Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(26): e2115190119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35737845

RESUMEN

Hearing depends on intricate morphologies and mechanical properties of diverse inner ear cell types. The individual contributions of various inner ear cell types into mechanical properties of the organ of Corti and the mechanisms of their integration are yet largely unknown. Using sub-100-nm spatial resolution atomic force microscopy (AFM), we mapped the Young's modulus (stiffness) of the apical surface of the different cells of the freshly dissected P5-P6 cochlear epithelium from wild-type and mice lacking either Trio and F-actin binding protein (TRIOBP) isoforms 4 and 5 or isoform 5 only. Variants of TRIOBP are associated with deafness in human and in Triobp mutant mouse models. Remarkably, nanoscale AFM mapping revealed unrecognized bidirectional radial stiffness gradients of different magnitudes and opposite orientations between rows of wild-type supporting cells and sensory hair cells. Moreover, the observed bidirectional radial stiffness gradients are unbalanced, with sensory cells being stiffer overall compared to neighboring supporting cells. Deafness-associated TRIOBP deficiencies significantly disrupted the magnitude and orientation of these bidirectional radial stiffness gradients. In addition, serial sectioning with focused ion beam and backscatter scanning electron microscopy shows that a TRIOBP deficiency results in ultrastructural changes of supporting cell apical phalangeal microfilaments and bundled cortical F-actin of hair cell cuticular plates, correlating with messenger RNA and protein expression levels and AFM stiffness measurements that exposed a softening of the apical surface of the sensory epithelium in mutant mice. Altogether, this additional complexity in the mechanical properties of the sensory epithelium is hypothesized to be an essential contributor to frequency selectivity and sensitivity of mammalian hearing.


Asunto(s)
Citoesqueleto de Actina , Sordera , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Cóclea/metabolismo , Sordera/metabolismo , Células Ciliadas Auditivas/metabolismo , Mamíferos/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Órgano Espiral , Isoformas de Proteínas/metabolismo
2.
Nanotechnology ; 26(35): 354004, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26266760

RESUMEN

The existing approach to characterize cell biomechanical properties typically utilizes switch-like models of mechanotransduction in which cell responses are analyzed in response to a single nanomechanical indentation or a transient pulsed stress. Although this approach provides effective descriptors at population-level, at a single-cell-level, there are significant overlaps in the biomechanical descriptors of non-metastatic and metastatic cells which precludes the use of biomechanical markers for single cell metastatic phenotyping. This study presents a new promising marker for biosensing metastatic and non-metastatic cells at a single-cell-level using the effects of a dynamic microenvironment on the biomechanical properties of cells. Two non-metastatic and two metastatic epithelial breast cell lines are subjected to a pulsed stresses regimen exerted by atomic force microscopy. The force-time data obtained for the cells revealed that the non-metastatic cells increase their resistance against deformation and become more stiffened when subjected to a series of nanomechanical indentations. On the other hand, metastatic cells become slightly softened when their mechanical microenvironment is subjected to a similar dynamical changes. This distinct behavior of the non-metastatic and metastatic cells to the pulsed stresses paradigm provided a signature for single-cell-level metastatic phenotyping with a high confidence level of ∼95%.

3.
Nanomedicine ; 10(5): 1013-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24407147

RESUMEN

We herein report, for the first time, the mechanical properties of ovarian cancer stem-like/tumor-initiating cells (CSC/TICs). The represented model is a spontaneously transformed murine ovarian surface epithelial (MOSE) cell line that mimics the progression of ovarian cancer from early/non-tumorigenic to late/highly aggressive cancer stages. Elastic modulus measurements via atomic force microscopy (AFM) illustrate that the enriched CSC/TICs population (0.32±0.12kPa) are 46%, 61%, and 72% softer (P<0.0001) than their aggressive late-stage, intermediate, and non-malignant early-stage cancer cells, respectively. Exposure to sphingosine, an anti-cancer agent, induced an increase in the elastic moduli of CSC/TICs by more than 46% (0.47±0.14kPa, P<0.0001). Altogether, our data demonstrate that the elastic modulus profile of CSC/TICs is unique and responsive to anti-cancer treatment strategies that impact the cytoskeleton architecture of cells. These findings increase the chance for obtaining distinctive cell biomechanical profiles with the intent of providing a means for effective cancer detection and treatment control. FROM THE CLINICAL EDITOR: This novel study utilized atomic force microscopy to demonstrate that the elastic modulus profile of cancer stem cell-like tumor initiating cells is unique and responsive to anti-cancer treatment strategies that impact the cytoskeleton of these cells. These findings pave the way to the development of unique means for effective cancer detection and treatment control.


Asunto(s)
Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/patología , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Femenino , Ratones , Microscopía de Fuerza Atómica
4.
Biofabrication ; 13(4)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34233304

RESUMEN

Selective spatial isolation and manipulation of single chromosomes and the controlled formation of defined chromosome ensembles in a droplet-based microfluidic system is presented. The multifunctional microfluidic technology employs elastomer valves and membrane displacement traps to support deterministic manipulation of individual droplets. Picoliter droplets are formed in the 2D array of microscale traps by self-discretization of a nanoliter sample plug, with membranes positioned over each trap allowing controllable metering or full release of selected droplets. By combining discretization, optical interrogation, and selective droplet release for sequential delivery to a downstream merging zone, the system enables efficient manipulation of multiple chromosomes into a defined ensemble with single macromolecule resolution. Key design and operational parameters are explored, and co-compartmentalization of three chromosome pairs is demonstrated as a first step toward formation of precisely defined chromosome ensembles for applications in genetic engineering and synthetic biology.


Asunto(s)
Cromosomas , Técnicas Analíticas Microfluídicas , Microfluídica
5.
Biomicrofluidics ; 14(1): 014112, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32038741

RESUMEN

A programmable microfluidic platform enabling on-demand sampling, compartmentalization, and manipulation of multiple aqueous volumes is presented. The system provides random-access actuation of a microtrap array supporting selective discretization of picoliter volumes from multiple sample inputs. The platform comprises two interconnected chips, with parallel T-junctions and multiplexed microvalves within one chip enabling programmable injection of aqueous sample plugs, and nanoliter volumes transferred to a second microtrap array chip in which the plugs are actively discretized into picoliter droplets within a static array of membrane displacement actuators. The system employs two different multiplexer designs that reduce the number of input signals required for both sample injection and discretization. This versatile droplet-based technology offers flexible sample workflows and functionalities for the formation and manipulation of heterogeneous picoliter droplets, with particular utility for applications in biochemical synthesis and cell-based assays requiring flexible and programmable operation of parallel and multistep droplet processes. The platform is used here for the selective encapsulation of differentially labeled cells within a discrete droplet array.

6.
Lab Chip ; 19(3): 493-502, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30623951

RESUMEN

A multifunctional microfluidic platform combining on-demand aqueous-phase droplet generation, multi-droplet storage, and controlled merging of droplets selected from a storage library in a single integrated microfluidic device is described. A unique aspect of the technology is a microfluidic trap design comprising a droplet trap chamber and lateral bypass channels integrated with a microvalve that supports the capture and merger of multiple droplets over a wide range of individual droplet sizes. A storage unit comprising an array of microfluidic traps operates in a first-in first-out manner, allowing droplets stored within the library to be analyzed before sequentially delivering selected droplets to a downstream merging zone, while shunting other droplets to waste. Performance of the microfluidic trap is investigated for variations in bypass/chamber hydrodynamic resistance ratio, micro-chamber geometry, trapped droplet volume, and overall flow rate. The integrated microfluidic platform is then utilized to demonstrate the operational steps necessary for cell-based assays requiring the isolation of defined cell populations with single cell resolution, including encapsulation of individual cells within an aqueous-phase droplet carrier, screening or incubation of the immobilized cell-encapsulated droplets, and generation of controlled combinations of individual cells through the sequential droplet merging process. Beyond its utility for cell analysis, the presented platform represents a versatile approach to robust droplet generation, storage, and merging for use in a wide range of droplet-based microfluidics applications.


Asunto(s)
Dispositivos Laboratorio en un Chip , Análisis de la Célula Individual/instrumentación , Comunicación Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Fibroblastos/citología , Humanos , Transducción de Señal
7.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 5350-5353, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30441545

RESUMEN

A microfluidic valve-based trap enabling controlled capture, release, and temporary immobilization of droplets together with on-demand merging of selected droplets is presented in this paper. The microfluidic trap technology can merge droplets passively or in active manner via a pneumatically actuated membrane. A microchip is developed with two functional units of droplet generator and merging mechanism to implement the passive or active merging performance of the microfluidic valve-based trap using a low and high surfactant concentrated continuous oil-phase.


Asunto(s)
Técnicas Analíticas Microfluídicas , Microfluídica
8.
Anal Methods ; 9(5): 847-855, 2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-29034007

RESUMEN

This study describes the development of a microfluidic biosensor called the iterative mechanical characteristics (iMECH) analyzer which enables label-free biomechanical profiling of individual cells for distinction between metastatic and non-metastatic human mammary cell lines. Previous results have demonstrated that pulsed mechanical nanoindentation can modulate the biomechanics of cells resulting in distinctly different biomechanical responses in metastatic and non-metastatic cell lines. The iMECH analyzer aims to move this concept into a microfluidic, clinically more relevant platform. The iMECH analyzer directs a cyclic deformation regimen by pulling cells through a test channel comprised of narrow deformation channels and interspersed with wider relaxation regions which together simulate a dynamic microenvironment. The results of the iMECH analysis of human breast cell lines revealed that cyclic deformations produce a resistance in non-metastatic 184A1 and MCF10A cells as determined by a drop in their average velocity in the iterative deformation channels after each relaxation. In contrast, metastatic MDA-MB-231 and MDA-MB-468 cells exhibit a loss of resistance as measured by a velocity raise after each relaxation. These distinctive modulatory mechanical responses of normal-like non-metastatic and metastatic cancer breast cells to the pulsed indentations paradigm provide a unique bio-signature. The iMECH analyzer represents a diagnostic microchip advance for discriminating metastatic cancer at the single-cell level.

9.
ACS Sens ; 2(2): 290-299, 2017 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-28723132

RESUMEN

A microfluidic device composed of variable numbers of multiconstriction channels is reported in this paper to differentiate a human breast cancer cell line, MDA-MB-231, and a nontumorigenic human breast cell line, MCF-10A. Differences between their mechanical properties were assessed by comparing the effect of single or multiple relaxations on their velocity profiles which is a novel measure of their deformation ability. Videos of the cells were recorded via a microscope using a smartphone, and imported to a tracking software to gain the position information on the cells. Our results indicated that a multiconstriction channel design with five deformation (50 µm in length, 10 µm in width, and 8 µm in height) separated by four relaxation (50 µm in length, 40 µm in width, and 30 µm in height) regions was superior to a single deformation design in differentiating MDA-MB-231 and MCF-10A cells. Velocity profile criteria can achieve a differentiation accuracy around 95% for both MDA-MB-231 and MCF-10A cells.

10.
Lab Chip ; 16(1): 188-98, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26607223

RESUMEN

Cancer progression and physiological changes within the cells are accompanied by alterations in the biophysical properties. Therefore, the cell biophysical properties can serve as promising markers for cancer detection and physiological activities. To aid in the investigation of the biophysical markers of cells, a microfluidic chip has been developed which consists of a constriction channel and embedded microelectrodes. Single-cell impedance magnitudes at four frequencies and entry and travel times are measured simultaneously during their transit through the constriction channel. This microchip provides a high-throughput, label-free, automated assay to identify biophysical signatures of malignant cells and monitor the therapeutic efficacy of drugs. Here, we monitored the dynamic cellular biophysical properties in response to sphingosine kinase inhibitors (SphKIs), and compared the effectiveness of drug delivery using poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) loaded with SphKIs versus conventional delivery. Cells treated with SphKIs showed significantly higher impedance magnitudes at all four frequencies. The bioelectrical parameters extracted using a model also revealed that the highly aggressive breast cells treated with SphKIs shifted electrically towards that of a less malignant phenotype; SphKI-treated cells exhibited an increase in cell-channel interface resistance and a significant decrease in specific membrane capacitance. Furthermore, SphKI-treated cells became slightly more deformable as measured by a decrease in their channel entry and travel times. We observed no significant difference in the bioelectrical changes produced by SphKI delivered conventionally or with NPs. However, NPs-packaged delivery of SphKI decreased the cell deformability. In summary, this study showed that while the bioelectrical properties of the cells were dominantly affected by SphKIs, the biomechanical properties were mainly changed by the NPs.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Capacidad Eléctrica , Nanopartículas/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Análisis de la Célula Individual/métodos , Antineoplásicos/química , Antineoplásicos/farmacología , Fenómenos Biomecánicos/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ácido Láctico/química , Técnicas Analíticas Microfluídicas/instrumentación , Estructura Molecular , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
11.
Integr Biol (Camb) ; 5(11): 1385-92, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24056950

RESUMEN

Cancer progression is associated with an increased deformability of cancer cells and reduced resistance to mechanical forces, enabling motility and invasion. This is important for metastases survival and outgrowth and as such could be a target for chemopreventive strategies. In this study, we determined the differential effects of exogenous sphingolipid metabolites on the elastic modulus of mouse ovarian surface epithelial cells as they transition to cancer. Treatment with ceramide or sphingosine-1-phosphate in non-toxic concentrations decreased the average elastic modulus by 21% (p≤ 0.001) in transitional and 15% (p≤ 0.02) in aggressive stages while exerting no appreciable effect on non-malignant cells. In contrast, sphingosine treatment on average increased the elastic modulus by 33% (p≤ 0.0002) in aggressive cells while not affecting precursor cells. These results indicate that tumor-supporting sphingolipid metabolites act by making cells softer, while the anti-cancer metabolite sphingosine partially reverses the decreased elasticity associated with cancer progression. Thus, sphingosine may be a valid alternative to conventional chemotherapeutics in ovarian cancer prevention or treatment.


Asunto(s)
Neoplasias Ováricas/patología , Esfingolípidos/metabolismo , Animales , Fenómenos Biomecánicos , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Ceramidas/metabolismo , Progresión de la Enfermedad , Módulo de Elasticidad , Femenino , Lisofosfolípidos/metabolismo , Ratones , Microscopía de Fuerza Atómica , Metástasis de la Neoplasia , Neoplasias Ováricas/metabolismo , Programas Informáticos , Esfingosina/análogos & derivados , Esfingosina/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-23366417

RESUMEN

Bioactive sphingolipid metabolites have emerged as important lipid second messengers in the regulation of cell growth, death, motility and many other events. These processes are important in cancer development and progression; thus, sphingolipid metabolites have been implicated in both cancer development and cancer prevention. Despite recent considerable progress in understanding the multi-faceted functions of these bioactive metabolites, little is known about their influence on the biomechanical property of cells. The biomechanical properties of cancer cells change during progression with aggressive and invasive cells being softer compared to their benign counterparts. In this paper, we investigated the effects of exogenous sphingolipid metabolites on the Young's modulus and cytoskeletal organization of cells representing aggressive ovarian cancer. Our findings demonstrate that the elasticity of aggressive ovarian cancer cells decreased ∼15% after treatment with ceramide and sphingosine-1-phosphate. In contrast, sphingosine treatment caused a ∼30% increase in the average elasticity which was associated with a more defined actin cytoskeleton organization. This indicates that sphingolipid metabolites differentially modulate the biomechanic properties of cancer cells which may have a critical impact on cancer cell survival and progression, and the use of sphingolipid metabolites as chemopreventive or chemo-therapeutic agents.


Asunto(s)
Neoplasias Ováricas/metabolismo , Esfingolípidos/metabolismo , Animales , Fenómenos Biomecánicos , Diferenciación Celular/fisiología , Ceramidas/metabolismo , Femenino , Humanos , Lisofosfolípidos/metabolismo , Ratones , Microscopía de Fuerza Atómica , Modelos Estadísticos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda