Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
1.
Plant Cell ; 36(4): 1159-1181, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38134410

ABSTRACT

Plants have unique responses to fluctuating light conditions. One such response involves chloroplast photorelocation movement, which optimizes photosynthesis under weak light by the accumulation of chloroplasts along the periclinal side of the cell, which prevents photodamage under strong light by avoiding chloroplast positioning toward the anticlinal side of the cell. This light-responsive chloroplast movement relies on the reorganization of chloroplast actin (cp-actin) filaments. Previous studies have suggested that CHLOROPLAST UNUSUAL POSITIONING 1 (CHUP1) is essential for chloroplast photorelocation movement as a regulator of cp-actin filaments. In this study, we conducted comprehensive analyses to understand CHUP1 function. Functional, fluorescently tagged CHUP1 colocalized with and was coordinately reorganized with cp-actin filaments on the chloroplast outer envelope during chloroplast movement in Arabidopsis thaliana. CHUP1 distribution was reversibly regulated in a blue light- and phototropin-dependent manner. X-ray crystallography revealed that the CHUP1-C-terminal domain shares structural homology with the formin homology 2 (FH2) domain, despite lacking sequence similarity. Furthermore, the CHUP1-C-terminal domain promoted actin polymerization in the presence of profilin in vitro. Taken together, our findings indicate that CHUP1 is a plant-specific actin polymerization factor that has convergently evolved to assemble cp-actin filaments and enables chloroplast photorelocation movement.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Actins , Arabidopsis Proteins/genetics , Polymerization , Chloroplast Proteins/genetics , Arabidopsis/genetics , Actin Cytoskeleton , Chloroplasts/physiology , Light , Movement
2.
Life Sci Alliance ; 6(1)2023 01.
Article in English | MEDLINE | ID: mdl-36288901

ABSTRACT

We report a case in which sub-stoichiometric binding of an actin-binding protein has profound structural and functional consequences, providing an insight into the fundamental properties of actin regulation. Rng2 is an IQGAP contained in contractile rings in the fission yeast Schizosaccharomyces pombe Here, we used high-speed atomic force microscopy and electron microscopy and found that sub-stoichiometric binding of the calponin-homology actin-binding domain of Rng2 (Rng2CHD) induces global structural changes in skeletal muscle actin filaments, including shortening of the filament helical pitch. Sub-stoichiometric binding of Rng2CHD also reduced the affinity between actin filaments and muscle myosin II carrying ADP and strongly inhibited the motility of actin filaments on myosin II in vitro. On skeletal muscle myosin II-coated surfaces, Rng2CHD stopped the actin movements at a binding ratio of 11%. Rng2CHD also inhibited actin movements on myosin II of the amoeba Dictyostelium, but in this case, by detaching actin filaments from myosin II-coated surfaces. Thus, sparsely bound Rng2CHD induces apparently cooperative structural changes in actin filaments and inhibits force generation by actomyosin II.


Subject(s)
Dictyostelium , Schizosaccharomyces , Actins/metabolism , Actomyosin/metabolism , Dictyostelium/metabolism , Skeletal Muscle Myosins/metabolism , Myosin Type II/metabolism , Actin Cytoskeleton/metabolism , Schizosaccharomyces/metabolism , Microfilament Proteins/metabolism , Cytoskeletal Proteins/metabolism , Adenosine Diphosphate/metabolism
3.
Genes Cells ; 27(5): 317-330, 2022 May.
Article in English | MEDLINE | ID: mdl-35194888

ABSTRACT

Actin is a major structural component of the cytoskeleton in eukaryotic cells, including fungi, plants, and animals, and exists not only in the cytoplasm as cytoskeleton but also in the nucleus. Recently, we developed a novel actin probe, ß-actin-EGFP fusion protein, which exhibited similar monomeric to filamentous ratio as that of endogenous actin, in contrast to the widely used EGFP-ß-actin fusion protein that over-assembles in cells. Unexpectedly, this novel probe visualized an interconnected meshwork of slightly curved beam-like bundles of actin filaments in the nucleus of U2OS cells. These structures were not labeled with rhodamine phalloidin, Lifeact-EGFP or anti-actin antibodies. In addition, immunofluorescence staining and expression of cofilin-EGFP revealed that this nuclear actin structures contained cofilin. We named these actin filaments as phalloidin-negative intranuclear (PHANIN) actin filaments. Since PHANIN actin filaments could not be detected by general detection methods for actin filaments, we propose that PHANIN actin filaments are different from previously reported nuclear actin structures.


Subject(s)
Actin Cytoskeleton , Actins , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors , Actins/metabolism , Animals , Cell Line, Tumor , Cytoplasm/metabolism , Green Fluorescent Proteins/metabolism , Humans , Phalloidine/analysis , Phalloidine/metabolism
4.
Biochem Biophys Res Commun ; 552: 59-65, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33740665

ABSTRACT

Fimbrin forms bundles of parallel actin filaments in filopodia, but it remains unclear how fimbrin forms well-ordered bundles. To address this issue, we focused on the cooperative interaction between the actin-binding domain of fimbrin and actin filaments. First, we loosely immobilized actin filaments on a glass surface via a positively charged lipid layer and observed the binding of GFP-fused actin-binding domain 2 of fimbrin using fluorescence microscopy. The actin-binding domain formed low-density clusters with unidirectional growth along actin filaments. When the actin filaments were tightly immobilized to the surface by increasing the charge density of the lipid layer, cluster formation was suppressed. This result suggests that the propagation of cooperative structural changes of actin filaments evoked by binding of the actin-binding domain was suppressed by a strong physical interaction with the glass surface. Interestingly, binding of the fimbrin actin-binding domain shortened the length of loosely immobilized actin filaments. Based on these results, we propose that fimbrin-actin interactions accompanied by unidirectional long-range allostery help the formation of well-ordered parallel actin filament bundles.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Surface Extensions/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Binding Sites/genetics , Dictyostelium/genetics , Dictyostelium/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Microscopy, Fluorescence , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Int J Mol Sci ; 21(9)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370032

ABSTRACT

A wide variety of uniquely localized actin-binding proteins (ABPs) are involved in various cellular activities, such as cytokinesis, migration, adhesion, morphogenesis, and intracellular transport. In a micrometer-scale space such as the inside of cells, protein molecules diffuse throughout the cell interior within seconds. In this condition, how can ABPs selectively bind to particular actin filaments when there is an abundance of actin filaments in the cytoplasm? In recent years, several ABPs have been reported to induce cooperative conformational changes to actin filaments allowing structural changes to propagate along the filament cables uni- or bidirectionally, thereby regulating the subsequent binding of ABPs. Such propagation of ABP-induced cooperative conformational changes in actin filaments may be advantageous for the elaborate regulation of cellular activities driven by actin-based machineries in the intracellular space, which is dominated by diffusion. In this review, we focus on long-range allosteric regulation driven by cooperative conformational changes of actin filaments that are evoked by binding of ABPs, and discuss roles of allostery of actin filaments in narrow intracellular spaces.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Actin Cytoskeleton/chemistry , Actins/chemistry , Allosteric Regulation , Animals , Carrier Proteins , Cytoskeleton , Humans , Protein Binding , Tropomyosin/chemistry , Tropomyosin/metabolism
6.
Biophys Rev ; 12(2): 225-232, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32157615

ABSTRACT

Biophysics in Waseda University was started in 1965 as one of the three key research areas that constitute the Physics Department. In the biophysics group, one theoretical lab and two experimental labs are now working on the cutting-edge themes on biophysics, disseminating the ideas and knowledge of biophysics to undergraduate and graduate students from the viewpoint of physics.

7.
Genes Cells ; 25(1): 6-21, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31957229

ABSTRACT

Motility often plays a decisive role in the survival of species. Five systems of motility have been studied in depth: those propelled by bacterial flagella, eukaryotic actin polymerization and the eukaryotic motor proteins myosin, kinesin and dynein. However, many organisms exhibit surprisingly diverse motilities, and advances in genomics, molecular biology and imaging have showed that those motilities have inherently independent mechanisms. This makes defining the breadth of motility nontrivial, because novel motilities may be driven by unknown mechanisms. Here, we classify the known motilities based on the unique classes of movement-producing protein architectures. Based on this criterion, the current total of independent motility systems stands at 18 types. In this perspective, we discuss these modes of motility relative to the latest phylogenetic Tree of Life and propose a history of motility. During the ~4 billion years since the emergence of life, motility arose in Bacteria with flagella and pili, and in Archaea with archaella. Newer modes of motility became possible in Eukarya with changes to the cell envelope. Presence or absence of a peptidoglycan layer, the acquisition of robust membrane dynamics, the enlargement of cells and environmental opportunities likely provided the context for the (co)evolution of novel types of motility.


Subject(s)
Cell Movement/genetics , Cell Movement/physiology , Flagella/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Bacteria , Biological Evolution , Dyneins/metabolism , Evolution, Molecular , Flagella/genetics , Humans , Kinesins/metabolism , Myosins/metabolism , Phylogeny
8.
Cells ; 8(8)2019 07 26.
Article in English | MEDLINE | ID: mdl-31357517

ABSTRACT

Dynamin is a large GTPase responsible for diverse cellular processes, such as endocytosis, division of organelles, and cytokinesis. The social amoebozoan, Dictyostelium discoideum, has five dynamin-like proteins: dymA, dymB, dlpA, dlpB, and dlpC. DymA, dlpA, or dlpB-deficient cells exhibited defects in cytokinesis. DlpA and dlpB were found to colocalize at cleavage furrows from the early phase, and dymA localized at the intercellular bridge connecting the two daughter cells, indicating that these dynamins contribute to cytokinesis at distinct dividing stages. Total internal reflection fluorescence microscopy revealed that dlpA and dlpB colocalized at individual dots at the furrow cortex. However, dlpA and dlpB did not colocalize with clathrin, suggesting that they are not involved in clathrin-mediated endocytosis. The fact that dlpA did not localize at the furrow in dlpB null cells and vice versa, as well as other several lines of evidence, suggests that hetero-oligomerization of dlpA and dlpB is required for them to bind to the furrow. The hetero-oligomers directly or indirectly associate with actin filaments, stabilizing them in the contractile rings. Interestingly, dlpA, but not dlpB, accumulated at the phagocytic cups independently of dlpB. Our results suggest that the hetero-oligomers of dlpA and dlpB contribute to cytokinesis cooperatively with dymA.


Subject(s)
Cytokinesis , Dictyostelium/physiology , Dynamins/metabolism , Actins/metabolism , Endocytosis , Fluorescent Antibody Technique , Humans , Protein Binding , Protein Transport , Proteolysis , Protozoan Proteins/metabolism
9.
PLoS One ; 14(4): e0214736, 2019.
Article in English | MEDLINE | ID: mdl-30946777

ABSTRACT

Although the distinct distribution of certain molecules along the anterior or posterior edge is essential for directed cell migration, the mechanisms to maintain asymmetric protein localization have not yet been fully elucidated. Here, we studied a mechanism for the distinct localizations of two Dictyostelium talin homologues, talin A and talin B, both of which play important roles in cell migration and adhesion. Using GFP fusion, we found that talin B, as well as its C-terminal actin-binding region, which consists of an I/LWEQ domain and a villin headpiece domain, was restricted to the leading edge of migrating cells. This is in sharp contrast to talin A and its C-terminal actin-binding domain, which co-localized with myosin II along the cell posterior cortex, as reported previously. Intriguingly, even in myosin II-null cells, talin A and its actin-binding domain displayed a specific distribution, co-localizing with stretched actin filaments. In contrast, talin B was excluded from regions rich in stretched actin filaments, although a certain amount of its actin-binding region alone was present in those areas. When cells were sucked by a micro-pipette, talin B was not detected in the retracting aspirated lobe where acto-myosin, talin A, and the actin-binding regions of talin A and talin B accumulated. Based on these results, we suggest that talin A predominantly interacts with actin filaments stretched by myosin II through its C-terminal actin-binding region, while the actin-binding region of talin B does not make such distinctions. Furthermore, talin B appears to have an additional, unidentified mechanism that excludes it from the region rich in stretched actin filaments. We propose that these actin-binding properties play important roles in the anterior and posterior enrichment of talin B and talin A, respectively, during directed cell migration.


Subject(s)
Cell Movement , Dictyostelium/metabolism , Protozoan Proteins/analysis , Talin/analysis , Actin Cytoskeleton/metabolism , Binding Sites , Lipid Metabolism , Lipids/chemistry , Protein Domains , Protozoan Proteins/chemistry , Protozoan Proteins/physiology , Talin/chemistry , Talin/physiology
10.
Sci Rep ; 9(1): 5353, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926871

ABSTRACT

Mutation of the Lys-336 residue of actin to Ile (K336I) or Asp (K336E) causes congenital myopathy. To understand the effect of this mutation on the function of actin filaments and gain insight into the mechanism of disease onset, we prepared and biochemically characterised K336I mutant actin from Dictyostelium discoideum. Subtilisin cleavage assays revealed that the structure of the DNase-I binding loop (D-loop) of monomeric K336I actin, which would face the adjacent actin-protomer in filaments, differed from that of wild type (WT) actin. Although K336I actin underwent normal salt-dependent reversible polymerisation and formed apparently normal filaments, interactions of K336I filaments with alpha-actinin, myosin II, and cofilin were disrupted. Furthermore, co-filaments of K336I and WT actins also exhibited abnormal interactions with cofilin, implying that K336I actin altered the structure of the neighbouring WT actin protomers such that interaction between cofilin and the WT actin protomers was prevented. We speculate that disruption of the interactions between co-filaments and actin-binding proteins is the primary reason why the K336I mutation induces muscle disease in a dominant fashion.


Subject(s)
Actins/genetics , Actins/metabolism , Alleles , Amino Acid Substitution , Microfilament Proteins/metabolism , Mutation , Promoter Regions, Genetic , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/administration & dosage , Actin Depolymerizing Factors/metabolism , Actins/chemistry , Myosins/metabolism , Protein Binding , Protein Multimerization , Recombinant Proteins
11.
Plasmid ; 98: 37-44, 2018 06.
Article in English | MEDLINE | ID: mdl-30196057

ABSTRACT

The CRISPR/Cas9 system is a powerful genome editing tool for disrupting the expression of specific genes in a variety of cells. However, the genome editing procedure using currently available vectors is laborious, and there is room for improvement to obtain knockout cells more efficiently. Therefore, we constructed a novel vector for high efficiency genome editing, named pGedit, which contains EGFP-Bsr as a selection marker, expression units of Cas9, and sgRNA without a terminator sequence of the U6 promoter. EGFP-Bsr is a fusion protein of EGFP and blasticidin S deaminase, and enables rapid selection and monitoring of transformants, as well as confirmation that the vector has not been integrated into the genome. By using pGedit, we targeted human ACTB, ACTG1 and mouse Nes genes coding for ß-actin, γ-actin and nestin, respectively. Knockout cell lines of each gene were easily and efficiently obtained in all three cases. In this report, we show that our novel vector, pGedit, significantly facilitates genome editing.


Subject(s)
Actins/antagonists & inhibitors , CRISPR-Cas Systems , Gene Editing/methods , Genetic Vectors , Nestin/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Actins/genetics , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Base Sequence , Gene Targeting , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Nestin/genetics , Promoter Regions, Genetic , Sequence Homology
12.
Sci Rep ; 8(1): 4381, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29531328

ABSTRACT

Flowering plants express multiple actin isoforms. Previous studies suggest that individual actin isoforms have specific functions; however, the subcellular localization of actin isoforms in plant cells remains obscure. Here, we transiently expressed and observed major Arabidopsis vegetative actin isoforms, AtACT2 and AtACT7, as fluorescent-fusion proteins. By optimizing the linker sequence between fluorescent protein and actin, we succeeded in observing filaments that contained these expressed actin isoforms fused with green fluorescent protein (GFP) in Arabidopsis protoplasts. Different colored fluorescent proteins fused with AtACT2 and AtACT7 and co-expressed in Nicotiana benthamiana mesophyll cells co-polymerized in a segregated manner along filaments. In epidermal cells, surprisingly, AtACT2 and AtACT7 tended to polymerize into different types of filaments. AtACT2 was incorporated into thinner filaments, whereas AtACT7 was incorporated into thick bundles. We conclude that different actin isoforms are capable of constructing unique filament arrays, depending on the cell type or tissue. Interestingly, staining patterns induced by two indirect actin filament probes, Lifeact and mTalin1, were different between filaments containing AtACT2 and those containing AtACT7. We suggest that filaments containing different actin isoforms bind specific actin-binding proteins in vivo, since the two probes comprise actin-binding domains from different actin-binding proteins.


Subject(s)
Actin Cytoskeleton/chemistry , Actins/genetics , Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , Actins/chemistry , Actins/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Green Fluorescent Proteins/metabolism , Microfilament Proteins/metabolism , Polymerization , Protein Binding , Protein Isoforms
13.
Mol Biol Cell ; 29(8): 911-922, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29467250

ABSTRACT

We demonstrated that myosin IIA and IIB are essential for the formation of transverse arcs and ventral stress fibers, respectively. Furthermore, we illustrated the roles of both isoforms in lamellar flattening and also raised the possibility that actin filaments in ventral stress fibers are in a stretched conformation.


Subject(s)
Cell Movement , Fibroblasts/cytology , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Stress Fibers/metabolism , Cell Line , Cell Polarity , Cell Shape , Gene Knockdown Techniques , Humans , Phosphorylation
14.
Biochem Biophys Res Commun ; 495(3): 2145-2151, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29248727

ABSTRACT

There are two classes of myosin, XI and VIII, in higher plants. Myosin XI moves actin filaments at high speed and its enzyme activity is also very high. In contrast, myosin VIII moves actin filaments very slowly with very low enzyme activity. Because most of these enzymatic and motile activities were measured using animal skeletal muscle α-actin, but not plant actin, they would not accurately reflect the actual activities in plant cells. We thus measured enzymatic and motile activities of the motor domains of two Arabidopsis myosin XI isoforms (MYA2, XI-B), and one Arabidopsis myosin VIII isoform (ATM1), by using three Arabidopsis actin isoforms (ACT1, ACT2, and ACT7). The measured activities were different from those measured by using muscle actin. Moreover, Arabidopsis myosins showed different enzymatic and motile activities when using different Arabidopsis actin isoforms. Our results suggest that plant actin should be used for measuring enzymatic and motile activities of plant myosins and that different actin isoforms in plant cells might function as different tracks along which affinities and velocities of each myosin isoform are modulated.


Subject(s)
Actins/chemistry , Arabidopsis Proteins/chemistry , Molecular Motor Proteins/chemistry , Motion , Myosins/chemistry , Actins/ultrastructure , Arabidopsis Proteins/ultrastructure , Enzyme Activation , Molecular Motor Proteins/ultrastructure , Myosins/ultrastructure , Protein Binding
15.
Cell Struct Funct ; 42(2): 131-140, 2017.
Article in English | MEDLINE | ID: mdl-28855440

ABSTRACT

Actin, a major component of microfilaments, is involved in various eukaryotic cellular functions. Over the past two decades, actin fused with fluorescent protein has been used as a probe to detect the organization and dynamics of the actin cytoskeleton in living eukaryotic cells. It is generally assumed that the expression of fusion protein of fluorescent protein does not disturb the distribution of endogenous actin throughout the cell, and that the distribution of the fusion protein reflects that of endogenous actin. However, we noticed that EGFP-ß-actin caused the excessive formation of microfilaments in several mammalian cell lines. To investigate whether the position of the EGFP tag on actin affected the formation of filaments, we constructed an expression vector harboring a ß-actin-EGFP gene. In contrast to EGFP-ß-actin, cells expressing ß-actin-EGFP showed actin filaments in a high background from the monomer actin in cytosol. Additionally, the detergent insoluble assay revealed that the majority of the detergent-insoluble cytoskeleton from cells expressing EGFP-ß-actin was recovered in the pellet. Furthermore, we found that the expression of EGFP-ß-actin affects the migration of NBT-L2b cells and the mechanical stiffness of U2OS cells. These results indicate that EGFP fused to the N-terminus of actin tend to form excessive actin filaments. In addition, EGFP-actin affects both the cellular morphological and physiological phenotypes as compared to actin-EGFP.Key words: actin, GFP, cytoskeleton and probe.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Green Fluorescent Proteins/metabolism , Actin Cytoskeleton/chemistry , Actins/analysis , Actins/chemistry , Animals , Cell Line , Cytosol/chemistry , Cytosol/metabolism , Green Fluorescent Proteins/analysis , Humans , Mammals
16.
Cytoskeleton (Hoboken) ; 74(12): 482-489, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28888060

ABSTRACT

Heavy meromyosin (HMM) forms clusters along actin filaments under low ATP concentrations. Here, we observed the growth of HMM clusters under low concentrations of ATP in real time using fluorescence microscopy. When actin filaments were loosely immobilized on positively charged lipid bilayers, clusters of HMM-GFP were readily formed. Time-lapse observation revealed that the clusters grew unidirectionally. When we used a mixture of actin filaments and copolymers of actin and acto-S1dC, a chimeric protein of actin and the myosin motor domain, HMM-GFP preferentially formed clusters along the copolymers. We thus suggest that binding of myosin motors carrying ADP and Pi induces unidirectional conformational changes in actin filaments and allosterically recruits more myosin binding. In contrast, when actin filaments and copolymers were anchored to glass substrate via stable biotin-avidin linkage, higher concentrations of HMM-GFP were required to form clusters than on the lipid bilayer. Moreover, actin filaments and copolymers were not discriminated regarding preferential cluster formation. This is presumably because the myosin-induced cooperative conformational changes in actin filaments involve changes in the helical twist. Consistent with this, cofilin clusters, which supertwist the helix, were readily formed along loosely immobilized actin filaments, but not along those anchored via biotin-avidin linkage.


Subject(s)
Actin Cytoskeleton/chemistry , Dictyostelium/chemistry , Myosin Subfragments/chemistry , Protozoan Proteins/chemistry , Microscopy, Fluorescence
17.
PLoS One ; 12(7): e0181171, 2017.
Article in English | MEDLINE | ID: mdl-28742155

ABSTRACT

We examined the movement of an actin filament sliding on a mixture of normal and genetically modified myosin molecules that were attached to a glass surface. For this purpose, we used a Dictyostelium G680V mutant myosin II whose release rates of Pi and ADP were highly suppressed relative to normal myosin, leading to a significantly extended life-time of the strongly bound state with actin and virtually no motility. When the mixing ratio of G680V mutant myosin II to skeletal muscle HMM (heavy myosin) was 0.01%, the actin filaments moved intermittently. When they moved, their sliding velocities were about two-fold faster than the velocity of skeletal HMM alone. Furthermore, sliding movements were also faster when the actin filaments were allowed to slide on skeletal muscle HMM-coated glass surfaces in the motility buffer solution containing G680V HMM. In this case no intermittent movement was observed. When the actin filaments used were copolymerized with a fusion protein consisting of Dictyostelium actin and Dictyostelium G680V myosin II motor domain, similar faster sliding movements were observed on skeletal muscle HMM-coated surfaces. The filament sliding velocities were about two-fold greater than the velocities of normal actin filaments. We found that the velocity of actin filaments sliding on skeletal muscle myosin molecules increased in the presence of a non-motile G680V mutant myosin motor.


Subject(s)
Actins/metabolism , Myosin Subfragments/metabolism , Myosin Type II/metabolism , Actins/chemistry , Animals , Dictyostelium/genetics , Dictyostelium/metabolism , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , In Vitro Techniques , Microscopy, Electron, Transmission , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Movement/physiology , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Muscle, Skeletal/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myosin Subfragments/chemistry , Myosin Type II/chemistry , Myosin Type II/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
Ann Biomed Eng ; 45(10): 2475-2486, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28744841

ABSTRACT

The use of magnetic nanoparticles (MNPs) is a promising technique for future advances in biomedical applications. This idea is supported by the availability of MNPs that can target specific cell components, the variety of shapes of MNPs and the possibility of finely controlling the applied magnetic forces. To examine this opportunity, here we review the current developments in the use of MNPs to mechanically stimulate cells and, specifically, the cell mechanotransduction systems. We analyze the cell components that may act as mechanosensors and their effect on cell fate and we focus on the promising possibilities of controlling stem-cell differentiation, inducing cancer-cell death and treating nervous-system diseases.


Subject(s)
Cell Differentiation , Magnetic Fields , Mechanotransduction, Cellular , Neoplasms , Nervous System Diseases , Stem Cells/metabolism , Animals , Cell Death , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Nervous System Diseases/therapy , Stem Cells/pathology
19.
Theranostics ; 7(6): 1735-1748, 2017.
Article in English | MEDLINE | ID: mdl-28529648

ABSTRACT

Magnetic nanoparticles (MNPs) functionalized with targeting moieties can recognize specific cell components and induce mechanical actuation under magnetic field. Their size is adequate for reaching tumors and targeting cancer cells. However, due to the nanometric size, the force generated by MNPs is smaller than the force required for largely disrupting key components of cells. Here, we show the magnetic assembly process of the nanoparticles inside the cells, to form elongated aggregates with the size required to produce elevated mechanical forces. We synthesized iron oxide nanoparticles doped with zinc, to obtain high magnetization, and functionalized with the epidermal growth factor (EGF) peptide for targeting cancer cells. Under a low frequency rotating magnetic field at 15 Hz and 40 mT, the internalized EGF-MNPs formed elongated aggregates and generated hundreds of pN to dramatically damage the plasma and lysosomal membranes. The physical disruption, including leakage of lysosomal hydrolases into the cytosol, led to programmed cell death and necrosis. Our work provides a novel strategy of designing magnetic nanomedicines for mechanical destruction of cancer cells.


Subject(s)
Epidermal Growth Factor/metabolism , Ferric Compounds/metabolism , Glioblastoma , Magnetic Field Therapy/methods , Metal Nanoparticles , Molecular Targeted Therapy/methods , Cell Death , Cell Line, Tumor , Humans
20.
Sci Rep ; 6: 35449, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27762277

ABSTRACT

Heavy meromyosin (HMM) of myosin II and cofilin each binds to actin filaments cooperatively and forms clusters along the filaments, but it is unknown whether the two cooperative bindings are correlated and what physiological roles they have. Fluorescence microscopy demonstrated that HMM-GFP and cofilin-mCherry each bound cooperatively to different parts of actin filaments when they were added simultaneously in 0.2 µM ATP, indicating that the two cooperative bindings are mutually exclusive. In 0.1 mM ATP, the motor domain of myosin (S1) strongly inhibited the formation of cofilin clusters along actin filaments. Under this condition, most actin protomers were unoccupied by S1 at any given moment, suggesting that transiently bound S1 alters the structure of actin filaments cooperatively and/or persistently to inhibit cofilin binding. Consistently, cosedimentation experiments using copolymers of actin and actin-S1 fusion protein demonstrated that the fusion protein affects the neighboring actin protomers, reducing their affinity for cofilin. In reciprocal experiments, cofilin-actin fusion protein reduced the affinity of neighboring actin protomers for S1. Thus, allosteric regulation by cooperative conformational changes of actin filaments contributes to mutually exclusive cooperative binding of myosin II and cofilin to actin filaments, and presumably to the differential localization of both proteins in cells.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Myosins/metabolism , Protein Conformation , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Humans , Microscopy, Fluorescence , Models, Biological , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...