Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Elife ; 132024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39093938

RESUMEN

The mechanism underlying the preferential and cooperative binding of cofilin and the expansion of clusters toward the pointed-end side of actin filaments remains poorly understood. To address this, we conducted a principal component analysis based on available filamentous actin (F-actin) and C-actin (cofilins were excluded from cofilactin) structures and compared to monomeric G-actin. The results strongly suggest that C-actin, rather than F-ADP-actin, represented the favourable structure for binding preference of cofilin. High-speed atomic force microscopy explored that the shortened bare half helix adjacent to the cofilin clusters on the pointed end side included fewer actin protomers than normal helices. The mean axial distance (MAD) between two adjacent actin protomers along the same long-pitch strand within shortened bare half helices was longer (5.0-6.3 nm) than the MAD within typical helices (4.3-5.6 nm). The inhibition of torsional motion during helical twisting, achieved through stronger attachment to the lipid membrane, led to more pronounced inhibition of cofilin binding and cluster formation than the presence of inorganic phosphate (Pi) in solution. F-ADP-actin exhibited more naturally supertwisted half helices than F-ADP.Pi-actin, explaining how Pi inhibits cofilin binding to F-actin with variable helical twists. We propose that protomers within the shorter bare helical twists, either influenced by thermal fluctuation or induced allosterically by cofilin clusters, exhibit characteristics of C-actin-like structures with an elongated MAD, leading to preferential and cooperative binding of cofilin.


Asunto(s)
Factores Despolimerizantes de la Actina , Actinas , Unión Proteica , Actinas/metabolismo , Actinas/química , Factores Despolimerizantes de la Actina/metabolismo , Factores Despolimerizantes de la Actina/química , Microscopía de Fuerza Atómica , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/química , Conformación Proteica , Modelos Moleculares , Animales
2.
Plant Cell ; 36(4): 1159-1181, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38134410

RESUMEN

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.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Actinas , Proteínas de Arabidopsis/genética , Polimerizacion , Proteínas de Cloroplastos/genética , Arabidopsis/genética , Citoesqueleto de Actina , Cloroplastos/fisiología , Luz , Movimiento
3.
Life Sci Alliance ; 6(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36288901

RESUMEN

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.


Asunto(s)
Dictyostelium , Schizosaccharomyces , Actinas/metabolismo , Actomiosina/metabolismo , Dictyostelium/metabolismo , Miosinas del Músculo Esquelético/metabolismo , Miosina Tipo II/metabolismo , Citoesqueleto de Actina/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas del Citoesqueleto/metabolismo , Adenosina Difosfato/metabolismo
4.
Genes Cells ; 27(5): 317-330, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35194888

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina , Actinas , Citoesqueleto de Actina/metabolismo , Factores Despolimerizantes de la Actina , Actinas/metabolismo , Animales , Línea Celular Tumoral , Citoplasma/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Faloidina/análisis , Faloidina/metabolismo
5.
Biochem Biophys Res Commun ; 552: 59-65, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33740665

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Extensiones de la Superficie Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Sitios de Unión/genética , Dictyostelium/genética , Dictyostelium/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Microfilamentos/genética , Microscopía Fluorescente , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
6.
Int J Mol Sci ; 21(9)2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32370032

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Citoesqueleto de Actina/química , Actinas/química , Regulación Alostérica , Animales , Proteínas Portadoras , Citoesqueleto , Humanos , Unión Proteica , Tropomiosina/química , Tropomiosina/metabolismo
7.
Biophys Rev ; 12(2): 225-232, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32157615

RESUMEN

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.

8.
Genes Cells ; 25(1): 6-21, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31957229

RESUMEN

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.


Asunto(s)
Movimiento Celular/genética , Movimiento Celular/fisiología , Flagelos/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animales , Bacterias , Evolución Biológica , Dineínas/metabolismo , Evolución Molecular , Flagelos/genética , Humanos , Cinesinas/metabolismo , Miosinas/metabolismo , Filogenia
9.
Cells ; 8(8)2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31357517

RESUMEN

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.


Asunto(s)
Citocinesis , Dictyostelium/fisiología , Dinaminas/metabolismo , Actinas/metabolismo , Endocitosis , Técnica del Anticuerpo Fluorescente , Humanos , Unión Proteica , Transporte de Proteínas , Proteolisis , Proteínas Protozoarias/metabolismo
10.
PLoS One ; 14(4): e0214736, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30946777

RESUMEN

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.


Asunto(s)
Movimiento Celular , Dictyostelium/metabolismo , Proteínas Protozoarias/análisis , Talina/análisis , Citoesqueleto de Actina/metabolismo , Sitios de Unión , Metabolismo de los Lípidos , Lípidos/química , Dominios Proteicos , Proteínas Protozoarias/química , Proteínas Protozoarias/fisiología , Talina/química , Talina/fisiología
11.
Sci Rep ; 9(1): 5353, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926871

RESUMEN

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.


Asunto(s)
Actinas/genética , Actinas/metabolismo , Alelos , Sustitución de Aminoácidos , Proteínas de Microfilamentos/metabolismo , Mutación , Regiones Promotoras Genéticas , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Factores Despolimerizantes de la Actina/administración & dosificación , Factores Despolimerizantes de la Actina/metabolismo , Actinas/química , Miosinas/metabolismo , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes
12.
Plasmid ; 98: 37-44, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30196057

RESUMEN

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.


Asunto(s)
Actinas/antagonistas & inhibidores , Sistemas CRISPR-Cas , Edición Génica/métodos , Vectores Genéticos , Nestina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Actinas/genética , Aminohidrolasas/genética , Aminohidrolasas/metabolismo , Animales , Secuencia de Bases , Marcación de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Nestina/genética , Regiones Promotoras Genéticas , Homología de Secuencia
13.
Sci Rep ; 8(1): 4381, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29531328

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/química , Actinas/genética , Proteínas de Arabidopsis/química , Arabidopsis/química , Actinas/química , Actinas/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Microfilamentos/metabolismo , Polimerizacion , Unión Proteica , Isoformas de Proteínas
14.
Mol Biol Cell ; 29(8): 911-922, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29467250

RESUMEN

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.


Asunto(s)
Movimiento Celular , Fibroblastos/citología , Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Fibras de Estrés/metabolismo , Línea Celular , Polaridad Celular , Forma de la Célula , Técnicas de Silenciamiento del Gen , Humanos , Fosforilación
15.
Biochem Biophys Res Commun ; 495(3): 2145-2151, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29248727

RESUMEN

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.


Asunto(s)
Actinas/química , Proteínas de Arabidopsis/química , Proteínas Motoras Moleculares/química , Movimiento (Física) , Miosinas/química , Actinas/ultraestructura , Proteínas de Arabidopsis/ultraestructura , Activación Enzimática , Proteínas Motoras Moleculares/ultraestructura , Miosinas/ultraestructura , Unión Proteica
16.
Cell Struct Funct ; 42(2): 131-140, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28855440

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Citoesqueleto de Actina/química , Actinas/análisis , Actinas/química , Animales , Línea Celular , Citosol/química , Citosol/metabolismo , Proteínas Fluorescentes Verdes/análisis , Humanos , Mamíferos
17.
Cytoskeleton (Hoboken) ; 74(12): 482-489, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28888060

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/química , Dictyostelium/química , Subfragmentos de Miosina/química , Proteínas Protozoarias/química , Microscopía Fluorescente
18.
Ann Biomed Eng ; 45(10): 2475-2486, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28744841

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Campos Magnéticos , Mecanotransducción Celular , Neoplasias , Enfermedades del Sistema Nervioso , Células Madre/metabolismo , Animales , Muerte Celular , Humanos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/terapia , Células Madre/patología
19.
PLoS One ; 12(7): e0181171, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28742155

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Subfragmentos de Miosina/metabolismo , Miosina Tipo II/metabolismo , Actinas/química , Animales , Dictyostelium/genética , Dictyostelium/metabolismo , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Técnicas In Vitro , Microscopía Electrónica de Transmisión , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Movimiento/fisiología , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Músculo Esquelético/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Subfragmentos de Miosina/química , Miosina Tipo II/química , Miosina Tipo II/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Conejos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
20.
Theranostics ; 7(6): 1735-1748, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28529648

RESUMEN

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.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Compuestos Férricos/metabolismo , Glioblastoma , Magnetoterapia/métodos , Nanopartículas del Metal , Terapia Molecular Dirigida/métodos , Muerte Celular , Línea Celular Tumoral , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA