Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 21, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167896

ABSTRACT

Membrane fusion and budding mediate fundamental processes like intracellular trafficking, exocytosis, and endocytosis. Fusion is thought to open a nanometer-range pore that may subsequently close or dilate irreversibly, whereas budding transforms flat membranes into vesicles. Reviewing recent breakthroughs in real-time visualization of membrane transformations well exceeding this classical view, we synthesize a new model and describe its underlying mechanistic principles and functions. Fusion involves hemi-to-full fusion, pore expansion, constriction and/or closure while fusing vesicles may shrink, enlarge, or receive another vesicle fusion; endocytosis follows exocytosis primarily by closing Ω-shaped profiles pre-formed through the flat-to-Λ-to-Ω-shape transition or formed via fusion. Calcium/SNARE-dependent fusion machinery, cytoskeleton-dependent membrane tension, osmotic pressure, calcium/dynamin-dependent fission machinery, and actin/dynamin-dependent force machinery work together to generate fusion and budding modes differing in pore status, vesicle size, speed and quantity, controls release probability, synchronization and content release rates/amounts, and underlies exo-endocytosis coupling to maintain membrane homeostasis. These transformations, underlying mechanisms, and functions may be conserved for fusion and budding in general.


Subject(s)
Calcium , Membrane Fusion , Cell Membrane , Exocytosis , Dynamins , Secretory Vesicles
2.
Methods Mol Biol ; 2565: 77-87, 2023.
Article in English | MEDLINE | ID: mdl-36205888

ABSTRACT

Recent advances in stimulated emission depletion (STED) microscopy offer an unparalleled avenue to study membrane dynamics of exo- and endocytosis, such as fusion pore opening, pore expansion, constriction, and closure, as well as the membrane transformation from flat-shaped to round-shaped vesicles in real time. Here we depict a method of using the state-of-the-art STED microscopy to image these membrane dynamics in bovine chromaffin cells. This method can potentially be applied to study other membrane structure dynamics in other cell model system.


Subject(s)
Chromaffin Cells , Microscopy , Animals , Cattle , Cell Membrane/metabolism , Endocytosis , Secretory Vesicles/metabolism
3.
Biochem Soc Trans ; 50(4): 1157-1167, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35960003

ABSTRACT

Visualization of cellular dynamics using fluorescent light microscopy has become a reliable and indispensable source of experimental evidence for biological studies. Over the past two decades, the development of super-resolution microscopy platforms coupled with innovations in protein and molecule labeling led to significant biological findings that were previously unobservable due to the barrier of the diffraction limit. As a result, the ability to image the dynamics of cellular processes is vastly enhanced. These imaging tools are extremely useful in cellular physiology for the study of vesicle fusion and endocytosis. In this review, we will explore the power of stimulated emission depletion (STED) and confocal microscopy in combination with various labeling techniques in real-time observation of the membrane transformation of fusion and endocytosis, as well as their underlying mechanisms. We will review how STED and confocal imaging are used to reveal fusion and endocytic membrane transformation processes in live cells, including hemi-fusion; hemi-fission; hemi-to-full fusion; fusion pore opening, expansion, constriction and closure; shrinking or enlargement of the Ω-shape membrane structure after vesicle fusion; sequential compound fusion; and the sequential endocytic membrane transformation from flat- to O-shape via the intermediate Λ- and Ω-shape transition. We will also discuss how the recent development of imaging techniques would impact future studies in the field.


Subject(s)
Endocytosis , Membrane Fusion , Cell Membrane/metabolism , Endocytosis/physiology , Exocytosis/physiology , Membrane Fusion/physiology , Microscopy, Confocal , Secretory Vesicles/physiology
4.
Nat Commun ; 13(1): 3697, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35760780

ABSTRACT

Membrane budding entails forces to transform flat membrane into vesicles essential for cell survival. Accumulated studies have identified coat-proteins (e.g., clathrin) as potential budding factors. However, forces mediating many non-coated membrane buddings remain unclear. By visualizing proteins in mediating endocytic budding in live neuroendocrine cells, performing in vitro protein reconstitution and physical modeling, we discovered how non-coated-membrane budding is mediated: actin filaments and dynamin generate a pulling force transforming flat membrane into Λ-shape; subsequently, dynamin helices surround and constrict Λ-profile's base, transforming Λ- to Ω-profile, and then constrict Ω-profile's pore, converting Ω-profiles to vesicles. These mechanisms control budding speed, vesicle size and number, generating diverse endocytic modes differing in these parameters. Their impact is widespread beyond secretory cells, as the unexpectedly powerful functions of dynamin and actin, previously thought to mediate fission and overcome tension, respectively, may contribute to many dynamin/actin-dependent non-coated-membrane buddings, coated-membrane buddings, and other membrane remodeling processes.


Subject(s)
Actins , Endocytosis , Actins/metabolism , Cell Membrane/metabolism , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Dynamins/metabolism
5.
Sci Adv ; 8(24): eabm6049, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35714180

ABSTRACT

Vesicle fusion at preestablished plasma membrane release sites releases transmitters and hormones to mediate fundamental functions like neuronal network activities and fight-or-flight responses. This half-a-century-old concept-fusion at well-established release sites in excitable cells-needs to be modified to include the sequential compound fusion reported here-vesicle fusion at previously fused Ω-shaped vesicular membrane. With superresolution STED microscopy in excitable neuroendocrine chromaffin cells, we real-time visualized sequential compound fusion pore openings and content releases in generating multivesicular and asynchronous release from single release sites, which enhances exocytosis strength and dynamic ranges in excitable cells. We also visualized subsequent compound fusion pore closure, a new mode of endocytosis termed compound kiss-and-run that enhances vesicle recycling capacity. These results suggest modifying current exo-endocytosis concepts by including rapid release-site assembly at fused vesicle membrane, where sequential compound fusion and kiss-and-run take place to enhance exo-endocytosis capacity and dynamic ranges.

6.
Cell Rep Methods ; 2(4): 100206, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35497501

ABSTRACT

Vesicle exo- and endocytosis mediate important biological functions, including synaptic transmission. In this issue of Cell Reports Methods, Seong J. An et al. found that the fluorescently tagged C2 domain of phospholipase A2 binds to membrane phosphatidylcholine and thus labels vesicle membrane, allowing for super-resolution and electron microscopic visualization of vesicle trafficking.


Subject(s)
Endocytosis , Synaptic Vesicles , Synaptic Vesicles/metabolism , Phospholipases A2/metabolism , Synaptic Transmission , Multimodal Imaging
7.
Front Synaptic Neurosci ; 14: 841704, 2022.
Article in English | MEDLINE | ID: mdl-35308832

ABSTRACT

Cytoskeletal filamentous actin (F-actin) has long been considered a molecule that may regulate exo- and endocytosis. However, its exact roles remained elusive. Recent studies shed new light on many crucial roles of F-actin in regulating exo- and endocytosis. Here, this progress is reviewed from studies of secretory cells, particularly neurons and endocrine cells. These studies reveal that F-actin is involved in mediating all kinetically distinguishable forms of endocytosis, including ultrafast, fast, slow, bulk, and overshoot endocytosis, likely via membrane pit formation. F-actin promotes vesicle replenishment to the readily releasable pool most likely via active zone clearance, which may sustain synaptic transmission and overcome short-term depression of synaptic transmission during repetitive firing. By enhancing plasma membrane tension, F-actin promotes fusion pore expansion, vesicular content release, and a fusion mode called shrink fusion involving fusing vesicle shrinking. Not only F-actin, but also the F-actin assembly pathway, including ATP hydrolysis, N-WASH, and formin, are involved in mediating these roles of exo- and endocytosis. Neurological disorders, including spinocerebellar ataxia 13 caused by Kv3.3 channel mutation, may involve impairment of F-actin and its assembly pathway, leading in turn to impairment of exo- and endocytosis at synapses that may contribute to neurological disorders.

8.
J Biol Chem ; 298(5): 101845, 2022 05.
Article in English | MEDLINE | ID: mdl-35307352

ABSTRACT

Enzymes within the de novo purine biosynthetic pathway spatially organize into dynamic intracellular assemblies called purinosomes. The formation of purinosomes has been correlated with growth conditions resulting in high purine demand, and therefore, the cellular advantage of complexation has been hypothesized to enhance metabolite flux through the pathway. However, the properties of this cellular structure are unclear. Here, we define the purinosome in a transient expression system as a biomolecular condensate using fluorescence microscopy. We show that purinosomes, as denoted by formylglycinamidine ribonucleotide synthase granules in purine-depleted HeLa cells, are spherical and appear to coalesce when two come into contact, all liquid-like characteristics that are consistent with previously reported condensates. We further explored the biophysical and biochemical means that drive the liquid-liquid phase separation of these structures. We found that the process of enzyme condensation into purinosomes is likely driven by the oligomeric state of the pathway enzymes and not a result of intrinsic disorder, the presence of low-complexity domains, the assistance of RNA scaffolds, or changes in intracellular pH. Finally, we demonstrate that the heat shock protein 90 KDa helps to regulate the physical properties of the condensate and maintain their liquid-like state inside HeLa cells. We show that disruption of heat shock protein 90 KDa activity induced the transformation of formylglycinamidine ribonucleotide synthase clusters into more irregularly shaped condensates, suggesting that its chaperone activity is essential for purinosomes to retain their liquid-like properties. This refined view of the purinosome offers new insight into how metabolic enzymes spatially organize into dynamic condensates within human cells.


Subject(s)
HSP90 Heat-Shock Proteins , Purines , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Molecular Chaperones/genetics , Purines/metabolism , Ribonucleotides
9.
iScience ; 25(2): 103809, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35198874

ABSTRACT

Clathrin-mediated endocytosis, the most prominent endocytic mode, is thought to be generated primarily from relatively flat patches of the plasma membrane. By employing conventional and platinum replica electron microscopy and super-resolution STED microscopy in neuroendocrine chromaffin cells, we found that large Ω-shaped or dome-shaped plasma membrane invaginations, previously thought of as the precursor of bulk endocytosis, are primary sites for clathrin-coated pit generation after depolarization. Clathrin-coated pits are more densely packed at invaginations rather than flat membranes, suggesting that invaginations are preferred sites for clathrin-coated pit formation, likely because their positive curvature facilitates coated-pit formation. Thus, clathrin-mediated endocytosis closely collaborates with bulk endocytosis to enhance endocytic capacity in active secretory cells. This direct collaboration between two classically independent endocytic pathways is of broad importance given the central role of both clathrin-mediated endocytosis and bulk endocytosis in neurons, endocrine cells, immune cells, and many other cell types throughout the body.

10.
Neuron ; 109(19): 3119-3134.e5, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34411513

ABSTRACT

Transformation of flat membrane into round vesicles is generally thought to underlie endocytosis and produce speed-, amount-, and vesicle-size-specific endocytic modes. Visualizing depolarization-induced exocytic and endocytic membrane transformation in live neuroendocrine chromaffin cells, we found that flat membrane is transformed into Λ-shaped, Ω-shaped, and O-shaped vesicles via invagination, Λ-base constriction, and Ω-pore constriction, respectively. Surprisingly, endocytic vesicle formation is predominantly from not flat-membrane-to-round-vesicle transformation but calcium-triggered and dynamin-mediated closure of (1) Ω profiles formed before depolarization and (2) fusion pores (called kiss-and-run). Varying calcium influxes control the speed, number, and vesicle size of these pore closures, resulting in speed-specific slow (more than ∼6 s), fast (less than ∼6 s), or ultrafast (<0.6 s) endocytosis, amount-specific compensatory endocytosis (endocytosis = exocytosis) or overshoot endocytosis (endocytosis > exocytosis), and size-specific bulk endocytosis. These findings reveal major membrane transformation mechanisms underlying endocytosis, diverse endocytic modes, and exocytosis-endocytosis coupling, calling for correction of the half-a-century concept that the flat-to-round transformation predominantly mediates endocytosis after physiological stimulation.


Subject(s)
Chromaffin Cells/physiology , Chromaffin Cells/ultrastructure , Endocytosis/physiology , Neuroendocrine Cells/physiology , Neuroendocrine Cells/ultrastructure , Animals , Calcium Signaling , Cattle , Cell Fusion , Cell Membrane/physiology , Cell Membrane/ultrastructure , Computer Systems , Dynamins/physiology , Exocytosis/physiology , Membrane Fusion , Primary Cell Culture , Synaptic Vesicles/metabolism
11.
Proc Natl Acad Sci U S A ; 115(51): 13009-13014, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30509995

ABSTRACT

To meet their purine demand, cells activate the de novo purine biosynthetic pathway and transiently cluster the pathway enzymes into metabolons called purinosomes. Recently, we have shown that purinosomes were spatially colocalized with mitochondria and microtubules, yet it remained unclear as to what drives these associations and whether a relationship between them exist. Here, we employed superresolution imaging methods to describe purinosome transit in the context of subcellular localization. Time-resolved imaging of purinosomes showed that these assemblies exhibit directed motion as they move along a microtubule toward mitochondria, where upon colocalization, a change in purinosome motion was observed. A majority of purinosomes colocalized with mitochondria were also deemed colocalized with microtubules. Nocodazole-dependent microtubule depolymerization resulted in a loss in the purinosome-mitochondria colocalization, suggesting that the association of purinosomes with mitochondria is facilitated by microtubule-directed transport, and thereby supporting our notion of an interdependency between these subcellular components in maximizing purine production through the de novo purine biosynthetic pathway.


Subject(s)
Cytosol/metabolism , Metabolome , Microtubules/metabolism , Mitochondria/metabolism , Purines/metabolism , Biosynthetic Pathways , HeLa Cells , Humans
12.
Lab Chip ; 18(10): 1411-1421, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29668002

ABSTRACT

Resolving the temporal dynamics of cell signaling pathways is essential for regulating numerous downstream functions, from gene expression to cellular responses. Mapping these signaling pathways requires the exposure of cells to time-varying chemical signals; these are difficult to generate and control over a wide temporal range. Herein, we present an acoustofluidic chemical signal generator based on a sharp-edge-based micromixing strategy. The device, simply by modulating the driving signals of an acoustic transducer including the ON/OFF switching frequency, actuation time and duty cycle, is capable of generating both single-pulse and periodic chemical signals that are temporally controllable in terms of stimulation period, stimulation duration and duty cycle. We also demonstrate the device's applicability and versatility for cell signaling studies by probing the calcium (Ca2+) release dynamics of three different types of cells stimulated by ionomycin signals of different shapes. Upon short single-pulse ionomycin stimulation (∼100 ms) generated by our device, we discover that cells tend to dynamically adjust the intracellular level of Ca2+ through constantly releasing and accepting Ca2+ to the cytoplasm and from the extracellular environment, respectively. With advantages such as simple fabrication and operation, compact device design, and reliability and versatility, our device will enable decoding of the temporal characteristics of signaling dynamics for various physiological processes.


Subject(s)
Acoustics , Microfluidic Analytical Techniques/methods , Models, Biological , Signal Transduction/physiology , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line , HeLa Cells , Humans , Ionomycin/pharmacology , Signal Transduction/drug effects
13.
Lab Chip ; 16(14): 2636-43, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27327102

ABSTRACT

The multicellular spheroid is an important 3D cell culture model for drug screening, tissue engineering, and fundamental biological research. Although several spheroid formation methods have been reported, the field still lacks high-throughput and simple fabrication methods to accelerate its adoption in drug development industry. Surface acoustic wave (SAW) based cell manipulation methods, which are known to be non-invasive, flexible, and high-throughput, have not been successfully developed for fabricating 3D cell assemblies or spheroids, due to the limited understanding on SAW-based vertical levitation. In this work, we demonstrated the capability of fabricating multicellular spheroids in the 3D acoustic tweezers platform. Our method used drag force from microstreaming to levitate cells in the vertical direction, and used radiation force from Gor'kov potential to aggregate cells in the horizontal plane. After optimizing the device geometry and input power, we demonstrated the rapid and high-throughput nature of our method by continuously fabricating more than 150 size-controllable spheroids and transferring them to Petri dishes every 30 minutes. The spheroids fabricated by our 3D acoustic tweezers can be cultured for a week with good cell viability. We further demonstrated that spheroids fabricated by this method could be used for drug testing. Unlike the 2D monolayer model, HepG2 spheroids fabricated by the 3D acoustic tweezers manifested distinct drug resistance, which matched existing reports. The 3D acoustic tweezers based method can serve as a novel bio-manufacturing tool to fabricate complex 3D cell assembles for biological research, tissue engineering, and drug development.


Subject(s)
Acoustics/instrumentation , Cell Culture Techniques/instrumentation , Drug Screening Assays, Antitumor/methods , Equipment Design/methods , Spheroids, Cellular/cytology , Cell Culture Techniques/methods , Drug Screening Assays, Antitumor/instrumentation , Fluorouracil/pharmacology , Hep G2 Cells , Humans , Lab-On-A-Chip Devices
14.
Science ; 351(6274): 733-7, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26912862

ABSTRACT

Purine biosynthetic enzymes organize into dynamic cellular bodies called purinosomes. Little is known about the spatiotemporal control of these structures. Using super-resolution microscopy, we demonstrated that purinosomes colocalized with mitochondria, and these results were supported by isolation of purinosome enzymes with mitochondria. Moreover, the number of purinosome-containing cells responded to dysregulation of mitochondrial function and metabolism. To explore the role of intracellular signaling, we performed a kinome screen using a label-free assay and found that mechanistic target of rapamycin (mTOR) influenced purinosome assembly. mTOR inhibition reduced purinosome-mitochondria colocalization and suppressed purinosome formation stimulated by mitochondria dysregulation. Collectively, our data suggest an mTOR-mediated link between purinosomes and mitochondria, and a general means by which mTOR regulates nucleotide metabolism by spatiotemporal control over protein association.


Subject(s)
Mitochondria/metabolism , Purines/metabolism , TOR Serine-Threonine Kinases/metabolism , HeLa Cells , Humans , Microscopy , Mitochondria/ultrastructure , Signal Transduction
15.
Lab Chip ; 15(21): 4166-76, 2015 Nov 07.
Article in English | MEDLINE | ID: mdl-26338516

ABSTRACT

The ability to generate stable, spatiotemporally controllable concentration gradients is critical for resolving the dynamics of cellular response to a chemical microenvironment. Here we demonstrate an acoustofluidic gradient generator based on acoustically oscillating sharp-edge structures, which facilitates in a step-wise fashion the rapid mixing of fluids to generate tunable, dynamic chemical gradients. By controlling the driving voltage of a piezoelectric transducer, we demonstrated that the chemical gradient profiles can be conveniently altered (spatially controllable). By adjusting the actuation time of the piezoelectric transducer, moreover, we generated pulsatile chemical gradients (temporally controllable). With these two characteristics combined, we have developed a spatiotemporally controllable gradient generator. The applicability and biocompatibility of our acoustofluidic gradient generator are validated by demonstrating the migration of human dermal microvascular endothelial cells (HMVEC-d) in response to a generated vascular endothelial growth factor (VEGF) gradient, and by preserving the viability of HMVEC-d cells after long-term exposure to an acoustic field. Our device features advantages such as simple fabrication and operation, compact and biocompatible device, and generation of spatiotemporally tunable gradients.


Subject(s)
Acoustics/instrumentation , Lab-On-A-Chip Devices , Cell Movement , Cell Survival , Endothelial Cells/cytology , Equipment Design , Humans , Spatio-Temporal Analysis
16.
J Biol Chem ; 290(11): 6705-13, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25605736

ABSTRACT

Enzymes in the de novo purine biosynthesis pathway are recruited to form a dynamic metabolic complex referred to as the purinosome. Previous studies have demonstrated that purinosome assembly responds to purine levels in culture medium. Purine-depleted medium or 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole (DMAT) treatment stimulates the purinosome assembly in HeLa cells. Here, several metabolomic technologies were applied to quantify the static cellular levels of purine nucleotides and measure the de novo biosynthesis rate of IMP, AMP, and GMP. Direct comparison of purinosome-rich cells (cultured in purine-depleted medium) and normal cells showed a 3-fold increase in IMP concentration in purinosome-rich cells and similar levels of AMP, GMP, and ratios of AMP/GMP and ATP/ADP for both. In addition, a higher level of IMP was also observed in HeLa cells treated with DMAT. Furthermore, increases in the de novo IMP/AMP/GMP biosynthetic flux rate under purine-depleted condition were observed. The synthetic enzymes, adenylosuccinate synthase (ADSS) and inosine monophosphate dehydrogenase (IMPDH), downstream of IMP were also shown to be part of the purinosome. Collectively, these results provide further evidence that purinosome assembly is directly related to activated de novo purine biosynthesis, consistent with the functionality of the purinosome.


Subject(s)
Metabolomics/methods , Multienzyme Complexes/metabolism , Purine Nucleotides/metabolism , Purines/metabolism , Adenylosuccinate Synthase/analysis , Adenylosuccinate Synthase/metabolism , Benzimidazoles/metabolism , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/analysis , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , HeLa Cells , Humans , IMP Dehydrogenase/analysis , IMP Dehydrogenase/metabolism , Magnetic Resonance Spectroscopy , Multienzyme Complexes/analysis , Purine Nucleotides/analysis , Purines/analysis , Spectrometry, Mass, Electrospray Ionization
17.
Proc Natl Acad Sci U S A ; 112(5): 1368-73, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605889

ABSTRACT

The de novo purine biosynthetic pathway relies on six enzymes to catalyze the conversion of phosphoribosylpyrophosphate to inosine 5'-monophosphate. Under purine-depleted conditions, these enzymes form a multienzyme complex known as the purinosome. Previous studies have revealed the spatial organization and importance of the purinosome within mammalian cancer cells. In this study, time-lapse fluorescence microscopy was used to investigate the cell cycle dependency on purinosome formation in two cell models. Results in HeLa cells under purine-depleted conditions demonstrated a significantly higher number of cells with purinosomes in the G1 phase, which was further confirmed by cell synchronization. HGPRT-deficient fibroblast cells also exhibited the greatest purinosome formation in the G1 phase; however, elevated levels of purinosomes were also observed in the S and G2/M phases. The observed variation in cell cycle-dependent purinosome formation between the two cell models tested can be attributed to differences in purine biosynthetic mechanisms. Our results demonstrate that purinosome formation is closely related to the cell cycle.


Subject(s)
Cell Cycle , Purines/biosynthesis , HeLa Cells , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Microscopy, Fluorescence , Single-Cell Analysis
18.
Biomicrofluidics ; 8(4): 046505, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25379110

ABSTRACT

In this article, we present a simple, rapid prototyped polystyrene-based microfluidic device with three-dimensional (3D) interconnected microporous walls for long term perfusion cell culture. Patterned 3D interconnected microporous structures were created by a chemical treatment together with a protective mask and the native hydrophobic nature of the microporous structures were selectively made hydrophilic using oxygen plasma treatment together with a protective mask. Using this polystyrene-based cell culture microfluidic device, we successfully demonstrated the support of four days perfusion cell culture of hepatocytes (C3A cells).

19.
Opt Lett ; 39(3): 501-4, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24487850

ABSTRACT

We studied the effects of absorption and radiative decay rates of surface plasmon polaritons on the field enhancement in periodic metallic arrays by temporal coupled mode theory and finite-difference time-domain simulation. When two rates are equal, the field enhancement is the strongest and the peak height of the orthogonal reflectivity reaches 0.25. To demonstrate this fact, we fabricated two series of two-dimensional Au and Ag nanohole arrays with different geometries and measured their corresponding reflectivity and decay rates. The experimental results agree well with the analytical and numerical results.

20.
Lab Chip ; 13(24): 4697-710, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24193241

ABSTRACT

Considerable advances have been made in the development of micro-physiological systems that seek to faithfully replicate the complexity and functionality of animal and human physiology in research laboratories. Sometimes referred to as "organs-on-chips", these systems provide key insights into physiological or pathological processes associated with health maintenance and disease control, and serve as powerful platforms for new drug development and toxicity screening. In this Focus article, we review the state-of-the-art designs and examples for developing multiple "organs-on-chips", and discuss the potential of this emerging technology to enhance our understanding of human physiology, and to transform and accelerate the drug discovery and preclinical testing process. This Focus article highlights some of the recent technological advances in this field, along with the challenges that must be addressed for these technologies to fully realize their potential.


Subject(s)
Drug Discovery/methods , Tissue Array Analysis/methods , Animals , Cell Culture Techniques , Drug Discovery/instrumentation , Humans , Systems Integration , Time Factors , Tissue Array Analysis/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...