Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmaceutics ; 15(9)2023 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-37765240

RESUMO

Overcoming the blood-brain barrier (BBB) remains a significant challenge with regard to drug delivery to the brain. By incorporating targeting ligands, and by carefully adjusting particle sizes, nanocarriers can be customized to improve drug delivery. Among these targeting ligands, transferrin stands out due to the high expression level of its receptor (i.e., transferrin receptor) on the BBB. Porous silicon nanoparticles (pSiNPs) are a promising drug nanocarrier to the brain due to their biodegradability, biocompatibility, and exceptional drug-loading capacity. However, an in-depth understanding of the optimal nanoparticle size and transferrin surface density, in order to maximize BBB penetration, is still lacking. To address this gap, a diverse library of pSiNPs was synthesized using bifunctional poly(ethylene glycol) linkers with methoxy or/and carboxyl terminal groups. These variations allowed us to explore different transferrin surface densities in addition to particle sizes. The effects of these parameters on the cellular association, uptake, and transcytosis in immortalized human brain microvascular endothelial cells (hCMEC/D3) were investigated using multiple in vitro systems of increasing degrees of complexity. These systems included the following: a 2D cell culture, a static Transwell model, and a dynamic BBB-on-a-chip model. Our results revealed the significant impact of both the ligand surface density and size of pSiNPs on their ability to penetrate the BBB, wherein intermediate-level transferrin densities and smaller pSiNPs exhibited the highest BBB transportation efficiency in vitro. Moreover, notable discrepancies emerged between the tested in vitro assays, further emphasizing the necessity of using more physiologically relevant assays, such as a microfluidic BBB-on-a-chip model, for nanocarrier testing and evaluation.

2.
Biosensors (Basel) ; 13(8)2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37622865

RESUMO

Microfluidic technology is applied across various research areas including organ-on-chip (OOC) systems. The main material used for microfluidics is polydimethylsiloxane (PDMS), a silicone elastomer material that is biocompatible, transparent, and easy to use for OOC systems with well-defined microstructures. However, PDMS-based OOC systems can absorb hydrophobic and small molecules, making it difficult and erroneous to make quantitative analytical assessments for such compounds. In this paper, we explore the use of a synthetic fluoropolymer, poly(4,5-difluoro-2,2-bis(trifluoromethyl)-1,3-dioxole-co-tetrafluoroethylene) (Teflon™ AF 2400), with excellent "non-stick" properties to functionalize OOC systems. Cannabinoids, including cannabidiol (CBD), are classes of hydrophobic compounds with a great potential for the treatment of anxiety, depression, pain, and cancer. By using CBD as a testing compound, we examined and systematically quantified CBD absorption into PDMS by means of an LC-MS/MS analysis. In comparison to the unmodified PDMS microchannels, an increase of approximately 30× in the CBD signal was detected with the fluoropolymer surface modification after 3 h of static incubation. Under perfusion conditions, we observed an increase of nearly 15× in the CBD signals from the surface-modified microchannels than from the unmodified microchannels. Furthermore, we also demonstrated that fluoropolymer-modified microchannels are compatible for culturing hCMEC/D3 endothelial cells and for CBD perfusion experiments.


Assuntos
Canabidiol , Canabinoides , Polímeros de Fluorcarboneto , Cromatografia Líquida , Células Endoteliais , Espectrometria de Massas em Tandem
3.
ACS Appl Bio Mater ; 6(9): 3532-3554, 2023 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-37294445

RESUMO

Recent preclinical and clinical studies have focused on the active area of therapeutic peptides due to their high potency, selectivity, and specificity in treating a broad range of diseases. However, therapeutic peptides suffer from multiple disadvantages, such as limited oral bioavailability, short half-life, rapid clearance from the body, and susceptibility to physiological conditions (e.g., acidic pH and enzymolysis). Therefore, high peptide dosages and dose frequencies are required for effective patient treatment. Recent innovations in pharmaceutical formulations have substantially improved therapeutic peptide administration by providing the following advantages: long-acting delivery, precise dose administration, retention of biological activity, and improvement of patient compliance. This review discusses therapeutic peptides and challenges in their delivery and explores recent peptide delivery formulations, including micro/nanoparticles (based on lipids, polymers, porous silicon, silica, and stimuli-responsive materials), (stimuli-responsive) hydrogels, particle/hydrogel composites, and (natural or synthetic) scaffolds. This review further covers the applications of these formulations for prolonged delivery and sustained release of therapeutic peptides and their impact on peptide bioactivity, loading efficiency, and (in vitro/in vivo) release parameters.


Assuntos
Hidrogéis , Peptídeos , Humanos , Polímeros
4.
Adv Biol (Weinh) ; 6(12): e2200152, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35999436

RESUMO

A continuous, sealed endothelial membrane is essential for the blood-brain barrier (BBB) to protect neurons from toxins present in systemic circulation. Endothelial cells are critical sensors of the capillary environment, where factors like fluid shear stress (FSS) and systemic signaling molecules activate intracellular pathways that either promote or disrupt the BBB. The brain vasculature exhibits complex heterogeneity across the bed, which is challenging to recapitulate in BBB microfluidic models with fixed dimensions and rectangular cross-section microchannels. Here, a Cayley-tree pattern, fabricated using lithography-less, fluid shaping technique in a modified Hele-Shaw cell is used to emulate the brain vasculature in a microfluidic chip. This geometry generates an inherent distribution of heterogeneous FSS, due to smooth variations in branch height and width. hCMEC/D3 endothelial cells cultured in the Cayley-tree designed chip generate a 3D monolayer of brain endothelium with branching hierarchy, enabling the study of the effect of heterogeneous FSS on the brain endothelium. The model is employed to study neuroinflammatory conditions by stimulating the brain endothelium with tumor necrosis factor-α under heterogeneous FSS conditions. The model has immense potential for studies involving drug transport across the BBB, which can be misrepresented in fixed dimension models.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Células Endoteliais/metabolismo , Encéfalo , Estresse Mecânico , Microfluídica
5.
Lab Chip ; 22(19): 3579-3602, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36004771

RESUMO

The blood-brain barrier (BBB) represents a key challenge in developing brain-penetrating therapeutic molecules. BBB dysfunction is also associated with the onset and progression of various brain diseases. The BBB-on-a-chip (µBBB), an organ-on-chip technology, has emerged as a powerful in vitro platform that closely mimics the human BBB microenvironments. While the µBBB technology has seen wide application in the study of brain cancer, its utility in other brain disease models ("µBBB+") is less appreciated. Based on the advances of the µBBB technology and the evolution of in vitro models for brain diseases over the last decade, we propose the concept of a "µBBB+" system and summarize its major promising applications in pathological studies, personalized medical research, drug development, and multi-organ-on-chip approaches. We believe that such a sophisticated "µBBB+" system is a highly tunable and promising in vitro platform for further advancement of the understanding of brain diseases.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Transporte Biológico , Encéfalo , Neoplasias Encefálicas/patologia , Humanos , Dispositivos Lab-On-A-Chip , Microambiente Tumoral
6.
ACS Appl Mater Interfaces ; 14(13): 15565-15575, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35286082

RESUMO

Porous silicon (pSi) is an established porous material that offers ample opportunities for biosensor design thanks to its tunable structure, versatile surface chemistry, and large surface area. Nonetheless, its potential for electrochemical sensing is relatively unexplored. This study investigates layered carbon-stabilized pSi nanostructures with site-specific functionalities as an electrochemical biosensor. A double-layer nanostructure combining a top hydrophilic layer of thermally carbonized pSi (TCpSi) and a bottom hydrophobic layer of thermally hydrocarbonized pSi (THCpSi) is prepared. The modified layers are formed in a stepwise process, involving first an electrochemical anodization step to generate a porous layer with precisely defined pore morphological features, followed by deposition of a thin thermally carbonized coating on the pore walls via temperature-controlled acetylene decomposition. The second layer is then generated beneath the first by following the same two-step process, but the acetylene decomposition conditions are adjusted to deposit a thermally hydrocarbonized coating. The double-layer platform features excellent electrochemical properties such as fast electron-transfer kinetics, which underpin the performance of a TCpSi-THCpSi voltammetric DNA sensor. The biosensor targets a 28-nucleotide single-stranded DNA sequence with a detection limit of 0.4 pM, two orders of magnitude lower than the values reported to date by any other pSi-based electrochemical DNA sensor.


Assuntos
Técnicas Biossensoriais , Nanoestruturas , Carbono/química , Nanoestruturas/química , Porosidade , Silício/química
7.
Lab Chip ; 21(20): 4016-4030, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34487130

RESUMO

Compartmentalized microfluidic devices are becoming increasingly popular and have proven to be valuable tools to probe neurobiological functions that are inherently difficult to study using traditional approaches. The ability of microfluidic devices to compartmentalize neurons offers considerable promise for disease modeling and drug discovery. Rodent cortical neurons/neural progenitors are commonly used in such studies but, while these cells mature rapidly, they do not possess the same receptors, ion channels and transport proteins found in human cortical neurons. Human pluripotent stem cell derived neurons offer a human phenotype, but their slow maturation offsets this phenotypic advantage, particularly over long-term culture where overgrowth and subsequent death of neurons may be a problem. In this work, we integrate the use of Matrigel as a 3D cell culture scaffold that enables high cell seeding density over a small fraction of the culture surface. This approach, in an open chamber microfluidic system, enables culture over a five-month period without the use of growth inhibitors. Matrigel was also uniquely utilized to hinder agonist diffusion across microchannels. We demonstrate the development of neuron-to-neuron communication networks by showing that electrical stimulation or the unilateral addition of agonists to one chamber resulted in activation of neurons in the adjacent chamber. Lastly, using a delayed neuron seeding strategy, we show that we can foster essentially one-way communication between separate populations of human forebrain and midbrain dopaminergic neuron containing cultures.


Assuntos
Microfluídica , Células-Tronco Pluripotentes , Diferenciação Celular , Neurônios Dopaminérgicos , Humanos , Mesencéfalo , Prosencéfalo
8.
Adv Sci (Weinh) ; 8(10): 2003937, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34026447

RESUMO

Neurological disorders such as Alzheimer's disease, stroke, and brain cancers are difficult to treat with current drugs as their delivery efficacy to the brain is severely hampered by the presence of the blood-brain barrier (BBB). Drug delivery systems have been extensively explored in recent decades aiming to circumvent this barrier. In particular, polymeric nanoparticles have shown enormous potentials owing to their unique properties, such as high tunability, ease of synthesis, and control over drug release profile. However, careful analysis of their performance in effective drug transport across the BBB should be performed using clinically relevant testing models. In this review, polymeric nanoparticle systems for drug delivery to the central nervous system are discussed with an emphasis on the effects of particle size, shape, and surface modifications on BBB penetration. Moreover, the authors critically analyze the current in vitro and in vivo models used to evaluate BBB penetration efficacy, including the latest developments in the BBB-on-a-chip models. Finally, the challenges and future perspectives for the development of polymeric nanoparticles to combat neurological disorders are discussed.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/administração & dosagem , Doenças Neurodegenerativas/tratamento farmacológico , Polímeros/administração & dosagem , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Humanos , Nanopartículas/química , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Tamanho da Partícula , Polímeros/química
9.
Nanoscale Adv ; 3(3): 682-691, 2021 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-36133829

RESUMO

Nanomaterials are widely used in industrial and clinical settings due to their unique physical and chemical properties. However, public health and environmental concerns have emerged owing to their undesired toxicity and ability to trigger immune responses. This paper presents the development of a microfluidic-based cell biochip device that enables the administration of nanoparticles under laminar flow to cells of the immune system to assess their cytotoxicity. The exposure of human B lymphocytes to 10 nm silver nanoparticles under fluid flow led to a 3-fold increase in toxicity compared to static conditions, possibly indicating enhanced cell-nanoparticle interactions. To investigate whether the administration under flow was the main contributing factor, we compared and validated the cytotoxicity of the same nanoparticles in different platforms, including the conventional well plate format and in-house fabricated microfluidic devices under both static and dynamic flow conditions. Our results suggest that commonly employed static platforms might not be well-suited to perform toxicological screening of nanomaterials and may lead to an underestimation of cytotoxic responses. The simplicity of the developed flow system makes this setup a valuable tool to preliminary screen nanomaterials.

10.
Biomacromolecules ; 22(2): 441-453, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33320642

RESUMO

Waterborne diarrheal diseases such as travelers' diarrhea and cholera remain a threat to public health in many countries. Rapid diagnosis of an infectious disease is critical in preventing the escalation of a disease outbreak into an epidemic. Many of the diagnostic tools for infectious diseases employed today are time-consuming and require specialized laboratory settings and trained personnel. There is hence a pressing need for fit-for-purpose point-of-care diagnostic tools with emphasis in sensitivity, specificity, portability, and low cost. We report work toward thermally reversible biosensors for detection of the carbohydrate-binding domain of the Escherichia coli heat-labile enterotoxin (LTB), a toxin produced by enterotoxigenic E. coli strains, which causes travelers' diarrhea. The biosensing platform is a hybrid of two materials, combining the optical properties of porous silicon (pSi) interferometric transducers and a thermoresponsive multivalent glycopolymer, to enable recognition of LTB. Analytical performance of our biosensors allows us to detect, using a label-free format, sub-micromolar concentrations of LTB in solution as low as 0.135 µM. Furthermore, our platform shows a temperature-mediated "catch-and-release" behavior, an exciting feature with potential for selective protein capture, multiple readouts, and regeneration of the sensor over consecutive cycles of use.


Assuntos
Toxinas Bacterianas , Infecções por Escherichia coli , Proteínas de Escherichia coli , Diarreia , Escherichia coli , Humanos , Viagem
11.
ACS Appl Mater Interfaces ; 12(51): 56753-56766, 2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33226228

RESUMO

Here, we have developed and evaluated a microfluidic-based human blood-brain-barrier (µBBB) platform that models and predicts brain tissue uptake of small molecule drugs and nanoparticles (NPs) targeting the central nervous system. By using a photocrosslinkable copolymer that was prepared from monomers containing benzophenone and N-hydroxysuccinimide ester functional groups, we were able to evenly coat and functionalize µBBB chip channels in situ, providing a covalently attached homogenous layer of extracellular matrix proteins. This novel approach allowed the coculture of human endothelial cells, pericytes, and astrocytes and resulted in the formation of a mimic of cerebral endothelium expressing tight junction markers and efflux proteins, resembling the native BBB. The permeability coefficients of a number of compounds, including caffeine, nitrofurantoin, dextran, sucrose, glucose, and alanine, were measured on our µBBB platform and were found to agree with reported values. In addition, we successfully visualized the receptor-mediated uptake and transcytosis of transferrin-functionalized NPs. The BBB-penetrating NPs were able to target glioma cells cultured in 3D in the brain compartment of our µBBB. In conclusion, our µBBB was able to accurately predict the BBB permeability of both small molecule pharmaceuticals and nanovectors and allowed time-resolved visualization of transcytosis. Our versatile chip design accommodates different brain disease models and is expected to be exploited in further BBB studies, aiming at replacing animal experiments.


Assuntos
Órgãos Artificiais , Barreira Hematoencefálica/metabolismo , Dispositivos Lab-On-A-Chip , Nanopartículas/química , Compostos Orgânicos/análise , Astrócitos/metabolismo , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/metabolismo , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Pericitos/metabolismo , Transferrina/química
12.
Trends Biotechnol ; 37(12): 1295-1314, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31130308

RESUMO

Therapeutic options for neurological disorders currently remain limited. The intrinsic complexity of the brain architecture prevents potential therapeutics from reaching their cerebral target, thus limiting their efficacy. Recent advances in microfluidic technology and organ-on-chip systems have enabled the development of a new generation of in vitro platforms that can recapitulate complex in vivo microenvironments and physiological responses. In this context, microfluidic-based in vitro models of the blood-brain barrier (BBB) are of particular interest as they provide an innovative approach for conducting research related to the brain, including modeling of neurodegenerative diseases and high-throughput drug screening. Here, we present the most recent advances in BBB-on-chip devices and examine validation steps that will strengthen their future applications.


Assuntos
Encefalopatias , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas , Modelos Cardiovasculares , Modelos Neurológicos , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encefalopatias/metabolismo , Encefalopatias/patologia , Humanos
13.
Biomicrofluidics ; 13(4): 044112, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31893008

RESUMO

Microfluidic screening is gaining attention as an efficient method for evaluating nanomaterial toxicity. Here, we consider a multiparameter treatment where nanomaterials interact with cells in the presence of a secondary exposure (UV radiation). The microfluidic device contains channels that permit immobilization of HaCaT cells (human skin cell line), delivery of titanium dioxide nanoparticles (TNPs), and exposure to a known dose of UV radiation. The effect of single-parameter exposures (UV or TNP) was first studied as a benchmark, and then multiparameter toxicity (UV and TNP) at different concentrations was explored. The results demonstrate a concentration-dependent protective effect of TNP when exposed to UV irradiation.

14.
Anal Chem ; 90(7): 4338-4347, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29498509

RESUMO

With advances in nanotechnology, particles with various size, shape, surface chemistry, and composition can be easily produced. Nano- and microparticles have been extensively explored in many industrial and clinical applications. Ensuring that the particles themselves are not possessing toxic effects to the biological system is of paramount importance. This paper describes a proof of concept method, in which a microfluidic system is used in conjunction with a cell microarray technique aiming to streamline the analysis of particle-cell interaction in a high throughput manner. Polymeric microparticles, with different particle surface functionalities, were first used to investigate the efficiency of particle-cell adhesion under dynamic flow. Silver nanoparticles (AgNPs, 10 nm in diameter) perfused at different concentrations (0 to 20 µg/mL) in parallel streams over the cell microarray exhibited a higher toxicity compared to the static culture in the 96-well-plate format. This developed microfluidic system can be easily scaled up to accommodate a larger number of microchannels for high throughput analysis of the potential toxicity of a wide range of particles in a single experiment.


Assuntos
Ensaios de Triagem em Larga Escala , Nanopartículas Metálicas/química , Técnicas Analíticas Microfluídicas , Prata/química , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Desenho de Equipamento , Humanos , Estrutura Molecular , Tamanho da Partícula , Prata/farmacologia , Propriedades de Superfície
15.
Biofabrication ; 10(1): 014101, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29058681

RESUMO

Binding and maintaining cells inside microfluidic channels is a challenging task due to the potential release of cells from the channels with the flow and accompanying shear stress. In this work we optimized the binding of human B-lymphocyte cells (HR1K) inside a microfluidic channel and determined the strength of this binding under shear stress of flowing liquid. In order to determine the parameters required for a live/dead test in microfluidic devices, populations of both living and dead cells were tested separately. Channels were prepared in glass-polydimethylsiloxane hybrid chips, with a self-assembled monolayer of 3-(glycidyloxypropyl)trimethoxysilane (GPTMS) before covalently immobilizing anti-CD20 antibody. Without GPTMS linker, ∼90% of the CD20-expressing cells detached at 200 µl min-1 (the highest flow rate studied). With GPTMS linker, the bonding method proved critical for sustained immobilization of HR1K cells under flow. Masking the channel area during plasma bonding preserves the antibody functionality; the masked surface gives 15% cell detachment at 200 µl min-1 compared with 80% for an unmasked surface. Sealing the chip via clamping (without plasma treatment) was similar to masked plasma treatment (20% detachment) and allowing a post-adhesion stasis time (30 min) did not significantly change the relative cell detachment for the flow rates studied. Membrane integrity and calcium spiking behaviour were measured fluorescently, and demonstrated that the live cells retained comparable functionality to unanchored cells for the duration of the flow experiments. Non-viable HR1K cells were found to detach more readily, exhibiting only 20% cell retention at 200 µl min-1 compared with >80% for live cells.


Assuntos
Linfócitos B/metabolismo , Microfluídica/métodos , Resistência ao Cisalhamento , Morte Celular , Linhagem Celular , Sobrevivência Celular , Humanos , Espectrometria de Massa de Íon Secundário , Propriedades de Superfície , Fatores de Tempo
16.
Lab Chip ; 17(3): 501-510, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28074962

RESUMO

This paper describes the use of crossed laminar flow microfluidics for the selective capture of multiple cell types on-chip aiming for high throughput screening of various cell treatment compounds. Parallel laminar streams containing different cell types were perfused and captured on a cell adhesion protein-functionalized reaction area. Thereafter, parallel streams containing cell treatment solutions were delivered orthogonally over the captured cells. Multiple cell types and a range of cell treatment conditions could therefore be assessed in a single experiment. We were also able to sort mixed cell populations via antibody array clusters, and to further deliver treatments to subpopulations of cells. Moreover, using solutions with different tonicities, we successfully demonstrated the incorporation of a live/dead cell viability assessment on-chip for a direct read out assay following the treatments. This crossed laminar flow microfluidics for generation of a cell-based assay could therefore offer an interesting platform for high throughput screening of potential drug candidates, nanoparticle toxicity testing, or other cellular and molecular interventions.


Assuntos
Separação Celular/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Linhagem Celular , Separação Celular/métodos , Desenho de Equipamento , Ensaios de Triagem em Larga Escala/métodos , Humanos , Técnicas Analíticas Microfluídicas/métodos
17.
Cell ; 157(3): 611-23, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24726433

RESUMO

Cell migration is a critical process for diverse (patho)physiological phenomena. Intriguingly, cell migration through physically confined spaces can persist even when typical hallmarks of 2D planar migration, such as actin polymerization and myosin II-mediated contractility, are inhibited. Here, we present an integrated experimental and theoretical approach ("Osmotic Engine Model") and demonstrate that directed water permeation is a major mechanism of cell migration in confined microenvironments. Using microfluidic and imaging techniques along with mathematical modeling, we show that tumor cells confined in a narrow channel establish a polarized distribution of Na+/H+ pumps and aquaporins in the cell membrane, which creates a net inflow of water and ions at the cell leading edge and a net outflow of water and ions at the trailing edge, leading to net cell displacement. Collectively, this study presents an alternate mechanism of cell migration in confinement that depends on cell-volume regulation via water permeation.


Assuntos
Movimento Celular , Modelos Biológicos , Água , Actinas/metabolismo , Animais , Aquaporina 5/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Tamanho Celular , Humanos , Camundongos , Trocadores de Sódio-Hidrogênio/metabolismo
18.
FASEB J ; 26(10): 4045-56, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22707566

RESUMO

Cell migration on planar surfaces is driven by cycles of actin protrusion, integrin-mediated adhesion, and myosin-mediated contraction; however, this mechanism may not accurately describe movement in 3-dimensional (3D) space. By subjecting cells to restrictive 3D environments, we demonstrate that physical confinement constitutes a biophysical stimulus that alters cell morphology and suppresses mesenchymal motility in human breast carcinoma (MDA-MB-231). Dorsoventral polarity, stress fibers, and focal adhesions are markedly attenuated by confinement. Inhibitors of myosin, Rho/ROCK, or ß1-integrins do not impair migration through 3-µm-wide channels (confinement), even though these treatments repress motility in 50-µm-wide channels (unconfined migration) by ≥50%. Strikingly, confined migration persists even when F-actin is disrupted, but depends largely on microtubule (MT) dynamics. Interfering with MT polymerization/depolymerization causes confined cells to undergo frequent directional changes, thereby reducing the average net displacement by ≥80% relative to vehicle controls. Live-cell EB1-GFP imaging reveals that confinement redirects MT polymerization toward the leading edge, where MTs continuously impact during advancement of the cell front. These results demonstrate that physical confinement can induce cytoskeletal alterations that reduce the dependence of migrating cells on adhesion-contraction force coupling. This mechanism may explain why integrins can exhibit reduced or altered function during migration in 3D environments.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Actinas/metabolismo , Amidas/farmacologia , Azepinas/farmacologia , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Humanos , Microtúbulos/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Naftalenos/farmacologia , Paclitaxel/farmacologia , Piridinas/farmacologia , Moduladores de Tubulina/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
19.
Integr Biol (Camb) ; 4(8): 847-56, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22627390

