Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 162(2): 243-245, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26186185

RESUMEN

Small molecule inhibitors of microtubule dynamics are widely used as cell biology research tools and clinically as cancer chemotherapeutics. By slight modification to the chemical structure of a known microtubule inhibitor, combretastatin A-4, Borowiak et al. develop a photoswitchable derivative that can be turned "on" and "off" with low-intensity light to spatially and temporally control microtubule dynamics.


Asunto(s)
Antimitóticos/química , Muerte Celular , Microtúbulos/efectos de los fármacos , Mitosis , Estilbenos/química , Animales , Humanos
2.
Biophys J ; 117(7): 1234-1249, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31493861

RESUMEN

Microtubules are multistranded polymers in eukaryotic cells that support key cellular functions such as chromosome segregation, motor-based cargo transport, and maintenance of cell polarity. Microtubules self-assemble via "dynamic instability," in which the dynamic plus ends switch stochastically between alternating phases of polymerization and depolymerization. A key question in the field is what are the atomistic origins of this switching, i.e., what is different between the GTP- and GDP-tubulin states that enables microtubule growth and shortening, respectively? More generally, a major challenge in biology is how to connect theoretical frameworks across length- and timescales, from atoms to cellular behavior. In this study, we describe a multiscale model by linking atomistic molecular dynamics (MD), molecular Brownian dynamics (BD), and cellular-level thermokinetic modeling of microtubules. Here, we investigated the underlying interaction energy when tubulin dimers associate laterally by performing all-atom MD simulations. We found that the lateral potential energy is not significantly different among three nucleotide states of tubulin, GTP, GDP, and GMPCPP and is estimated to be ≅ -11 kBT. Furthermore, using MD potential energy in our BD simulations of tubulin dimers confirms that the lateral bond is weak on its own, with a mean lifetime of ∼0.1 µs, implying that the longitudinal bond is required for microtubule assembly. We conclude that nucleotide-dependent lateral-bond strength is not the key mediator microtubule dynamic instability, implying that GTP acts elsewhere to exert its stabilizing influence on microtubule polymer. Furthermore, the estimated lateral-bond strength (ΔGlat0≅ -5 kBT) is well-aligned with earlier estimates based on thermokinetic modeling and light microscopy measurements. Thus, we have computationally connected atomistic-level structural information, obtained by cryo-electron microscopy, to cellular-scale microtubule assembly dynamics using a combination of MD, BD, and thermokinetic models to bridge from Ångstroms to micrometers and from femtoseconds to minutes.


Asunto(s)
Simulación de Dinámica Molecular , Tubulina (Proteína)/metabolismo , Cinética , Unión Proteica , Conformación Proteica , Termodinámica , Tubulina (Proteína)/química
3.
Mol Pharm ; 15(9): 3892-3900, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30048137

RESUMEN

Multidrug resistance and toxic side effects are the major challenges in cancer treatment with microtubule-targeting agents (MTAs), and thus, there is an urgent clinical need for new therapies. Chalcone, a common simple scaffold found in many natural products, is widely used as a privileged structure in medicinal chemistry. We have previously validated tubulin as the anticancer target for chalcone derivatives. In this study, an α-methyl-substituted indole-chalcone (FC77) was synthesized and found to exhibit an excellent cytotoxicity against the NCI-60 cell lines (average concentration causing 50% growth inhibition = 6 nM). More importantly, several multidrug-resistant cancer cell lines showed no resistance to FC77, and the compound demonstrated good selective toxicity against cancer cells versus normal CD34+ blood progenitor cells. A further mechanistic study demonstrated that FC77 could arrest cells that relate to the binding to tubulin and inhibit the microtubule dynamics. The National Cancer Institute COMPARE analysis and molecular modeling indicated that FC77 had a mechanism of action similar to that of colchicine. Overall, our data demonstrate that this indole-chalcone represents a novel MTA template for further development of potential drug candidates for the treatment of multidrug-resistant cancers.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Chalconas/química , Indoles/química , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Antineoplásicos/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Relación Estructura-Actividad
4.
Biophys J ; 105(11): 2528-40, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24314083

RESUMEN

The structure and free energy of multistranded linear polymer ends evolves as individual subunits are added and lost. Thus, the energetic state of the polymer end is not constant, as assembly theory has assumed. Here we utilize a Brownian dynamics approach to simulate the addition and loss of individual subunits at the polymer tip. Using the microtubule as a primary example, we examined how the structure of the polymer tip dictates the rate at which units are added to and lost from individual protofilaments. We find that freely diffusing subunits arrive less frequently to lagging protofilaments but bind more efficiently, such that there is no kinetic difference between leading and lagging protofilaments within a tapered tip. However, local structure at the nanoscale has up to an order-of-magnitude effect on the rate of addition. Thus, the kinetic on-rate constant, integrated across the microtubule tip (kon,MT), is an ensemble average of the varying individual protofilament on-rate constants (kon,PF). Our findings have implications for both catastrophe and rescue of the dynamic microtubule end, and provide a subnanoscale framework for understanding the mechanism of action of microtubule-associated proteins and microtubule-directed drugs. Although we utilize the specific example of the microtubule here, the findings are applicable to multistranded polymers generally.


Asunto(s)
Microtúbulos/química , Modelos Biológicos , Animales , Difusión , Humanos , Cinética , Microtúbulos/metabolismo , Movimiento (Física) , Multimerización de Proteína , Termodinámica
5.
iScience ; 23(12): 101782, 2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-33294790

RESUMEN

Plus-end tracking proteins (+TIPs) associate with the growing end of microtubules and mediate important cellular functions. The majority of +TIPs are directed to the plus-end through a family of end-binding proteins (EBs), which preferentially bind the stabilizing cap of GTP-tubulin present during microtubule growth. One outstanding question is whether there may exist other microtubule-associated proteins (MAPs) that preferentially bind specific nucleotide states of tubulin. Here, we report that the neuronal MAP tau preferentially binds GDP-tubulin (K D = 0.26 µM) over GMPCPP-tubulin (K D = 1.1 µM) in vitro, as well as GTP-tubulin at the tips of growing microtubules, causing tau binding to lag behind the plus-end both in vitro and in live cells. Thus, tau is a microtubule tip avoiding protein, establishing the framework for a possible new class of tip avoiding MAPs. We speculate that disease-relevant tau mutations may exert their phenotype by their failure to properly recognize GDP-tubulin.

6.
Sci Adv ; 5(3): eaau1086, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30891490

RESUMEN

In sickle cell disease, the aberrant assembly of hemoglobin fibers induces changes in red blood cell morphology and stiffness, which leads to downstream symptoms of the disease. Therefore, understanding of this assembly process will be important for the treatment of sickle cell disease. By performing the highest spatiotemporal resolution measurements (55 nm at 1 Hz) of single sickle hemoglobin fiber assembly to date and combining them with a model that accounts for the multistranded structure of the fibers, we show that the rates of sickle hemoglobin addition and loss have been underestimated in the literature by at least an order of magnitude. These results reveal that the sickle hemoglobin self-assembly process is very rapid and inefficient (4% efficient versus 96% efficient based on previous analyses), where net growth is the small difference between over a million addition-loss events occurring every second.


Asunto(s)
Hemoglobina Falciforme/ultraestructura , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Microscopía de Interferencia/métodos , Anemia de Células Falciformes/sangre , Tampones (Química) , Eritrocitos/química , Hemoglobina Falciforme/química , Humanos , Soluciones , Sulfitos/química
7.
Cytoskeleton (Hoboken) ; 76(11-12): 571-585, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31512404

RESUMEN

Cell migration and traction are essential to many biological phenomena, and one of their key features is sensitivity to substrate stiffness, which biophysical models, such as the motor-clutch model and the cell migration simulator can predict and explain. However, these models have not accounted for the finite size of adhesions, the spatial distribution of forces within adhesions. Here, we derive an expression that relates varying adhesion radius ( R) and spatial distribution of force within an adhesion (described by s) to the effective substrate stiffness ( κsub ), as a function of the Young's modulus of the substrate ( E Y ), which yields the relation, κsub=RsEY , for two-dimensional cell cultures. Experimentally, we found that a cone-shaped force distribution ( s = 1.05) can describe the observed displacements of hydrogels deformed by adherent U251 glioma cells. Also, we found that the experimentally observed adhesion radius increases linearly with the cell protrusion force, consistent with the predictions of the motor-clutch model with spatially distributed clutches. We also found that, theoretically, the influence of one protrusion on another through a continuous elastic environment is negligible. Overall, we conclude cells can potentially control their own interpretation of the mechanics of the environment by controlling adhesion size and spatial distribution of forces within an adhesion.


Asunto(s)
Neoplasias de la Mama/patología , Adhesión Celular , Movimiento Celular , Módulo de Elasticidad , Mecanotransducción Celular , Músculo Liso Vascular/fisiología , Células Cultivadas , Femenino , Humanos , Músculo Liso Vascular/citología
8.
Curr Opin Cell Biol ; 50: 8-13, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29351860

RESUMEN

Microtubule self-assembly dynamics serve to facilitate many vital cellular functions, such as chromosome segregation during mitosis and synaptic plasticity. However, the detailed atomistic basis of assembly dynamics has remained an unresolved puzzle. A key challenge is connecting together the vast range of relevant length-time scales, events happening at time scales ranging from nanoseconds, such as tubulin molecular interactions (Å-nm), to minutes-hours, such as the cellular response to microtubule dynamics during mitotic progression (µm). At the same time, microtubule interactions with associated proteins and binding agents, such as anti-cancer drugs, can strongly affect this dynamic process through atomic-level mechanisms that remain to be elucidated. New high-resolution technologies for investigating these interactions, including cryo-electron microscopy (EM) techniques and total internal reflection fluorescence (TIRF) microscopy, are yielding important new insights. Here, we focus on recent studies of microtubule dynamics, both theoretical and experimental, and how these findings shed new light on this complex phenomenon across length-time scales, from Å to µm and from nanoseconds to minutes.


Asunto(s)
Segregación Cromosómica , Microtúbulos/metabolismo , Microscopía por Crioelectrón , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/efectos de los fármacos , Mitosis , Neoplasias/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Termodinámica , Tubulina (Proteína)/metabolismo
9.
Mol Biol Cell ; 28(9): 1238-1257, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28298489

RESUMEN

Microtubule-targeting agents (MTAs), widely used as biological probes and chemotherapeutic drugs, bind directly to tubulin subunits and "kinetically stabilize" microtubules, suppressing the characteristic self-assembly process of dynamic instability. However, the molecular-level mechanisms of kinetic stabilization are unclear, and the fundamental thermodynamic and kinetic requirements for dynamic instability and its elimination by MTAs have yet to be defined. Here we integrate a computational model for microtubule assembly with nanometer-scale fluorescence microscopy measurements to identify the kinetic and thermodynamic basis of kinetic stabilization by the MTAs paclitaxel, an assembly promoter, and vinblastine, a disassembly promoter. We identify two distinct modes of kinetic stabilization in live cells, one that truly suppresses on-off kinetics, characteristic of vinblastine, and the other a "pseudo" kinetic stabilization, characteristic of paclitaxel, that nearly eliminates the energy difference between the GTP- and GDP-tubulin thermodynamic states. By either mechanism, the main effect of both MTAs is to effectively stabilize the microtubule against disassembly in the absence of a robust GTP cap.


Asunto(s)
Microtúbulos/metabolismo , Paclitaxel/farmacología , Vinblastina/farmacología , Animales , Simulación por Computador , Procesamiento de Imagen Asistido por Computador , Cinética , Microscopía Fluorescente/métodos , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/fisiología , Porcinos , Termodinámica , Tubulina (Proteína)/metabolismo
10.
Methods Enzymol ; 540: 35-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24630100

RESUMEN

Microtubules are dynamic polymers of the cytoskeleton, which play important roles in cell division, polarization, and intracellular transport. Self-assembly of microtubule polymer from αß-tubulin heterodimers is highly variable, with stochastic switching between alternate states of net growth and net shortening, a phenomenon known as dynamic instability. Microtubule tip structures are also variable and directly influence the kinetics of assembly and vice versa. TipTracker, a semiautomated, image processing-based tool, permits high spatial and temporal resolution measurements from fluorescence microscopy images (~10-40 nm, or 1-5 dimer lengths, at 1-10 Hz) with simultaneous tip structure estimation. We provide a walkthrough of the TipTracker code to demonstrate methods used to (1) fit the coordinates of the microtubule backbone; (2) track microtubule tip position; and (3) estimate tip structure from the spatial decay of the tip fluorescence distribution, discuss possible sources of error, and include an example protocol for nanometer-scale tip tracking in living cells. Additionally, we evaluate TipTracker's accuracy on simulated digital images and fixed microtubules to estimate accuracy under realistic imaging conditions. In summary, this chapter demonstrates the use of TipTracker in making robust, high-resolution measurements of microtubule tip dynamics and structures, facilitating quantitative investigations into nanoscale/molecular control of microtubule assembly. Although our primary focus is on microtubules, these methods are, in principle, suitable for other polymer structures, such as F-actin.


Asunto(s)
Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Algoritmos , Animales , Línea Celular , Cinética , Microscopía Fluorescente/métodos , Microtúbulos/química , Porcinos , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/ultraestructura
11.
Curr Biol ; 22(18): 1681-7, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22902755

RESUMEN

Microtubules (MTs) polymerize via net addition of GTP-tubulin subunits to the MT plus end, which subsequently hydrolyze to GDP-tubulin in the MT lattice. Relatively stable GTP-tubulin subunits create a "GTP cap" at the growing MT plus end that suppresses catastrophe. To understand MT assembly regulation, we need to understand GTP hydrolysis reaction kinetics and the GTP cap size. In vitro, the GTP cap has been estimated to be as small as one layer (13 subunits) or as large as 100-200 subunits. GTP cap size estimates in vivo have not yet been reported. Using EB1-EGFP as a marker for GTP-tubulin in epithelial cells, we find on average (1) 270 EB1 dimers bound to growing MT plus ends, and (2) a GTP cap size of ∼750 tubulin subunits. Thus, in vivo, the GTP cap is far larger than previous estimates in vitro, and ∼60-fold larger than a single layer cap. We also find that the tail of a large GTP cap promotes MT rescue and suppresses shortening. We speculate that a large GTP cap provides a locally concentrated scaffold for tip-tracking proteins and confers persistence to assembly in the face of physical barriers such as the cell cortex.


Asunto(s)
Guanosina Trifosfato/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Células Epiteliales/metabolismo
12.
Cell Mol Bioeng ; 4(1): 116-121, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21892361

RESUMEN

Morphogen gradients dictate the spatial patterning of multicellular organisms and are established via transport mechanisms. One of the best-characterized morphogens, Bicoid, acts as a polarity determinant in the Drosophila embryo through spatial-temporal control of gap gene expression. The prevailing model for establishment of the gradient has been localized anterior translation, subsequent diffusion, and spatially uniform degradation, consistent with the observed exponential anterior-posterior decay. However, a recent direct measurement of the Bicoid diffusion coefficient via fluorescence recovery after photobleaching (FRAP) resulted in a surprisingly low estimate, which challenged the prevailing model and led to more complicated active transport models. Here, we reassessed this conclusion using a detailed computational model of the FRAP experiment and analysis. In our model, we found disagreement between the input diffusion coefficient and the resulting estimated diffusion coefficient, as measured by previous methods. By using the model to reproduce the original data, we estimate that Bicoid's mitotic diffusion coefficient is 3-fold larger than the originally reported value. Thus, the long-standing diffusive transport model still holds.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA