Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Traffic ; 24(10): 463-474, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37679870

RESUMEN

To understand force generation under a wide range of loads, the stepping of single kinesin molecules was measured at loads from -20 to 42 pN by optical tweezers with high temporal resolution. The optical trap has been improved to halve positional noise and increase bandwidth by using 200-nm beads. The step size of the forward and backward steps was 8.2 nm even over a wide range of loads. Histograms of the dwell times of backward steps and detachment fit well to two independent exponential equations with fast (~0.4 ms) and slow (>3 ms) time constants, indicating the existence of a fast step in addition to the conventional slow step. The dwell times of the fast steps were almost independent of the load and ATP concentration, while those of the slow backward steps and detachment depended on those. We constructed the kinetic model to explain the fast and slow steps under a wide range of loads.


Asunto(s)
Cinesinas , Cinética
2.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34088833

RESUMEN

Changes in the molecular properties of cardiac myosin strongly affect the interactions of myosin with actin that result in cardiac contraction and relaxation. However, it remains unclear how myosin molecules work together in cardiac myofilaments and which properties of the individual myosin molecules impact force production to drive cardiac contractility. Here, we measured the force production of cardiac myofilaments using optical tweezers. The measurements revealed that stepwise force generation was associated with a higher frequency of backward steps at lower loads and higher stall forces than those of fast skeletal myofilaments. To understand these unique collective behaviors of cardiac myosin, the dynamic responses of single cardiac and fast skeletal myosin molecules, interacting with actin filaments, were evaluated under load. The cardiac myosin molecules switched among three distinct conformational positions, ranging from pre- to post-power stroke positions, in 1 mM ADP and 0 to 10 mM phosphate solution. In contrast to cardiac myosin, fast skeletal myosin stayed primarily in the post-power stroke position, suggesting that cardiac myosin executes the reverse stroke more frequently than fast skeletal myosin. To elucidate how the reverse stroke affects the force production of myofilaments and possibly heart function, a simulation model was developed that combines the results from the single-molecule and myofilament experiments. The results of this model suggest that the reversal of the cardiac myosin power stroke may be key to characterizing the force output of cardiac myosin ensembles and possibly to facilitating heart contractions.


Asunto(s)
Contracción Miocárdica , Miocardio/metabolismo , Miofibrillas/metabolismo , Miosinas/metabolismo , Animales , Porcinos
3.
Sensors (Basel) ; 21(2)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33450927

RESUMEN

Elucidation of cell-level transport mediated by vesicles within a living cell provides key information regarding viral infection processes and also drug delivery mechanisms. Although the single-particle tracking method has enabled clear analysis of individual vesicle trajectories, information regarding the entire cell-level intracellular transport is hardly obtainable, due to the difficulty in collecting a large dataset with current methods. In this paper, we propose a visualization method of vesicle transport using optical flow, based on geometric cell center estimation and vector analysis, for measuring the trafficking directions. As a quantitative visualization method for determining the intracellular transport status, the proposed method is expected to be universally exploited in various biomedical cell image analyses.


Asunto(s)
Flujo Optico , Procesamiento de Imagen Asistido por Computador
4.
Biophys J ; 115(10): 1981-1992, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30396511

RESUMEN

Dimeric motor proteins, kinesin-1, cytoplasmic dynein-1, and myosin-V, move stepwise along microtubules and actin filaments with a regular step size. The motors take backward as well as forward steps. The step ratio r and dwell time τ, which are the ratio of the number of backward steps to the number of forward steps and the time between consecutive steps, respectively, were observed to change with the load. To understand the movement of motor proteins, we constructed a unified and simple mathematical model to explain the load dependencies of r and of τ measured for the above three types of motors quantitatively. Our model consists of three states, and the forward and backward steps are represented by the cycles of transitions visiting different pairs of states among the three, implying that a backward step is not the reversal of a forward step. Each of r and τ is given by a simple expression containing two exponential functions. The experimental data for r and τ for dynein available in the literature are not sufficient for a quantitative analysis, which is in contrast to those for kinesin and myosin-V. We reanalyze the data to obtain r and τ of native dynein to make up the insufficient data to fit them to the model. Our model successfully describes the behavior of r and τ for all of the motors in a wide range of loads from large assisting loads to superstall loads.


Asunto(s)
Cinesinas/química , Cinesinas/metabolismo , Fenómenos Mecánicos , Modelos Moleculares , Multimerización de Proteína , Fenómenos Biomecánicos , Miosina Tipo V/metabolismo , Estructura Cuaternaria de Proteína
5.
Opt Express ; 26(13): 16236-16249, 2018 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-30119458

RESUMEN

The detection of the precise movement of a vesicle during transport in a live cell provides key information for the intracellular delivery process. Here we report a novel numerical method for analyzing three-dimensional vesicle movement. Since the vesicle moves along a linear cytoskeleton during the active transport, our method first detects the orientation and position of the cytoskeleton as a linear section based on angle correlation and linear regression, after noise reduction. Then, the precise vesicle movement is calculated using vector analysis, in terms of rotation angle and translational displacement. Using this method, various vesicle trajectories obtained via high spatiotemporal resolution microscopy can be understood..


Asunto(s)
Transporte Biológico Activo/fisiología , Vesículas Citoplasmáticas/fisiología , Citoesqueleto/metabolismo , Neoplasias Hepáticas/metabolismo , Humanos , Imagenología Tridimensional , Neoplasias Hepáticas/patología , Células Tumorales Cultivadas
6.
Sci Technol Adv Mater ; 17(1): 473-482, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27877897

RESUMEN

The processes involved in malignant gliomas damage were quantitatively evaluated by microscopy. The near-infrared fluorescent dye IR700 that is conjugated to an anti-CD133 antibody (IR700-CD133) specifically targets malignant gliomas (U87MG) and stem cells (BT142) and is endocytosed into the cells. The gliomas are then photodamaged by the release of reactive oxygen species (ROS) and the heat induced by illumination of IR700 by a red laser, and the motility of the vesicles within these cells is altered as a result of cellular damage. To investigate these changes in motility, we developed a new method that measures fluctuations in the intensity of phase-contrast images obtained from small areas within cells. The intensity fluctuation in U87MG cells gradually decreased as cell damage progressed, whereas the fluctuation in BT142 cells increased. The endocytosed IR700 dye was co-localized in acidic organelles such as endosomes and lysosomes. The pH in U87MG cells, as monitored by a pH indicator, was decreased and then gradually increased by the illumination of IR700, while the pH in BT142 cells increased monotonically. In these experiments, the processes of cell damage were quantitatively evaluated according to the motility of vesicles and changes in pH.

7.
Cell Mol Life Sci ; 70(22): 4275-92, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23685901

RESUMEN

In muscles, the arrays of skeletal myosin molecules interact with actin filaments and continuously generate force at various contraction speeds. Therefore, it is crucial for myosin molecules to generate force collectively and minimize the interference between individual myosin molecules. Knowledge of the elasticity of myosin molecules is crucial for understanding the molecular mechanisms of muscle contractions because elasticity directly affects the working and drag (resistance) force generation when myosin molecules are positively or negatively strained. The working stroke distance is also an important mechanical property necessary for elucidation of the thermodynamic efficiency of muscle contractions at the molecular level. In this review, we focus on these mechanical properties obtained from single-fiber and single-molecule studies and discuss recent findings associated with these mechanical properties. We also discuss the potential molecular mechanisms associated with reduction of the drag effect caused by negatively strained myosin molecules.


Asunto(s)
Músculo Esquelético/fisiología , Miosinas/metabolismo , Fenómenos Biomecánicos , Elasticidad , Humanos , Modelos Biológicos , Contracción Muscular
8.
Development ; 137(18): 3037-46, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20685736

RESUMEN

Motile cilia generate constant fluid flow over epithelial tissue, and thereby influence diverse physiological processes. Such functions of ciliated cells depend on the planar polarity of the cilia and on their basal bodies being oriented in the downstream direction of fluid flow. Recently, another type of basal body planar polarity, characterized by the anterior localization of the basal bodies in individual cells, was reported in the multiciliated ependymal cells that line the surface of brain ventricles. However, little is known about the cellular and molecular mechanisms by which this polarity is established. Here, we report in mice that basal bodies move in the apical cell membrane during differentiation to accumulate in the anterior region of ependymal cells. The planar cell polarity signaling pathway influences basal body orientation, but not their anterior migration, in the neonatal brain. Moreover, we show by pharmacological and genetic studies that non-muscle myosin II is a key regulator of this distribution of basal bodies. This study demonstrates that the orientation and distribution of basal bodies occur by distinct mechanisms.


Asunto(s)
Movimiento Celular , Polaridad Celular , Epéndimo/crecimiento & desarrollo , Epéndimo/metabolismo , Miosina Tipo II/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Cilios/metabolismo , Epéndimo/citología , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Rastreo , Miosina Tipo II/genética , Biosíntesis de Proteínas
9.
Sci Rep ; 13(1): 10514, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37386019

RESUMEN

Dynein is a minus-end-directed motor that generates oscillatory motion in eukaryotic flagella. Cyclic beating, which is the most significant feature of a flagellum, occurs by sliding spatiotemporal regulation by dynein along microtubules. To elucidate oscillation generated by dynein in flagellar beating, we examined its mechanochemical properties under three different axonemal dissection stages. By starting from the intact 9 + 2 structure, we reduced the number of interacting doublets and determined three parameters, namely, the duty ratio, dwell time and step size, of the generated oscillatory forces at each stage. Intact dynein molecules in the axoneme, doublet bundle and single doublet were used to measure the force with optical tweezers. The mean forces per dynein determined under three axonemal conditions were smaller than the previously reported stall forces of axonemal dynein; this phenomenon suggests that the duty ratio is lower than previously thought. This possibility was further confirmed by an in vitro motility assay with purified dynein. The dwell time and step size estimated from the measured force were similar. The similarity in these parameters suggests that the essential properties of dynein oscillation are inherent to the molecule and independent of the axonemal architecture, composing the functional basis of flagellar beating.


Asunto(s)
Dineínas Axonemales , Axonema , Cilios
10.
J Biol Chem ; 286(12): 10581-92, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21199870

RESUMEN

The mechanism by which HIV-1-Tat protein transduction domain (TatP) enters the cell remains unclear because of an insufficient understanding of the initial kinetics of peptide entry. Here, we report the successful visualization and tracking of TatP molecular kinetics on the cell surface with 7-nm spatial precision using quantum dots. Strong cell binding was only observed with a TatP valence of ≥8, whereas monovalent TatP binding was negligible. The requirement of the cell-surface heparan sulfate (HS) chains of HS proteoglycans (HSPGs) for TatP binding and intracellular transport was demonstrated by the enzymatic removal of HS and simultaneous observation of two individual particles. Multivalent TatP induces HSPG cross-linking, recruiting activated Rac1 to adjacent lipid rafts and thereby enhancing the recruitment of TatP/HSPG to actin-associated microdomains and its internalization by macropinocytosis. These findings clarify the initial binding mechanism of TatP to the cell surface and demonstrate the importance of TatP valence for strong surface binding and signal transduction. Our data also shed light on the ability of TatP to exploit the machinery of living cells, using HSPG signaling to activate Rac1 and alter TatP mobility and internalization. This work should guide the future design of TatP-based peptides as therapeutic nanocarriers with efficient transduction.


Asunto(s)
VIH-1/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Microdominios de Membrana/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , VIH-1/genética , Células HeLa , Proteoglicanos de Heparán Sulfato/genética , Humanos , Cinética , Microdominios de Membrana/genética , Pinocitosis/genética , Estructura Terciaria de Proteína , Transporte de Proteínas/genética , Proteína de Unión al GTP rac1/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
11.
Elife ; 112022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35749159

RESUMEN

Bending of cilia and flagella occurs when axonemal dynein molecules on one side of the axoneme produce force and move toward the microtubule (MT) minus end. These dyneins are then pulled back when the axoneme bends in the other direction, meaning oscillatory back and forth movement of dynein during repetitive bending of cilia/flagella. There are various factors that may regulate the dynein activity, e.g. the nexin-dynein regulatory complex, radial spokes, and central apparatus. In order to understand the basic mechanism of dynein's oscillatory movement, we constructed a simple model system composed of MTs, outer-arm dyneins, and crosslinks between the MTs made of DNA origami. Electron microscopy (EM) showed pairs of parallel MTs crossbridged by patches of regularly arranged dynein molecules bound in two different orientations, depending on which of the MTs their tails bind to. The oppositely oriented dyneins are expected to produce opposing forces when the pair of MTs have the same polarity. Optical trapping experiments showed that the dynein-MT-DNA-origami complex actually oscillates back and forth after photolysis of caged ATP. Intriguingly, the complex, when held at one end, showed repetitive bending motions. The results show that a simple system composed of ensembles of oppositely oriented dyneins, MTs, and inter-MT crosslinkers, without any additional regulatory structures, has an intrinsic ability to cause oscillation and repetitive bending motions.


Asunto(s)
Chlamydomonas reinhardtii , Dineínas , Dineínas Axonemales/metabolismo , Axonema/metabolismo , Chlamydomonas reinhardtii/metabolismo , ADN/metabolismo , Dineínas/metabolismo , Flagelos/fisiología , Microtúbulos/metabolismo , Movimiento/fisiología
12.
J Biol Chem ; 285(25): 19605-14, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20392702

RESUMEN

Filopodia are actin-rich finger-like cytoplasmic projections extending from the leading edge of cells. Unconventional myosin-X is involved in the protrusion of filopodia. However, the underlying mechanism of myosin-X-induced filopodia formation is obscure. Here, we studied the movements of myosin-X during filopodia protrusion using a total internal reflection microscope to clarify the mechanism of myosin-X-induced filopodia formation. Myosin-X was recruited to the discrete site at the leading edge where it assembles with exponential kinetics before the filopodia extension. The myosin-X-induced filopodia showed repeated extension-retraction cycles with each extension of 2.4 microm, which was critical to produce long filopodia. Myosin-X, lacking the FERM domain, could move to the tip as does the wild type. However, it was transported toward the cell body during filopodia retraction, did not undergo multiple extension-retraction cycles, and failed to produce long filopodia. During the filopodia protrusion, the single molecules of full-length myosin-X moved within filopodia. The majority of the fluorescence spots showed two-step photobleaching, suggesting that the moving myosin-X is a dimer. Deletion of the FERM domain did not change the movement at the single molecule level with the same velocity of approximately 600 nm/s as wild-type, suggesting that the myosin-X in filopodia moves without interaction with the attached membrane via the FERM domain. Based upon these results, we have proposed a model of myosin-X-induced filopodia protrusion.


Asunto(s)
Miosinas/metabolismo , Seudópodos/metabolismo , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , Cinética , Luz , Microscopía Fluorescente/métodos , Modelos Biológicos , Estructura Terciaria de Proteína , Factores de Tiempo
13.
J Biol Chem ; 285(4): 2750-7, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19917603

RESUMEN

Changes in membrane morphology and membrane protein dynamics based on its fluidity are critical for cancer metastasis. However, this subject has remained unclear, because the spatial precision of previous in vivo imaging has been limited to the micrometer level and single molecule imaging is impossible. Here, we have imaged the membrane dynamics of tumor cells in mice with a spatial precision of 7-9 nm under a confocal microscope. A metastasis-promoting factor on the cell membrane, protease-activated receptor 1 (PAR1), was labeled with quantum dots conjugated with an anti-PAR1 antibody. Movements of cancer cells and PAR1 during metastasis were clearly observed in vivo. Images used to assess PAR1 dynamics were taken of representative cells for four stages of metastasis; i.e. cancer cells far from blood vessels in tumor, near the vessel, in the bloodstream, and adherent to the inner vascular surface in the normal tissues near tumor were photographed. The diffusion constant of PAR1 in static cells far from tumor blood vessels was smaller than in moving cells near the vessels and in the bloodstream. The diffusion constant of cells adhering to the inner vascular surface in the normal tissues was also very small. Cells formed membrane protrusion during migration. The PAR1 diffusion constant on these pseudopodia was greater than in other membrane regions in the same cell. Thus, the dynamics of PAR1 movement showed that membrane fluidity increases during intravasation, reaches a peak in the vessel, decreases during extravasation, and is also higher at locally formed pseudopodia.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/secundario , Membrana Celular/metabolismo , Fluidez de la Membrana/fisiología , Puntos Cuánticos , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Difusión , Mapeo Epitopo , Femenino , Humanos , Ratones , Ratones SCID , Microscopía Confocal , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Seudópodos/metabolismo , Receptor PAR-1/química , Receptor PAR-1/metabolismo
14.
Curr Opin Cell Biol ; 14(1): 50-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11792544

RESUMEN

Analysis of a mutant with altered directionality has led to new insights into motor directionality. The prediction from current models for processivity of a two-heads-bound state has been confirmed by electron microscopy for myosin V and by unbinding experiments for kinesin. Evidence is emerging that non-processive motors bind their filament with one head, hydrolyze ATP and then release, requiring binding by a second motor to complete a step.


Asunto(s)
Proteínas Motoras Moleculares/metabolismo , Animales , Citoesqueleto/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos , Modelos Moleculares , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/genética , Mutación , Miosinas/química , Miosinas/metabolismo
15.
Nat Cell Biol ; 4(10): 790-7, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12360289

RESUMEN

The molecular motor kinesin travels processively along a microtubule in a stepwise manner. Here we have studied the chemomechanical coupling of the hydrolysis of ATP to the mechanical work of kinesin by analysing the individual stepwise movements according to the directionality of the movements. Kinesin molecules move primarily in the forward direction and only occasionally in the backward direction. The hydrolysis of a single ATP molecule is coupled to either the forward or the backward movement. This bidirectional movement is well described by a model of Brownian motion assuming an asymmetric potential of activation energy. Thus, the stepwise movement along the microtubule is most probably due to Brownian motion that is biased towards the forward direction by chemical energy stored in ATP molecules.


Asunto(s)
Adenosina Trifosfato/metabolismo , Transporte Biológico/fisiología , Células Eucariotas/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Proteínas Motoras Moleculares/metabolismo , Adenosina Trifosfato/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Bovinos , Relación Dosis-Respuesta a Droga , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Cinesinas/efectos de los fármacos , Cinética , Modelos Biológicos , Proteínas Motoras Moleculares/efectos de los fármacos , Factores de Tiempo
16.
Nat Cell Biol ; 5(12): 1079-82, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14634664

RESUMEN

A conventional kinesin molecule travels continuously along a microtubule in discrete 8-nm steps. This processive movement is generally explained by models in which the two identical heads of a kinesin move in a 'hand-over-hand' manner. Here, we show that a single heterodimeric kinesin molecule (in which one of the two heads is mutated in a nucleotide-binding site) exhibits fast and slow (with the dwell time at least 10 times longer than that of the fast step) 8-nm steps alternately, presumably corresponding to the displacement by the wild-type and mutant heads, respectively. Our results provide the first direct evidence for models in which the roles of the two heads alternate every 8-nm step.


Asunto(s)
Cinesinas/química , Microtúbulos/fisiología , Proteínas Motoras Moleculares/química , Dimerización , Cinesinas/fisiología , Modelos Moleculares , Conformación Molecular , Proteínas Motoras Moleculares/fisiología , Mutación/fisiología , Transporte de Proteínas/fisiología , Factores de Tiempo
17.
Nat Struct Mol Biol ; 12(2): 127-32, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15665867

RESUMEN

Myosin V is a calmodulin-binding motor protein. The dissociation of single calmodulin molecules from individual myosin V molecules at 1 microM Ca(2+) correlates with a reduction in sliding velocity in an in vitro motility assay. The dissociation of two calmodulin molecules at 5 microM Ca(2+) correlates with a detachment of actin filaments from myosin V. To mimic the regulation of myosin V motility by Ca(2+) in a cell, caged Ca(2+) coupled with a UV flash system was used to produce Ca(2+) transients. During the Ca(2+) transient, myosin V goes through the functional cycle of reduced sliding velocity, actin detachment and reattachment followed by the recovery of the sliding velocity. These results indicate that myosin V motility is regulated by Ca(2+) through a reduction in actin-binding affinity resulting from the dissociation of single calmodulin molecules.


Asunto(s)
Calmodulina/metabolismo , Miosina Tipo V/metabolismo , Animales , Calcio/farmacología , Bovinos , Pollos , Movimiento/efectos de los fármacos , Unión Proteica
18.
Nature ; 424(6948): 574-7, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12891363

RESUMEN

Conventional isoforms of the motor protein kinesin behave functionally not as 'single molecules' but as 'two molecules' paired. This dimeric structure poses a barrier to solving its mechanism. To overcome this problem, we used an unconventional kinesin KIF1A (refs 5, 6) as a model molecule. KIF1A moves processively as an independent monomer, and can also work synergistically as a functional dimer. Here we show, by measuring its movement with an optical trapping system, that a single ATP hydrolysis triggers a single stepping movement of a single KIF1A monomer. The step size is distributed stochastically around multiples of 8 nm with a gaussian-like envelope and a standard deviation of 15 nm. On average, the step is directional to the microtubule's plus-end against a load force of up to 0.15 pN. As the source for this directional movement, we show that KIF1A moves to the microtubule's plus-end by approximately 3 nm on average on binding to the microtubule, presumably by preferential binding to tubulin on the plus-end side. We propose a simple physical formulation to explain the movement of KIF1A.


Asunto(s)
Cinesinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Tubulina (Proteína)/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Hidrólisis , Microtúbulos/química , Microtúbulos/metabolismo , Movimiento , Unión Proteica , Distribuciones Estadísticas , Procesos Estocásticos
19.
Sci Rep ; 10(1): 20468, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33235297

RESUMEN

The heart rhythm is maintained by oscillatory changes in [Ca2+]. However, it has been suggested that the rapid drop in blood pressure that occurs with a slow decrease in [Ca2+] preceding early diastolic filling is related to the mechanism of rapid sarcomere lengthening associated with spontaneous tension oscillation at constant intermediate [Ca2+]. Here, we analyzed a new type of oscillation called hyperthermal sarcomeric oscillation. Sarcomeres in rat neonatal cardiomyocytes that were warmed at 38-42 °C oscillated at both slow (~ 1.4 Hz), Ca2+-dependent frequencies and fast (~ 7 Hz), Ca2+-independent frequencies. Our high-precision experimental observations revealed that the fast sarcomeric oscillation had high and low peak-to-peak amplitude at low and high [Ca2+], respectively; nevertheless, the oscillation period remained constant. Our numerical simulations suggest that the regular and fast rthythm is maintained by the unchanged cooperative binding behavior of myosin molecules during slow oscillatory changes in [Ca2+].


Asunto(s)
Calcio/metabolismo , Miocitos Cardíacos/fisiología , Sarcómeros/fisiología , Animales , Animales Recién Nacidos , Homeostasis , Calor , Contracción Miocárdica , Miosinas/metabolismo , Ratas , Análisis Espacio-Temporal
20.
Transl Oncol ; 13(6): 100764, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32403030

RESUMEN

Anticancer drug efficacy varies because the delivery of drugs within tumors and tumor responses are heterogeneous; however, these features are often more homogenous in vitro. This difference makes it difficult to accurately determine drug efficacy. Therefore, it is important to use living tumor tissues in preclinical trials to observe the heterogeneity in drug distribution and cell characteristics in tumors. In the present study, to accurately evaluate the efficacy of an antibody-drug conjugate (ADC) containing a microtubule inhibitor, we established a cell line that expresses a fusion of end-binding protein 1 and enhanced green fluorescent protein that serves as a microtubule plus-end-tracking protein allowing the visualization of microtubule dynamics. This cell line was xenografted into mice to create a model of living tumor tissue. The tumor cells possessed a greater number of microtubules with plus-ends, a greater number of meandering microtubules, and a slower rate of microtubule polymerization than the in vitro cells. In tumor tissues treated with fluorescent dye-labeled ADCs, heterogeneity was observed in the delivery of the drug to tumor cells, and microtubule dynamics were inhibited in a concentration-dependent manner. Moreover, a difference in drug sensitivity was observed between in vitro cells and tumor cells; compared with in vitro cells, tumor cells were more sensitive to changes in the concentration of the ADC. This study is the first to simultaneously evaluate the delivery and intracellular efficacy of ADCs in living tumor tissue. Accurate evaluation of the efficacy of ADCs is important for the development of effective anticancer drugs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA