Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
Add more filters

Publication year range
1.
Cell ; 154(5): 1047-1059, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23954413

ABSTRACT

Key cellular decisions, such as proliferation or growth arrest, typically occur at spatially defined locations within tissues. Loss of this spatial control is a hallmark of many diseases, including cancer. Yet, how these patterns are established is incompletely understood. Here, we report that physical and architectural features of a multicellular sheet inform cells about their proliferative capacity through mechanical regulation of YAP and TAZ, known mediators of Hippo signaling and organ growth. YAP/TAZ activity is confined to cells exposed to mechanical stresses, such as stretching, location at edges/curvatures contouring an epithelial sheet, or stiffness of the surrounding extracellular matrix. We identify the F-actin-capping/severing proteins Cofilin, CapZ, and Gelsolin as essential gatekeepers that limit YAP/TAZ activity in cells experiencing low mechanical stresses, including contact inhibition of proliferation. We propose that mechanical forces are overarching regulators of YAP/TAZ in multicellular contexts, setting responsiveness to Hippo, WNT, and GPCR signaling.


Subject(s)
Actin Capping Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Phosphoproteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Actins/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Breast Neoplasms/pathology , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Mechanical Phenomena , Phosphoproteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , YAP-Signaling Proteins
2.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36897576

ABSTRACT

Actin dynamics play an important role in tissue morphogenesis, yet the control of actin filament growth takes place at the molecular level. A challenge in the field is to link the molecular function of actin regulators with their physiological function. Here, we report an in vivo role of the actin-capping protein CAP-1 in the Caenorhabditis elegans germline. We show that CAP-1 is associated with actomyosin structures in the cortex and rachis, and its depletion or overexpression led to severe structural defects in the syncytial germline and oocytes. A 60% reduction in the level of CAP-1 caused a twofold increase in F-actin and non-muscle myosin II activity, and laser incision experiments revealed an increase in rachis contractility. Cytosim simulations pointed to increased myosin as the main driver of increased contractility following loss of actin-capping protein. Double depletion of CAP-1 and myosin or Rho kinase demonstrated that the rachis architecture defects associated with CAP-1 depletion require contractility of the rachis actomyosin corset. Thus, we uncovered a physiological role for actin-capping protein in regulating actomyosin contractility to maintain reproductive tissue architecture.


Subject(s)
Actomyosin , Caenorhabditis elegans , Animals , Actomyosin/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Actins/metabolism , Actin Capping Proteins/metabolism , Actin Cytoskeleton/metabolism , Myosins/metabolism , Germ Cells/metabolism
3.
Nat Rev Mol Cell Biol ; 15(10): 677-89, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25207437

ABSTRACT

Capping protein (CP) binds the fast growing barbed end of the actin filament and regulates actin assembly by blocking the addition and loss of actin subunits. Recent studies provide new insights into how CP and barbed-end capping are regulated. Filament elongation factors, such as formins and ENA/VASP (enabled/vasodilator-stimulated phosphoprotein), indirectly regulate CP by competing with CP for binding to the barbed end, whereas other molecules, including V-1 and phospholipids, directly bind to CP and sterically block its interaction with the filament. In addition, a diverse and unrelated group of proteins interact with CP through a conserved 'capping protein interaction' (CPI) motif. These proteins, including CARMIL (capping protein, ARP2/3 and myosin I linker), CD2AP (CD2-associated protein) and the WASH (WASP and SCAR homologue) complex subunit FAM21, recruit CP to specific subcellular locations and modulate its actin-capping activity via allosteric effects.


Subject(s)
Actin Capping Proteins/metabolism , Actin Cytoskeleton/physiology , DNA-Binding Proteins/metabolism , Actin Capping Proteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/physiology , DNA-Binding Proteins/physiology , Humans , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Models, Molecular , Phosphatidylinositol Phosphates/chemistry , Protein Binding , Protein Conformation
4.
Proteins ; 92(1): 37-43, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37497763

ABSTRACT

Capping protein (CP) binds to the barbed end of an actin-filament and inhibits its elongation. CARMIL binds CP and dissociates it from the barbed end of the actin-filament. The binding of CARMIL peptide alters the flexibility of CP, which is considered to facilitate the dissociation. Twinfilin also binds to CP through its C-terminal tail. The complex structures of the CP/twinfilin-tail (TW-tail) peptide indicate that the binding sites of CARMIL and TW-tail overlap. However, TW-tail binding does not facilitate the dissociation of CP from the barbed end. We extensively investigated the flexibilities of CP in the CP/TW-tail or CP/CARMIL complexes using an elastic network model and concluded that TW-tail binding does not alter the flexibility of CP. Our extensive analysis also highlighted that the strong contacts of peptides with the two domains of CP, that is, the CP-L and CP-S domains, are key to changing the flexibilities of CP. CARMIL peptides can interact strongly with both of the domains, while TW-tail peptides exclusively interact with the CP-S domain because the binding site of TW-tail on CP relatively shifts to the CP-S domain compared with that of CP/CARMIL. This result supports our hypothesis that the dissociation of CP from the barbed end is regulated by the flexibility of CP.


Subject(s)
Actin Capping Proteins , Microfilament Proteins , Microfilament Proteins/metabolism , Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Protein Binding , Actins/metabolism , Actin Cytoskeleton/metabolism , Peptides/chemistry
5.
Funct Integr Genomics ; 24(4): 120, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38960936

ABSTRACT

The Drosophila egg chamber (EC) starts as a spherical tissue at the beginning. With maturation, the outer follicle cells of EC collectively migrate in a direction perpendicular to the anterior-posterior axis, to shape EC from spherical to ellipsoidal. Filamentous actin (F-actin) plays a significant role in shaping individual migratory cells to the overall EC shape, like in every cell migration. The primary focus of this article is to unveil the function of different Actin Binding Proteins (ABPs) in regulating mature Drosophila egg shape. We have screened 66 ABPs, and the genetic screening data revealed that individual knockdown of Arp2/3 complex genes and the "capping protein ß" (cpb) gene have severely altered the egg phenotype. Arpc1 and cpb RNAi mediated knockdown resulted in the formation of spherical eggs which are devoid of dorsal appendages. Studies also showed the role of Arpc1 and cpb on the number of laid eggs and follicle cell morphology. Furthermore, the depletion of Arpc1 and cpb resulted in a change in F-actin quantity. Together, the data indicate that Arpc1 and cpb regulate Drosophila egg shape, F-actin management, egg-laying characteristics and dorsal appendages formation.


Subject(s)
Actins , Drosophila Proteins , Morphogenesis , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Actins/metabolism , Actins/genetics , Female , Morphogenesis/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin Capping Proteins/metabolism , Actin Capping Proteins/genetics , Ovum/metabolism , Ovum/growth & development
6.
J Cell Sci ; 135(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35583107

ABSTRACT

Capping protein Arp2/3 myosin I linker (CARMIL) proteins are multi-domain scaffold proteins that regulate actin dynamics by regulating the activity of capping protein (CP). Here, we characterize CARMIL-GAP (GAP for GTPase-activating protein), a Dictyostelium CARMIL isoform that contains a ∼130 residue insert that, by homology, confers GTPase-activating properties for Rho-related GTPases. Consistent with this idea, this GAP domain binds Dictyostelium Rac1a and accelerates its rate of GTP hydrolysis. CARMIL-GAP concentrates with F-actin in phagocytic cups and at the leading edge of chemotaxing cells, and CARMIL-GAP-null cells exhibit pronounced defects in phagocytosis and chemotactic streaming. Importantly, these defects are fully rescued by expressing GFP-tagged CARMIL-GAP in CARMIL-GAP-null cells. Finally, rescue with versions of CARMIL-GAP that lack either GAP activity or the ability to regulate CP show that, although both activities contribute significantly to CARMIL-GAP function, the GAP activity plays the bigger role. Together, our results add to the growing evidence that CARMIL proteins influence actin dynamics by regulating signaling molecules as well as CP, and that the continuous cycling of the nucleotide state of Rho GTPases is often required to drive Rho-dependent biological processes.


Subject(s)
Actin Capping Proteins , Dictyostelium , Actin Capping Proteins/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Dictyostelium/genetics , Dictyostelium/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism
7.
PLoS Biol ; 18(9): e3000848, 2020 09.
Article in English | MEDLINE | ID: mdl-32898131

ABSTRACT

Improper lengths of actin-thin filaments are associated with altered contractile activity and lethal myopathies. Leiomodin, a member of the tropomodulin family of proteins, is critical in thin filament assembly and maintenance; however, its role is under dispute. Using nuclear magnetic resonance data and molecular dynamics simulations, we generated the first atomic structural model of the binding interface between the tropomyosin-binding site of cardiac leiomodin and the N-terminus of striated muscle tropomyosin. Our structural data indicate that the leiomodin/tropomyosin complex only forms at the pointed end of thin filaments, where the tropomyosin N-terminus is not blocked by an adjacent tropomyosin protomer. This discovery provides evidence supporting the debated mechanism where leiomodin and tropomodulin regulate thin filament lengths by competing for thin filament binding. Data from experiments performed in cardiomyocytes provide additional support for the competition model; specifically, expression of a leiomodin mutant that is unable to interact with tropomyosin fails to displace tropomodulin at thin filament pointed ends and fails to elongate thin filaments. Together with previous structural and biochemical data, we now propose a molecular mechanism of actin polymerization at the pointed end in the presence of bound leiomodin. In the proposed model, the N-terminal actin-binding site of leiomodin can act as a "swinging gate" allowing limited actin polymerization, thus making leiomodin a leaky pointed-end cap. Results presented in this work answer long-standing questions about the role of leiomodin in thin filament length regulation and maintenance.


Subject(s)
Actin Cytoskeleton/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Actin Cytoskeleton/chemistry , Actins/chemistry , Actins/metabolism , Animals , Animals, Newborn , Binding Sites , Cells, Cultured , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Myocardium/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Interaction Domains and Motifs , Rats , Sarcomeres/metabolism
8.
Cell ; 133(5): 765-7, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18510919

ABSTRACT

Capping proteins limit actin filament growth, but paradoxically increase actin-based cell motility. This has been attributed to funneling of actin monomers to the filament ends that remain uncapped. Using a reconstituted motility system, Akin and Mullins (2008) now demonstrate that filament capping increases Arp2/3-based nucleation and branching, rather than elevating the rate of filament elongation.


Subject(s)
Actins/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Actin Capping Proteins/metabolism , Actin Cytoskeleton/metabolism , Models, Biological
9.
Cell ; 133(5): 841-51, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18510928

ABSTRACT

Capping protein (CP) is an integral component of Arp2/3-nucleated actin networks that drive amoeboid motility. Increasing the concentration of capping protein, which caps barbed ends of actin filaments and prevents elongation, increases the rate of actin-based motility in vivo and in vitro. We studied the synergy between CP and Arp2/3 using an in vitro actin-based motility system reconstituted from purified proteins. We find that capping protein increases the rate of motility by promoting more frequent filament nucleation by the Arp2/3 complex and not by increasing the rate of filament elongation as previously suggested. One consequence of this coupling between capping and nucleation is that, while the rate of motility depends strongly on the concentration of CP and Arp2/3, the net rate of actin assembly is insensitive to changes in either factor. By reorganizing their architecture, dendritic actin networks harness the same assembly kinetics to drive different rates of motility.


Subject(s)
Actin Capping Proteins/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Cell Movement , Actin Capping Proteins/isolation & purification , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/isolation & purification , Actin Depolymerizing Factors/metabolism , Actin-Related Protein 2-3 Complex/isolation & purification , Actins/isolation & purification , Animals , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Brain Chemistry , Cattle , Cell-Free System , Kinetics , Listeria monocytogenes , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Microspheres , Polystyrenes/metabolism , Profilins/isolation & purification , Profilins/metabolism
10.
Cell Mol Life Sci ; 79(2): 125, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35132495

ABSTRACT

Apicomplexan parasites, such as Plasmodium spp., rely on an unusual actomyosin motor, termed glideosome, for motility and host cell invasion. The actin filaments are maintained by a small set of essential regulators, which provide control over actin dynamics in the different stages of the parasite life cycle. Actin filament capping proteins (CPs) are indispensable heterodimeric regulators of actin dynamics. CPs have been extensively characterized in higher eukaryotes, but their role and functional mechanism in Apicomplexa remain enigmatic. Here, we present the first crystal structure of a homodimeric CP from the malaria parasite and compare the homo- and heterodimeric CP structures in detail. Despite retaining several characteristics of a canonical CP, the homodimeric Plasmodium berghei (Pb)CP exhibits crucial differences to the canonical heterodimers. Both homo- and heterodimeric PbCPs regulate actin dynamics in an atypical manner, facilitating rapid turnover of parasite actin, without affecting its critical concentration. Homo- and heterodimeric PbCPs show partially redundant activities, possibly to rescue actin filament capping in life cycle stages where the ß-subunit is downregulated. Our data suggest that the homodimeric PbCP also influences actin kinetics by recruiting lateral actin dimers. This unusual function could arise from the absence of a ß-subunit, as the asymmetric PbCP homodimer lacks structural elements essential for canonical barbed end interactions suggesting a novel CP binding mode. These findings will facilitate further studies aimed at elucidating the precise actin filament capping mechanism in Plasmodium.


Subject(s)
Actin Capping Proteins , Antigens, Protozoan , Malaria/parasitology , Plasmodium/metabolism , Protozoan Proteins , Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Kinetics , Models, Molecular , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
11.
Int J Mol Sci ; 23(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35886903

ABSTRACT

The infection of a mammalian host by the pathogenic fungus Candida albicans involves fungal resistance to reactive oxygen species (ROS)-induced DNA damage stress generated by the defending macrophages or neutrophils. Thus, the DNA damage response in C. albicans may contribute to its pathogenicity. Uncovering the transcriptional changes triggered by the DNA damage-inducing agent MMS in many model organisms has enhanced the understanding of their DNA damage response processes. However, the transcriptional regulation triggered by MMS remains unclear in C. albicans. Here, we explored the global transcription profile in response to MMS in C. albicans and identified 306 defined genes whose transcription was significantly affected by MMS. Only a few MMS-responsive genes, such as MGT1, DDR48, MAG1, and RAD7, showed potential roles in DNA repair. GO term analysis revealed that a large number of induced genes were involved in antioxidation responses, and some downregulated genes were involved in nucleosome packing and IMP biosynthesis. Nevertheless, phenotypic assays revealed that MMS-induced antioxidation gene CAP1 and glutathione metabolism genes GST2 and GST3 showed no direct roles in MMS resistance. Furthermore, the altered transcription of several MMS-responsive genes exhibited RAD53-related regulation. Intriguingly, the transcription profile in response to MMS in C. albicans shared a limited similarity with the pattern in S. cerevisiae, including COX17, PRI2, and MGT1. Overall, C. albicans cells exhibit global transcriptional changes to the DNA damage agent MMS; these findings improve our understanding of this pathogen's DNA damage response pathways.


Subject(s)
Candida albicans , Methyl Methanesulfonate , Actin Capping Proteins/genetics , Actin Capping Proteins/metabolism , Animals , Candida albicans/drug effects , Candida albicans/genetics , Candida albicans/metabolism , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Mammals/metabolism , Methyl Methanesulfonate/pharmacology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
12.
J Biol Chem ; 295(45): 15366-15375, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32868296

ABSTRACT

Heterodimeric capping protein (CP) binds the rapidly growing barbed ends of actin filaments and prevents the addition (or loss) of subunits. Capping activity is generally considered to be essential for actin-based motility induced by Arp2/3 complex nucleation. By stopping barbed end growth, CP favors nucleation of daughter filaments at the functionalized surface where the Arp2/3 complex is activated, thus creating polarized network growth, which is necessary for movement. However, here using an in vitro assay where Arp2/3 complex-based actin polymerization is induced on bead surfaces in the absence of CP, we produce robust polarized actin growth and motility. This is achieved either by adding the actin polymerase Ena/VASP or by boosting Arp2/3 complex activity at the surface. Another actin polymerase, the formin FMNL2, cannot substitute for CP, showing that polymerase activity alone is not enough to override the need for CP. Interfering with the polymerase activity of Ena/VASP, its surface recruitment or its bundling activity all reduce Ena/VASP's ability to maintain polarized network growth in the absence of CP. Taken together, our findings show that CP is dispensable for polarized actin growth and motility in situations where surface-directed polymerization is favored by whatever means over the growth of barbed ends in the network.


Subject(s)
Actin Capping Proteins/metabolism , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , DNA-Binding Proteins/metabolism , Formins/metabolism , Animals , Mice , Polymerization , Rabbits , Swine
13.
Proc Natl Acad Sci U S A ; 115(9): 2138-2143, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29439200

ABSTRACT

During cytokinesis, a cleavage furrow generated by actomyosin ring contraction is restructured into the midbody, a platform for the assembly of the abscission machinery that controls the final separation of daughter cells. The polymerization state of F-actin is important during assembly, ingression, disassembly, and closure of the contractile ring and for the cytoskeletal remodeling that accompanies midbody formation and progression to abscission. Actin filaments must be cleared from the abscission sites before the final cut can take place. Although many conserved proteins interact with and influence the polymerization state of actin filaments, it is poorly understood how they regulate cytokinesis in higher eukaryotes. We report here that the actin capping protein (CP), a barbed end actin binding protein, participates in the control of actin polymerization during later stages of cytokinesis in human cells. Cells depleted of CP furrow and form early midbodies, but they fail cytokinesis. Appropriate recruitment of the ESCRT-III abscission machinery to the midbody is impaired, preventing the cell from progressing to the abscission stage. To generate actin filaments of optimal length, different actin nucleators, such as formins, balance CP's activity. Loss of actin capping activity leads to excessive accumulation of formin-based linear actin filaments. Depletion of the formin FHOD1 results in partial rescue of CP-induced cytokinesis failure, suggesting that it can antagonize CP activity during midbody maturation. Our work suggests that the actin cytoskeleton is remodeled in a stepwise manner during cytokinesis, with different regulators at different stages required for successful progression to abscission.


Subject(s)
Actin Capping Proteins/physiology , Actin Cytoskeleton/physiology , Cytokinesis/physiology , Actins , Cell Membrane , Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Cells/physiology , Epithelium, Corneal/cytology , Fetal Proteins/genetics , Fetal Proteins/metabolism , Formins , Gene Expression Regulation/physiology , HeLa Cells , Humans , Microfilament Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
14.
Biochem Biophys Res Commun ; 525(3): 681-686, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32139121

ABSTRACT

Actin capping proteins belong to the core set of proteins minimally required for actin-based motility and are present in virtually all eukaryotic cells. They bind to the fast-growing barbed end of an actin filament, preventing addition and loss of monomers, thus restricting growth to the slow-growing pointed end. Actin capping proteins are usually heterodimers of two subunits. The Plasmodium orthologs are an exception, as their α subunits are able to form homodimers. We show here that, while the ß subunit alone is unstable, the α subunit of the Plasmodium actin capping protein forms functional homo- and heterodimers. This implies independent functions for the αα homo- and αß heterodimers in certain stages of the parasite life cycle. Structurally, the homodimers resemble canonical αß heterodimers, although certain rearrangements at the interface must be required. Both homo- and heterodimers bind to actin filaments in a roughly equimolar ratio, indicating they may also bind other sites than barbed ends.


Subject(s)
Actin Capping Proteins/metabolism , Malaria/parasitology , Parasites/metabolism , Protein Multimerization , Protozoan Proteins/metabolism , Actin Capping Proteins/chemistry , Actin Cytoskeleton/metabolism , Animals , Plasmodium/metabolism , Protein Binding , Protein Folding , Solutions , Temperature
15.
Cell Commun Signal ; 18(1): 121, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32771000

ABSTRACT

BACKGROUND: The PIM family kinases promote cancer cell survival and motility as well as metastatic growth in various types of cancer. We have previously identified several PIM substrates, which support cancer cell migration and invasiveness. However, none of them are known to regulate cellular movements by directly interacting with the actin cytoskeleton. Here we have studied the phosphorylation-dependent effects of PIM1 on actin capping proteins, which bind as heterodimers to the fast-growing actin filament ends and stabilize them. METHODS: Based on a phosphoproteomics screen for novel PIM substrates, we have used kinase assays and fluorescence-based imaging techniques to validate actin capping proteins as PIM1 substrates and interaction partners. We have analysed the functional consequences of capping protein phosphorylation on cell migration and adhesion by using wound healing and real-time impedance-based assays. We have also investigated phosphorylation-dependent effects on actin polymerization by analysing the protective role of capping protein phosphomutants in actin disassembly assays. RESULTS: We have identified capping proteins CAPZA1 and CAPZB2 as PIM1 substrates, and shown that phosphorylation of either of them leads to increased adhesion and migration of human prostate cancer cells. Phosphorylation also reduces the ability of the capping proteins to protect polymerized actin from disassembly. CONCLUSIONS: Our data suggest that PIM kinases are able to induce changes in actin dynamics to support cell adhesion and movement. Thus, we have identified a novel mechanism through which PIM kinases enhance motility and metastatic behaviour of cancer cells. Video abstract.


Subject(s)
Actin Capping Proteins/metabolism , Cell Movement , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-pim-1/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Surface Extensions/metabolism , Cytoplasm/metabolism , Humans , Male , Mice , Phosphorylation , Protein Multimerization , Protein Subunits/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors
16.
Immunity ; 35(3): 388-99, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21835647

ABSTRACT

Dendritic cells (DCs) flexibly adapt to different microenvironments by using diverse migration strategies that are ultimately dependent on the dynamics and structural organization of the actin cytoskeleton. Here, we have shown that DCs require the actin capping activity of the signaling adaptor Eps8 to polarize and to form elongated migratory protrusions. DCs from Eps8-deficient mice are impaired in directional and chemotactic migration in 3D in vitro and are delayed in reaching the draining lymph node (DLN) in vivo after inflammatory challenge. Hence, Eps8-deficient mice are unable to mount a contact hypersensitivity response. We have also shown that the DC migratory defect is cell autonomous and that Eps8 is required for the proper architectural organization of the actin meshwork and dynamics of cell protrusions. Yet, Eps8 is not necessary for antigen uptake, processing, and presentation. Thus, we have identified Eps8 as a unique actin capping protein specifically required for DC migration.


Subject(s)
Actin Capping Proteins/immunology , Adaptor Proteins, Signal Transducing/immunology , Cytoskeletal Proteins/immunology , Dendritic Cells/immunology , Signal Transduction , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigen Presentation , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Dermatitis, Contact/immunology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
17.
PLoS Genet ; 13(5): e1006814, 2017 May.
Article in English | MEDLINE | ID: mdl-28542408

ABSTRACT

Actin organization is a conserved cellular process that regulates the growth and development of eukaryotic cells. It also governs the virulence process of pathogenic fungi, such as the rice blast fungus Magnaporthe oryzae, with mechanisms not yet fully understood. In a previous study, we found that actin-regulating kinase MoArk1 displays conserved functions important in endocytosis and actin organization, and MoArk1 is required for maintaining the growth and full virulence of M. oryzae. To understand how MoArk1 might function, we identified capping protein homologs from M. oryzae (MoCAP) that interact with MoArk1 in vivo. MoCAP is heterodimer consisting of α and ß subunits MoCapA and MoCapB. Single and double deletions of MoCAP subunits resulted in abnormal mycelial growth and conidia formation. The ΔMocap mutants also exhibited reduced appressorium penetration and invasive hyphal growth within host cells. Furthermore, the ΔMocap mutants exhibited delayed endocytosis and abnormal cytoskeleton assembly. Consistent with above findings, MoCAP proteins interacted with MoAct1, co-localized with actin during mycelial development, and participated in appressorial actin ring formation. Further analysis revealed that the S85 residue of MoCapA and the S285 residue of MoCapB were subject to phosphorylation by MoArk1 that negatively regulates MoCAP functions. Finally, the addition of exogenous phosphatidylinositol 4,5-bisphosphate (PIP2) failed to modulate actin ring formation in ΔMocap mutants, in contrast to the wild-type strain, suggesting that MoCAP may also mediate phospholipid signaling in the regulation of the actin organization. These results together demonstrate that MoCAP proteins whose functions are regulated by MoArk1 and PIP2 are important for endocytosis and actin dynamics that are directly linked to growth, conidiation and pathogenicity of M. oryzae.


Subject(s)
Actin Capping Proteins/metabolism , Actins/metabolism , Endocytosis , Fungal Proteins/metabolism , Magnaporthe/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Actin Capping Proteins/genetics , Magnaporthe/growth & development , Magnaporthe/pathogenicity , Mycelium/growth & development , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Virulence
18.
FASEB J ; 32(6): 3119-3132, 2018 06.
Article in English | MEDLINE | ID: mdl-29401618

ABSTRACT

IL-1 signaling is adhesion-restricted in many cell types, but the mechanism that drives it is not defined. We screened for proteins recruited to nascent adhesions in IL-1-treated human fibroblasts with tandem mass tag-mass spectrometry. We used fibronectin bead preparations to enrich 10 actin-associated proteins. There was a 1.2 times log 2-fold enrichment of actin capping protein (ACP) at 30 min after IL-1 stimulation. Knockdown (KD) of ACP by siRNA reduced IL-1-induced ERK activation(by 56%, matrix metalloproteinase-3 (MMP-3) expression by 48%, and MMP-9 expression by 62% (in all reductions, P < 0.01). Confocal or structured illumination microscopy showed that ACP was diffused throughout the cytosol but strongly accumulated at the ruffled border of spreading cells. ACP colocalized with nascent paxillin- and vinculin-containing adhesions at the ruffled border, but not with mature adhesions in the center. ACP KD promoted the formation of large, stable adhesions. Immunoprecipitation and proximity ligation analysis showed that ACP was associated with the IL-1 signal transduction proteins myeloid differentiation factor 88 (MyD88) and IL-1 receptor-associated kinase (IRAK) at the ruffled border of the leading edge. IL-1-induced phospho-ERK and MyD88 or IRAK colocalized at the leading edge. We concluded that ACP is required for recruitment and function of IL-1 signaling complexes in nascent adhesions at the leading edge of the cell.-Wang, Q., Delcorde, J., Tang, T., Downey, G. P., McCulloch, C. A. Regulation of IL-1 signaling through control of focal adhesion assembly.


Subject(s)
Actin Capping Proteins/metabolism , Fibroblasts/metabolism , Focal Adhesions/metabolism , Interleukin-1/metabolism , MAP Kinase Signaling System/physiology , Actin Capping Proteins/genetics , Fibroblasts/cytology , Focal Adhesions/genetics , Gene Knockdown Techniques , Humans , Interleukin-1/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism
19.
Proc Natl Acad Sci U S A ; 113(43): E6610-E6619, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27791032

ABSTRACT

Capping Protein (CP) plays a central role in the creation of the Arp2/3-generated branched actin networks comprising lamellipodia and pseudopodia by virtue of its ability to cap the actin filament barbed end, which promotes Arp2/3-dependent filament nucleation and optimal branching. The highly conserved protein V-1/Myotrophin binds CP tightly in vitro to render it incapable of binding the barbed end. Here we addressed the physiological significance of this CP antagonist in Dictyostelium, which expresses a V-1 homolog that we show is very similar biochemically to mouse V-1. Consistent with previous studies of CP knockdown, overexpression of V-1 in Dictyostelium reduced the size of pseudopodia and the cortical content of Arp2/3 and induced the formation of filopodia. Importantly, these effects scaled positively with the degree of V-1 overexpression and were not seen with a V-1 mutant that cannot bind CP. V-1 is present in molar excess over CP, suggesting that it suppresses CP activity in the cytoplasm at steady state. Consistently, cells devoid of V-1, like cells overexpressing CP described previously, exhibited a significant decrease in cellular F-actin content. Moreover, V-1-null cells exhibited pronounced defects in macropinocytosis and chemotactic aggregation that were rescued by V-1, but not by the V-1 mutant. Together, these observations demonstrate that V-1 exerts significant influence in vivo on major actin-based processes via its ability to sequester CP. Finally, we present evidence that V-1's ability to sequester CP is regulated by phosphorylation, suggesting that cells may manipulate the level of active CP to tune their "actin phenotype."


Subject(s)
Actin Capping Proteins/genetics , Actin-Related Protein 2-3 Complex/genetics , Dictyostelium/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Protozoan Proteins/genetics , Pseudopodia/metabolism , Actin Capping Proteins/metabolism , Actin-Related Protein 2/genetics , Actin-Related Protein 2/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 3/genetics , Actin-Related Protein 3/metabolism , Actins/genetics , Actins/metabolism , Amino Acid Sequence , Animals , Chemotaxis/genetics , Conserved Sequence , Dictyostelium/genetics , Dictyostelium/ultrastructure , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Kinetics , Mice , Mutation , Phosphorylation , Pinocytosis/genetics , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Protozoan Proteins/metabolism , Pseudopodia/genetics , Pseudopodia/ultrastructure , Sequence Alignment , Signal Transduction
20.
J Biol Chem ; 292(33): 13566-13583, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28642367

ABSTRACT

Disheveled-associated activator of morphogenesis (DAAM) is a diaphanous-related formin protein essential for the regulation of actin cytoskeleton dynamics in diverse biological processes. The conserved formin homology 1 and 2 (FH1-FH2) domains of DAAM catalyze actin nucleation and processively mediate filament elongation. These activities are indirectly regulated by the N- and C-terminal regions flanking the FH1-FH2 domains. Recently, the C-terminal diaphanous-autoregulatory domain (DAD) and the C terminus (CT) of formins have also been shown to regulate actin assembly by directly interacting with actin. Here, to better understand the biological activities of DAAM, we studied the role of DAD-CT regions of Drosophila DAAM in its interaction with actin with in vitro biochemical and in vivo genetic approaches. We found that the DAD-CT region binds actin in vitro and that its main actin-binding element is the CT region, which does not influence actin dynamics on its own. However, we also found that it can tune the nucleating activity and the filament end-interaction properties of DAAM in an FH2 domain-dependent manner. We also demonstrate that DAD-CT makes the FH2 domain more efficient in antagonizing with capping protein. Consistently, in vivo data suggested that the CT region contributes to DAAM-mediated filopodia formation and dynamics in primary neurons. In conclusion, our results demonstrate that the CT region of DAAM plays an important role in actin assembly regulation in a biological context.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Models, Molecular , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Pseudopodia/metabolism , Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Actin Cytoskeleton/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Animals , Cells, Cultured , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Embryo, Nonmammalian/cytology , Gene Deletion , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL