Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
ALTEX ; 40(3): 534-540, 2023.
Article in English | MEDLINE | ID: mdl-36888967

ABSTRACT

Progress in developing new tools, assays, and approaches to assess human hazard and health risk provides an opportunity to re-evaluate the necessity of dog studies for the safety evaluation of agrochemicals. A workshop was held where partic­ipants discussed the strengths and limitations of past use of dogs for pesticide evaluations and registrations. Opportunities were identified to support alternative approaches to answer human safety questions without performing the required 90-day dog study. Development of a decision tree for determining when the dog study might not be necessary to inform pesticide safety and risk assessment was proposed. Such a process will require global regulatory authority participation to lead to its acceptance. The identification of unique effects in dogs that are not identified in rodents will need further evaluation and determination of their relevance to humans. The establishment of in vitro and in silico approaches that can provide critical data on relative species sensitivity and human relevance will be an important tool to advance the decision process. Promising novel tools including in vitro comparative metabolism studies, in silico models, and high-throughput assays able to identify metabolites and mechanisms of action leading to development of adverse outcome pathways will need further development. To replace or eliminate the 90-day dog study, a collaborative, multidisciplinary, international effort that transcends organi­zations and regulatory agencies will be needed in order to develop guidance on when the study would not be necessary for human safety and risk assessment.


Subject(s)
Adverse Outcome Pathways , Pesticides , Animals , Dogs , Humans , Agrochemicals/toxicity , Pesticides/toxicity , Risk Assessment , Computer Simulation
4.
Commun Med (Lond) ; 2(1): 154, 2022 Dec 06.
Article in English | MEDLINE | ID: mdl-36473994

ABSTRACT

BACKGROUND: Conventional preclinical models often miss drug toxicities, meaning the harm these drugs pose to humans is only realized in clinical trials or when they make it to market. This has caused the pharmaceutical industry to waste considerable time and resources developing drugs destined to fail. Organ-on-a-Chip technology has the potential improve success in drug development pipelines, as it can recapitulate organ-level pathophysiology and clinical responses; however, systematic and quantitative evaluations of Organ-Chips' predictive value have not yet been reported. METHODS: 870 Liver-Chips were analyzed to determine their ability to predict drug-induced liver injury caused by small molecules identified as benchmarks by the Innovation and Quality consortium, who has published guidelines defining criteria for qualifying preclinical models. An economic analysis was also performed to measure the value Liver-Chips could offer if they were broadly adopted in supporting toxicity-related decisions as part of preclinical development workflows. RESULTS: Here, we show that the Liver-Chip met the qualification guidelines across a blinded set of 27 known hepatotoxic and non-toxic drugs with a sensitivity of 87% and a specificity of 100%. We also show that this level of performance could generate over $3 billion annually for the pharmaceutical industry through increased small-molecule R&D productivity. CONCLUSIONS: The results of this study show how incorporating predictive Organ-Chips into drug development workflows could substantially improve drug discovery and development, allowing manufacturers to bring safer, more effective medicines to market in less time and at lower costs.


Drug development is lengthy and costly, as it relies on laboratory models that fail to predict human reactions to potential drugs. Because of this, toxic drugs sometimes go on to harm humans when they reach clinical trials or once they are in the marketplace. Organ-on-a-Chip technology involves growing cells on small devices to mimic organs of the body, such as the liver. Organ-Chips could potentially help identify toxicities earlier, but there is limited research into how well they predict these effects compared to conventional models. In this study, we analyzed 870 Liver-Chips to determine how well they predict drug-induced liver injury, a common cause of drug failure, and found that Liver-Chips outperformed conventional models. These results suggest that widespread acceptance of Organ-Chips could decrease drug attrition, help minimize harm to patients, and generate billions in revenue for the pharmaceutical industry.

5.
Nat Biomed Eng ; 4(4): 407-420, 2020 04.
Article in English | MEDLINE | ID: mdl-31988458

ABSTRACT

Organ chips can recapitulate organ-level (patho)physiology, yet pharmacokinetic and pharmacodynamic analyses require multi-organ systems linked by vascular perfusion. Here, we describe an 'interrogator' that employs liquid-handling robotics, custom software and an integrated mobile microscope for the automated culture, perfusion, medium addition, fluidic linking, sample collection and in situ microscopy imaging of up to ten organ chips inside a standard tissue-culture incubator. The robotic interrogator maintained the viability and organ-specific functions of eight vascularized, two-channel organ chips (intestine, liver, kidney, heart, lung, skin, blood-brain barrier and brain) for 3 weeks in culture when intermittently fluidically coupled via a common blood substitute through their reservoirs of medium and endothelium-lined vascular channels. We used the robotic interrogator and a physiological multicompartmental reduced-order model of the experimental system to quantitatively predict the distribution of an inulin tracer perfused through the multi-organ human-body-on-chips. The automated culture system enables the imaging of cells in the organ chips and the repeated sampling of both the vascular and interstitial compartments without compromising fluidic coupling.


Subject(s)
Cell Culture Techniques/methods , Lab-On-A-Chip Devices , Microfluidics/methods , Robotics/methods , Blood-Brain Barrier , Brain , Calibration , Cell Culture Techniques/instrumentation , Equipment Design , Heart , Humans , Intestines , Kidney , Liver , Lung , Robotics/instrumentation , Skin
6.
Elife ; 92020 01 14.
Article in English | MEDLINE | ID: mdl-31933478

ABSTRACT

Induction of intestinal drug metabolizing enzymes can complicate the development of new drugs, owing to the potential to cause drug-drug interactions (DDIs) leading to changes in pharmacokinetics, safety and efficacy. The development of a human-relevant model of the adult intestine that accurately predicts CYP450 induction could help address this challenge as species differences preclude extrapolation from animals. Here, we combined organoids and Organs-on-Chips technology to create a human Duodenum Intestine-Chip that emulates intestinal tissue architecture and functions, that are relevant for the study of drug transport, metabolism, and DDI. Duodenum Intestine-Chip demonstrates the polarized cell architecture, intestinal barrier function, presence of specialized cell subpopulations, and in vivo relevant expression, localization, and function of major intestinal drug transporters. Notably, in comparison to Caco-2, it displays improved CYP3A4 expression and induction capability. This model could enable improved in vitro to in vivo extrapolation for better predictions of human pharmacokinetics and risk of DDIs.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Drug Interactions , Duodenum/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Caco-2 Cells , Computational Biology , Cytochrome P-450 CYP3A/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Microvilli , Organ Culture Techniques , Organoids/metabolism , Permeability , Transcriptome
7.
Sci Transl Med ; 11(517)2019 11 06.
Article in English | MEDLINE | ID: mdl-31694927

ABSTRACT

Nonclinical rodent and nonrodent toxicity models used to support clinical trials of candidate drugs may produce discordant results or fail to predict complications in humans, contributing to drug failures in the clinic. Here, we applied microengineered Organs-on-Chips technology to design a rat, dog, and human Liver-Chip containing species-specific primary hepatocytes interfaced with liver sinusoidal endothelial cells, with or without Kupffer cells and hepatic stellate cells, cultured under physiological fluid flow. The Liver-Chip detected diverse phenotypes of liver toxicity, including hepatocellular injury, steatosis, cholestasis, and fibrosis, and species-specific toxicities when treated with tool compounds. A multispecies Liver-Chip may provide a useful platform for prediction of liver toxicity and inform human relevance of liver toxicities detected in animal studies to better determine safety and human risk.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/pathology , Lab-On-A-Chip Devices , Liver/pathology , Animals , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dogs , Humans , Kupffer Cells/metabolism , Liver/injuries , Liver Diseases/pathology , Phenotype , Rats , Reproducibility of Results , Risk Factors , Species Specificity
8.
Arch Toxicol ; 93(4): 1021-1037, 2019 04.
Article in English | MEDLINE | ID: mdl-30915487

ABSTRACT

Drug-induced liver injury remains a frequent reason for drug withdrawal. Accordingly, more predictive and translational models are required to assess human hepatotoxicity risk. This study presents a comprehensive evaluation of two promising models to assess mechanistic hepatotoxicity, microengineered Organ-Chips and 3D hepatic spheroids, which have enhanced liver phenotype, metabolic activity and stability in culture not attainable with conventional 2D models. Sensitivity of the models to two hepatotoxins, acetaminophen (APAP) and fialuridine (FIAU), was assessed across a range of cytotoxicity biomarkers (ATP, albumin, miR-122, α-GST) as well as their metabolic functionality by quantifying APAP, FIAU and CYP probe substrate metabolites. APAP and FIAU produced dose- and time-dependent increases in miR-122 and α-GST release as well as decreases in albumin secretion in both Liver-Chips and hepatic spheroids. Metabolic turnover of CYP probe substrates, APAP and FIAU, was maintained over the 10-day exposure period at concentrations where no cytotoxicity was detected and APAP turnover decreased at concentrations where cytotoxicity was detected. With APAP, the most sensitive biomarkers were albumin in the Liver-Chips (EC50 5.6 mM, day 1) and miR-122 and ATP in the liver spheroids (14-fold and EC50 2.9 mM, respectively, day 3). With FIAU, the most sensitive biomarkers were albumin in the Liver-Chip (EC50 126 µM) and miR-122 (15-fold) in the liver spheroids, both on day 7. In conclusion, both models exhibited integrated toxicity and metabolism, and broadly similar sensitivity to the hepatotoxicants at relevant clinical concentrations, demonstrating the utility of these models for improved hepatotoxicity risk assessment.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/drug effects , Liver/drug effects , Models, Biological , Spheroids, Cellular/drug effects , Acetaminophen/toxicity , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/toxicity , Biomarkers/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Spheroids, Cellular/metabolism
9.
Lab Chip ; 19(3): 410-421, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30663729

ABSTRACT

Organ-Chips are micro-engineered systems that aim to recapitulate the organ microenvironment. Implementation of Organ-Chips within the pharmaceutical industry aims to improve the probability of success of drugs reaching late stage clinical trial by generating models for drug discovery that are of human origin and have disease relevance. We are adopting the use of Organ-Chips for enhancing pre-clinical efficacy and toxicity evaluation and prediction. Whilst capturing cellular phenotype via imaging in response to drug exposure is a useful readout in these models, application has been limited due to difficulties in imaging the chips at scale. Here we created an end-to-end, automated workflow to capture and analyse confocal images of multicellular Organ-Chips to assess detailed cellular phenotype across large batches of chips. By automating this process, we not only reduced acquisition time, but we also minimised process variability and user bias. This enabled us to establish, for the first time, a framework of statistical best practice for Organ-Chip imaging, creating the capability of using Organ-Chips and imaging for routine testing in drug discovery applications that rely on quantitative image data for decision making. We tested our approach using benzbromarone, whose mechanism of toxicity has been linked to mitochondrial damage with subsequent induction of apoptosis and necrosis, and staurosporine, a tool inducer of apoptosis. We also applied this workflow to assess the hepatotoxic effect of an active AstraZeneca drug candidate illustrating its applicability in drug safety assessment beyond testing tool compounds. Finally, we have demonstrated that this approach could be adapted to Organ-Chips of different shapes and sizes through application to a Kidney-Chip.


Subject(s)
Lab-On-A-Chip Devices , Optical Imaging/instrumentation , Animals , Automation , Drug Evaluation, Preclinical , Humans , Kidney/diagnostic imaging , Kidney/drug effects , Liver/diagnostic imaging , Liver/drug effects , Rats
10.
Annu Rev Pharmacol Toxicol ; 58: 65-82, 2018 01 06.
Article in English | MEDLINE | ID: mdl-29029591

ABSTRACT

Enhancing the early detection of new therapies that are likely to carry a safety liability in the context of the intended patient population would provide a major advance in drug discovery. Microphysiological systems (MPS) technology offers an opportunity to support enhanced preclinical to clinical translation through the generation of higher-quality preclinical physiological data. In this review, we highlight this technological opportunity by focusing on key target organs associated with drug safety and metabolism. By focusing on MPS models that have been developed for these organs, alongside other relevant in vitro models, we review the current state of the art and the challenges that still need to be overcome to ensure application of this technology in enhancing drug discovery.


Subject(s)
Drug Discovery/methods , Pharmaceutical Preparations/chemistry , Animals , Drug Evaluation, Preclinical/methods , Humans
11.
ACS Nano ; 11(1): 730-741, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28051852

ABSTRACT

We present a method to induce cell directional behavior using slanted nanocilia arrays. NIH-3T3 fibroblasts demonstrated bidirectional polarization in a rectangular arrangement on vertical nanocilia arrays and exhibited a transition from a bidirectional to a unidirectional polarization pattern when the angle of the nanocilia was decreased from 90° to 30°. The slanted nanocilia guided and facilitated spreading by allowing the cells to contact the sidewalls of the nanocilia, and the directional migration of the cells opposed the direction of the slant due to the anisotropic bending stiffness of the slanted nanocilia. Although the cells recognized the underlying anisotropic geometry when the nanocilia were coated with fibronectin, collagen type I, and Matrigel, the cells lost their directionality when the nanocilia were coated with poly-d-lysine and poly-l-lysine. Furthermore, although the cells recognized geometrical anisotropy on fibronectin coatings, pharmacological perturbation of PI3K-Rac signaling hindered the directional elongation of the cells on both the slanted and vertical nanocilia. Furthermore, myosin light chain II was required for the cells to obtain polarized morphologies. These results indicated that the slanted nanocilia array provided anisotropic contact guidance cues to the interacting cells. The polarization of cells was controlled through two steps: the recognition of underlying geometrical anisotropy and the subsequent directional spreading according to the guidance cues.

12.
Sci Rep ; 5: 11542, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26100219

ABSTRACT

Tympanic membrane (TM) perforation, in particular chronic otitis media, is one of the most common clinical problems in the world and can present with sensorineural healing loss. Here, we explored an approach for TM regeneration where the latent progenitor or stem cells within TM epithelial layers may play an important regulatory role. We showed that potential TM stem cells present highly positive staining for epithelial stem cell markers in all areas of normal TM tissue. Additionally, they are present at high levels in perforated TMs, especially in proximity to the holes, regardless of acute or chronic status, suggesting that TM stem cells may be a potential factor for TM regeneration. Our study suggests that latent TM stem cells could be potential regulators of regeneration, which provides a new insight into this clinically important process and a potential target for new therapies for chronic otitis media and other eardrum injuries.


Subject(s)
Regeneration , Stem Cells/cytology , Tympanic Membrane/physiopathology , Animals , Biomarkers/metabolism , Cell Separation , Female , Integrin beta1/metabolism , Keratin-19/metabolism , Rats, Sprague-Dawley , Stem Cell Transplantation , Tympanic Membrane Perforation/pathology , Tympanic Membrane Perforation/therapy
13.
Annu Rev Pathol ; 10: 195-262, 2015.
Article in English | MEDLINE | ID: mdl-25621660

ABSTRACT

The ultimate goal of most biomedical research is to gain greater insight into mechanisms of human disease or to develop new and improved therapies or diagnostics. Although great advances have been made in terms of developing disease models in animals, such as transgenic mice, many of these models fail to faithfully recapitulate the human condition. In addition, it is difficult to identify critical cellular and molecular contributors to disease or to vary them independently in whole-animal models. This challenge has attracted the interest of engineers, who have begun to collaborate with biologists to leverage recent advances in tissue engineering and microfabrication to develop novel in vitro models of disease. As these models are synthetic systems, specific molecular factors and individual cell types, including parenchymal cells, vascular cells, and immune cells, can be varied independently while simultaneously measuring system-level responses in real time. In this article, we provide some examples of these efforts, including engineered models of diseases of the heart, lung, intestine, liver, kidney, cartilage, skin and vascular, endocrine, musculoskeletal, and nervous systems, as well as models of infectious diseases and cancer. We also describe how engineered in vitro models can be combined with human inducible pluripotent stem cells to enable new insights into a broad variety of disease mechanisms, as well as provide a test bed for screening new therapies.


Subject(s)
Models, Biological , Pathology/methods , Animals , Disease , Humans , In Vitro Techniques
14.
J Biomed Mater Res B Appl Biomater ; 103(6): 1320-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25377936

ABSTRACT

In recent years, cell chip-based platforms have begun to show promise as a means of corroborating the findings of in vivo animal tests for cytotoxicity, and perhaps in the future partially replacing the need for such animal models. In contrast to the conventional culture methods, micro- and nanofabrication techniques can be utilized to provide a set of mechanostimulatory signals to the cells that mimic the context of extracellular matrix (ECM) of the tissue in which a particular cell line resides. Here, we report periodic lateral topographic striations, with a pitch ranging approximately from 200 to 800 nm with an intention to mimic a common geometry of fibrils in the ECM such as collagen or elastin, as a platform for investigating anticancer drug-induced cytotoxicity in stem cells. The ECM cues could facilitate perimeter, elongation, and gap junction formation of mesenchymal stem cells (MSCs), which eventually influenced the fate of cells in terms of death and survival against the common chemotherapeutic agent cisplatin. Interestingly, the appropriate inhibition of gap junctions of MSCs on the ECM mimicking substrates could prevent the cisplatin-induced cytotoxicity through the inhibition of the cisplatin-induced 'death signal communication' as compared to that on the flat substrates. Our results imply that nanoscale topography is an important consideration for chip-based cytotoxicity assays, which uniquely enable the consideration and rational design of ECM-like topographic features, and furthermore, that the natural topography of the ECM in the context of stem cell niches may serve as an important indicator for chemotherapeutic agent sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Biomimetic Materials/chemistry , Extracellular Matrix/chemistry , Mesenchymal Stem Cells/metabolism , Cell Survival/drug effects , Gap Junctions/metabolism , HeLa Cells , Humans , Mesenchymal Stem Cells/cytology
15.
Integr Biol (Camb) ; 5(9): 1119-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23644926

ABSTRACT

Kidney toxicity is one of the most frequent adverse events reported during drug development. The lack of accurate predictive cell culture models and the unreliability of animal studies have created a need for better approaches to recapitulate kidney function in vitro. Here, we describe a microfluidic device lined by living human kidney epithelial cells exposed to fluidic flow that mimics key functions of the human kidney proximal tubule. Primary kidney epithelial cells isolated from human proximal tubule are cultured on the upper surface of an extracellular matrix-coated, porous, polyester membrane that splits the main channel of the device into two adjacent channels, thereby creating an apical 'luminal' channel and a basal 'interstitial' space. Exposure of the epithelial monolayer to an apical fluid shear stress (0.2 dyne cm(-2)) that mimics that found in living kidney tubules results in enhanced epithelial cell polarization and primary cilia formation compared to traditional Transwell culture systems. The cells also exhibited significantly greater albumin transport, glucose reabsorption, and brush border alkaline phosphatase activity. Importantly, cisplatin toxicity and Pgp efflux transporter activity measured on-chip more closely mimic the in vivo responses than results obtained with cells maintained under conventional culture conditions. While past studies have analyzed kidney tubular cells cultured under flow conditions in vitro, this is the first report of a toxicity study using primary human kidney proximal tubular epithelial cells in a microfluidic 'organ-on-a-chip' microdevice. The in vivo-like pathophysiology observed in this system suggests that it might serve as a useful tool for evaluating human-relevant renal toxicity in preclinical safety studies.


Subject(s)
Drug Evaluation, Preclinical/methods , Kidney Tubules, Proximal/metabolism , Microfluidic Analytical Techniques/methods , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Albumins/metabolism , Alkaline Phosphatase/metabolism , Biological Transport , Cisplatin/pharmacokinetics , Cisplatin/toxicity , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Glucose/metabolism , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/ultrastructure , Microscopy, Fluorescence
16.
Integr Biol (Camb) ; 3(2): 134-41, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21079870

ABSTRACT

In vivo, renal tubular epithelial cells are exposed to luminal fluid shear stress (FSS) and a transepithelial osmotic gradient. In this study, we used a simple collecting-duct-on-a-chip to investigate the role of an altered luminal microenvironment in the translocation of aquaporin-2 (AQP2) and the reorganization of actin cytoskeleton (F-actin) in primary cultured inner medullary collecting duct (IMCD) cells of rat kidney. Immunocytochemistry demonstrated that 3 h of exposure to luminal FSS at 1 dyn cm(-2) was sufficient to induce depolymerization of F-actin in those cells. We observed full actin depolymerization after 5 h exposure and substantial re-polymerization within 2 h of removing the luminal FSS, suggesting that the process is reversible and the fluidic environment regulates the reorganization of intracellular F-actin. We demonstrate that several factors (i.e., luminal FSS, hormonal stimulation, transepithelial osmotic gradient) collectively exert a profound effect on the AQP2 trafficking in the collecting ducts, which is associated with actin cytoskeletal reorganization.


Subject(s)
Actins/metabolism , Aquaporin 2/metabolism , Cytoskeleton/metabolism , Kidney Tubules, Collecting/metabolism , Animals , Cell Line , Epithelial Cells/metabolism , Immunohistochemistry , Kidney Tubules, Collecting/cytology , Microfluidic Analytical Techniques/methods , Protein Transport , Rats , Stress, Mechanical
17.
Anal Chem ; 82(7): 3016-22, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20218573

ABSTRACT

We present a simple analytical method to measure adhesion of human umbilical vein endothelial cells (HUVECs) and calf pulmonary artery endothelial cells (CPAEs) using nanopatterned, biodegradable poly(lactic-co-glycolic acid) (PLGA) surfaces for potential applications to artificial tissue-engineered blood vessel. Various nanostructured PLGA surfaces (350 nm wide ridges/350 nm grooves, 350 nm ridges/700 nm grooves, 350 nm ridges/1750 nm grooves, 700 nm ridges/350 nm grooves, 1050 nm ridges/350 nm grooves, 1750 nm ridges/350 nm grooves) and flat (unpatterned) surfaces were fabricated on the bottom of polydimethylsiloxane (PDMS) microfluidic channel of 2 mm width and 60 microm height by using thermal imprinting and irreversible channel bonding. To measure adhesion strength of HUVECs and CPAEs, the cells were exposed to a range of shear stress (12, 40, and 80 dyn/cm(2)) within the channels for 20 min after a preculture for 3 days and the remaining cells were counted under each condition. The highest adhesion strength was found on the surface of 700 nm wide ridges, 350 nm wide grooves for both cell types. The enhanced adhesion on nanopatterned surfaces can be attributed to two aspects: (i) contact guidance along the line direction and (ii) clustered focal adhesions. In particular, the contact guidance induced cell alignment along the line directions, which in turn lowers wall shear stress applied to the cell surface, as supported by a simple hydrodynamic model based on cell morphology.


Subject(s)
Cell Adhesion , Endothelial Cells/cytology , Microfluidic Analytical Techniques/methods , Nanostructures/chemistry , Cells, Cultured , Dimethylpolysiloxanes/chemistry , Fluorescent Dyes/chemistry , Humans , Lactic Acid/chemistry , Microscopy, Electron, Scanning , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Surface Properties
18.
PLoS One ; 5(12): e15966, 2010 Dec 30.
Article in English | MEDLINE | ID: mdl-21209862

ABSTRACT

BACKGROUND: The process of neurite outgrowth is the initial step in producing the neuronal processes that wire the brain. Current models about neurite outgrowth have been derived from classic two-dimensional (2D) cell culture systems, which do not recapitulate the topographical cues that are present in the extracellular matrix (ECM) in vivo. Here, we explore how ECM nanotopography influences neurite outgrowth. METHODOLOGY/PRINCIPAL FINDINGS: We show that, when the ECM protein laminin is presented on a line pattern with nanometric size features, it leads to orientation of neurite outgrowth along the line pattern. This is also coupled with a robust increase in neurite length. The sensing mechanism that allows neurite orientation occurs through a highly stereotypical growth cone behavior involving two filopodia populations. Non-aligned filopodia on the distal part of the growth cone scan the pattern in a lateral back and forth motion and are highly unstable. Filopodia at the growth cone tip align with the line substrate, are stabilized by an F-actin rich cytoskeleton and enable steady neurite extension. This stabilization event most likely occurs by integration of signals emanating from non-aligned and aligned filopodia which sense different extent of adhesion surface on the line pattern. In contrast, on the 2D substrate only unstable filopodia are observed at the growth cone, leading to frequent neurite collapse events and less efficient outgrowth. CONCLUSIONS/SIGNIFICANCE: We propose that a constant crosstalk between both filopodia populations allows stochastic sensing of nanotopographical ECM cues, leading to oriented and steady neurite outgrowth. Our work provides insight in how neuronal growth cones can sense geometric ECM cues. This has not been accessible previously using routine 2D culture systems.


Subject(s)
Extracellular Matrix/metabolism , Growth Cones/metabolism , Laminin/chemistry , Neurites/metabolism , Brain/physiology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Humans , Indoles/chemistry , Laminin/metabolism , Microscopy, Phase-Contrast/methods , Neurons/metabolism , Pseudopodia/metabolism , Tubulin/chemistry
19.
Lab Chip ; 10(1): 36-42, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20024048

ABSTRACT

We have developed a simple multi-layer microfluidic device by integrating a polydimethyl siloxane (PDMS) microfluidic channel and a porous membrane substrate to culture and analyze the renal tubular cells. As a model cell type, primary rat inner medullary collecting duct (IMCD) cells were cultured inside the channel. To generate in vivo-like tubular environments for the cells, a fluidic shear stress of 1 dyn/cm(2) was applied for 5 hours, allowing for optimal fluidic conditions for the cultured cells, as verified by enhanced cell polarization, cytoskeletal reorganization, and molecular transport by hormonal stimulations. These results suggest that the microfluidic device presented here is useful for resembling an in vivo renal tubule system and has potential applications in drug screening and advanced tissue engineering.


Subject(s)
Cell Culture Techniques/methods , Kidney Tubules, Collecting/cytology , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Animals , Cell Culture Techniques/instrumentation , Cells, Cultured , Dimethylpolysiloxanes/chemistry , Equipment Design , Male , Membranes, Artificial , Microfluidic Analytical Techniques/instrumentation , Porosity , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...