed molecular understanding of the spatial mechanisms regulating cytokinesis. Division plane position in this unicellular rod-shaped organism, which divides by the assembly and constriction of a medially placed actomyosin ring, largely depends on the anillin-like protein Mid1. Therefore, the major pathways controlling the position of the division plane converge on Mid1. In this review, we make an overview of the studies that have deciphered how Mid1 localization and scaffolding activities are controlled over the cell cycle to ensure the symmetrical division of fission yeast cells. These studies have revealed new mechanisms generating spatial information based on nuclear shuttling of the division plane factor Mid1 and on the establishment of cortical inhibitory gradients of the cell polarity kinase Pom1.


Subject(s)
Contractile Proteins/metabolism , Cytokinesis , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Cell Nucleus/metabolism , Contractile Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Signal Transduction
17.
Mol Biol Cell ; 23(20): 3993-4007, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918954

ABSTRACT

Fission yeast cells depend on the anillin-related protein Mid1p for reliable cytokinesis. Insolubility limits the purification of full-length Mid1p for biophysical analysis, and lack of knowledge about the structural domains of Mid1p limits functional analysis. We addressed these limitations by identifying in a bacterial expression screen of random Mid1p fragments five soluble segments that can be purified and one insoluble segment. Using complementation experiments in Δmid1 cells, we tested the biological functions of these six putative domains that account for full-length Mid1p. The N-terminal domain (residues 1-149) is essential for correct positioning and orientation of septa. The third domain (residues 309-452) allows the construct composed of the first three domains (residues 1-452) to form hydrodynamically well-behaved octamers. Constructs consisting of residues 1-452 or 1-578 carry out most functions of full-length Mid1p, including concentration at the equatorial cortex in nodes that accumulate myosin-II and other contractile ring proteins during mitosis. However, cells depending on these constructs without the insoluble domain (residues 579-797) form equatorially located rings slowly from strands rather than by direct condensation of nodes. We conclude that residues 1-578 assemble node components myosin-II, Rng2p, and Cdc15p, and the insoluble domain facilitates the normal, efficient condensation of nodes into rings.


Subject(s)
Contractile Proteins/chemistry , Contractile Proteins/metabolism , Cytokinesis , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Cell Division , Cell Polarity , Genetic Complementation Test , Hydrodynamics , Models, Biological , Protein Structure, Tertiary , Protein Transport , Sequence Deletion , Solubility , Structure-Activity Relationship , Ultracentrifugation
18.
PLoS One ; 7(7): e40448, 2012.
Article in English | MEDLINE | ID: mdl-22802962

ABSTRACT

Filamins are important actin cross-linking proteins implicated in scaffolding, membrane stabilization and signal transduction, through interaction with ion channels, receptors and signaling proteins. Here we report the physical and functional interaction between filamins and polycystin-2, a TRP-type cation channel mutated in 10-15% patients with autosomal dominant polycystic kidney disease. Yeast two-hybrid and GST pull-down experiments demonstrated that the C-termini of filamin isoforms A, B and C directly bind to both the intracellular N- and C-termini of polycystin-2. Reciprocal co-immunoprecipitation experiments showed that endogenous polycystin-2 and filamins are in the same complexes in renal epithelial cells and human melanoma A7 cells. We then examined the effect of filamin on polycystin-2 channel function by electrophysiology studies with a lipid bilayer reconstitution system and found that filamin-A substantially inhibits polycystin-2 channel activity. Our study indicates that filamins are important regulators of polycystin-2 channel function, and further links actin cytoskeletal dynamics to the regulation of this channel protein.


Subject(s)
Contractile Proteins/chemistry , Microfilament Proteins/chemistry , TRPP Cation Channels/physiology , Animals , Calcium Channels/drug effects , Calcium Channels/physiology , Cell Line , Cell Line, Tumor , Contractile Proteins/physiology , Filamins , Humans , Immunoprecipitation , Lipid Bilayers , Microfilament Proteins/physiology , Protein Isoforms/metabolism , Two-Hybrid System Techniques
19.
Bioorg Khim ; 38(2): 242-50, 2012.
Article in Russian | MEDLINE | ID: mdl-22792729

ABSTRACT

Reactions ofnucleophilic substitution and enzymatic processes with participation of metal-porphyrins (MP) are considered from the point of view of Zn-tetraphenylporphin (Zn-TPhP) coordination with corresponding ligand/nucleophyl/substrate/base. Linear correlations perform between kinetic parameters of process of coordination of Zn-TPhP in chloroform (constants of stability) and reactions of nucleophilic substitution both in aqueous and organic solvents with participation ofpyridines, N-oxides ofpyridines, anilines, primary amines and oxidation of anilines by horseradish peroxidase in aqueous solutions (rate constants). Thermodynamic parameters of complexation and nucleophilic substitution mutually correlate linearly in the case of pyridines, anilines and primary amines.


Subject(s)
Horseradish Peroxidase/chemistry , Metalloporphyrins/chemistry , Zinc/chemistry , Contractile Proteins/chemistry , Kinetics , Oxidation-Reduction
20.
J Cell Sci ; 125(Pt 13): 3051-60, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22797913

ABSTRACT

Many essential cellular functions in health and disease are closely linked to the ability of cells to respond to mechanical forces. In the context of cell adhesion to the extracellular matrix, the forces that are generated within the actin cytoskeleton and transmitted through integrin-based focal adhesions are essential for the cellular response to environmental clues, such as the spatial distribution of adhesive ligands or matrix stiffness. Whereas substantial progress has been made in identifying mechanosensitive molecules that can transduce mechanical force into biochemical signals, much less is known about the nature of cytoskeletal force generation and transmission that regulates the magnitude, duration and spatial distribution of forces imposed on these mechanosensitive complexes. By focusing on cell-matrix adhesion to flat elastic substrates, on which traction forces can be measured with high temporal and spatial resolution, we discuss our current understanding of the physical mechanisms that integrate a large range of molecular mechanotransduction events on cellular scales. Physical limits of stability emerge as one important element of the cellular response that complements the structural changes affected by regulatory systems in response to mechanical processes.


Subject(s)
Actin Cytoskeleton/chemistry , Cell-Matrix Junctions/chemistry , Mechanotransduction, Cellular , Actin Cytoskeleton/physiology , Animals , Biomechanical Phenomena , Cell-Matrix Junctions/physiology , Contractile Proteins/chemistry , Contractile Proteins/physiology , Elastic Modulus , Materials Testing/methods , Microscopy, Atomic Force/methods , Protein Stability , Pseudopodia/chemistry , Pseudopodia/physiology , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...