RESUMO

Receptor-ligand adhesive interactions play a pivotal role in diverse biological processes including inflammation and cancer metastasis. Cell adhesion is mediated by the molecular recognition of membrane-bound receptors by their cognate ligands on apposing cells. Cell-cell binding is regulated by distinct parameters such as the receptor-ligand binding kinetics, the tensile strength of individual bonds, the involvement of multiple bonds and their modulation by hydrodynamic shear. This work aims to investigate the interplay of these parameters on selectin-mediated cell adhesion in shear flow. We designed a microfluidic device that delivers cells in a single file over a receptor-functionalized substrate, thereby permitting accurate determination of the cell flux. The selectin(s) was presented on striped patches of fixed width and varying length. We identified the critical patch lengths of P- and L-selectin for the initiation of HL-60 cell binding in shear flow. This characteristic length is governed by the time required to form multiple-bond interactions, as revealed by a multiple-bond mathematical model. The number of bonds required to support cell binding increases with the applied shear stress (0.5-2 dyn cm(-2)) for L- but not P-selectin. This finding is explained by differences in the tensile strength of P- and L-selectin for PSGL-1. Our integrated experimental and mathematical approach advances our understanding of receptor-mediated cell adhesion in the vasculature. Detailed knowledge of how molecular interactions modulate macroscopic cell binding behavior pertinent to inflammation and metastasis would facilitate the development of promising diagnostic tools to combat these diseases.


Assuntos
Selectina L/metabolismo , Selectina-P/metabolismo , Adesão Celular , Comunicação Celular , Relação Dose-Resposta a Droga , Desenho de Equipamento , Európio/química , Células HL-60 , Humanos , Hidrodinâmica , Íons , Cinética , Teste de Materiais , Glicoproteínas de Membrana/metabolismo , Técnicas Analíticas Microfluídicas , Ligação Proteica , Resistência ao Cisalhamento , Estresse Mecânico , Propriedades de Superfície , Resistência à Tração
20.
PLoS One ; 7(1): e29211, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22279529

RESUMO

Cell migration is crucial for both physiological and pathological processes. Current in vitro cell motility assays suffer from various drawbacks, including insufficient temporal and/or optical resolution, or the failure to include a controlled chemotactic stimulus. Here, we address these limitations with a migration chamber that utilizes a self-sustaining chemotactic gradient to induce locomotion through confined environments that emulate physiological settings. Dynamic real-time analysis of both population-scale and single-cell movement are achieved at high resolution. Interior surfaces can be functionalized through adsorption of extracellular matrix components, and pharmacological agents can be administered to cells directly, or indirectly through the chemotactic reservoir. Direct comparison of multiple cell types can be achieved in a single enclosed system to compare inherent migratory potentials. Our novel microfluidic design is therefore a powerful tool for the study of cellular chemotaxis, and is suitable for a wide range of biological and biomedical applications.


Assuntos
Movimento Celular/fisiologia , Quimiotaxia/fisiologia , Técnicas Analíticas Microfluídicas/métodos , Análise de Célula Única/métodos , Actinas/genética , Actinas/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Dimetilpolisiloxanos/química , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Paclitaxel/farmacologia , Reprodutibilidade dos Testes , Análise de Célula Única/instrumentação , Tiazolidinas/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...