Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Philip C. Samson is active.

Publication


Featured researches published by Philip C. Samson.


Biomicrofluidics | 2015

Recreating blood-brain barrier physiology and structure on chip: A novel neurovascular microfluidic bioreactor.

Jacquelyn A. Brown; Virginia Pensabene; Dmitry A. Markov; Vanessa Allwardt; M. Diana Neely; Mingjian Shi; Clayton M. Britt; Orlando S. Hoilett; Qing Yang; Bryson M. Brewer; Philip C. Samson; Lisa J. McCawley; James M. May; Donna J. Webb; Deyu Li; Aaron B. Bowman; Ronald S. Reiserer; John P. Wikswo

The blood-brain barrier (BBB) is a critical structure that serves as the gatekeeper between the central nervous system and the rest of the body. It is the responsibility of the BBB to facilitate the entry of required nutrients into the brain and to exclude potentially harmful compounds; however, this complex structure has remained difficult to model faithfully in vitro. Accurate in vitro models are necessary for understanding how the BBB forms and functions, as well as for evaluating drug and toxin penetration across the barrier. Many previous models have failed to support all the cell types involved in the BBB formation and/or lacked the flow-created shear forces needed for mature tight junction formation. To address these issues and to help establish a more faithful in vitro model of the BBB, we have designed and fabricated a microfluidic device that is comprised of both a vascular chamber and a brain chamber separated by a porous membrane. This design allows for cell-to-cell communication between endothelial cells, astrocytes, and pericytes and independent perfusion of both compartments separated by the membrane. This NeuroVascular Unit (NVU) represents approximately one-millionth of the human brain, and hence, has sufficient cell mass to support a breadth of analytical measurements. The NVU has been validated with both fluorescein isothiocyanate (FITC)-dextran diffusion and transendothelial electrical resistance. The NVU has enabled in vitro modeling of the BBB using all human cell types and sampling effluent from both sides of the barrier.


IEEE Transactions on Biomedical Engineering | 2013

Engineering Challenges for Instrumenting and Controlling Integrated Organ-on-Chip Systems

John P. Wikswo; Frank E. Block; David E. Cliffel; Cody R. Goodwin; Christina C. Marasco; Dmitry A. Markov; David L. McLean; John A. McLean; Jennifer R. McKenzie; Ronald S. Reiserer; Philip C. Samson; David K. Schaffer; Kevin T. Seale; Stacy D. Sherrod

The sophistication and success of recently reported microfabricated organs-on-chips and human organ constructs have made it possible to design scaled and interconnected organ systems that may significantly augment the current drug development pipeline and lead to advances in systems biology. Physiologically realistic live microHuman (μHu) and milliHuman (mHu) systems operating for weeks to months present exciting and important engineering challenges such as determining the appropriate size for each organ to ensure appropriate relative organ functional activity, achieving appropriate cell density, providing the requisite universal perfusion media, sensing the breadth of physiological responses, and maintaining stable control of the entire system, while maintaining fluid scaling that consists of ~5 mL for the mHu and ~5 μL for the μHu. We believe that successful mHu and μHu systems for drug development and systems biology will require low-volume microdevices that support chemical signaling, microfabricated pumps, valves and microformulators, automated optical microscopy, electrochemical sensors for rapid metabolic assessment, ion mobility-mass spectrometry for real-time molecular analysis, advanced bioinformatics, and machine learning algorithms for automated model inference and integrated electronic control. Toward this goal, we are building functional prototype components and are working toward top-down system integration.


Journal of Neuroinflammation | 2016

Metabolic consequences of inflammatory disruption of the blood-brain barrier in an organ-on-chip model of the human neurovascular unit

Jacquelyn A. Brown; Simona G. Codreanu; Mingjian Shi; Stacy D. Sherrod; Dmitry A. Markov; M. Diana Neely; Clayton M. Britt; Orlando S. Hoilett; Ronald S. Reiserer; Philip C. Samson; Lisa J. McCawley; Donna J. Webb; Aaron B. Bowman; John A. McLean; John P. Wikswo

BackgroundUnderstanding blood-brain barrier responses to inflammatory stimulation (such as lipopolysaccharide mimicking a systemic infection or a cytokine cocktail that could be the result of local or systemic inflammation) is essential to understanding the effect of inflammatory stimulation on the brain. It is through the filter of the blood-brain barrier that the brain responds to outside influences, and the blood-brain barrier is a critical point of failure in neuroinflammation. It is important to note that this interaction is not a static response, but one that evolves over time. While current models have provided invaluable information regarding the interaction between cytokine stimulation, the blood-brain barrier, and the brain, these approaches—whether in vivo or in vitro—have often been only snapshots of this complex web of interactions.MethodsWe utilize new advances in microfluidics, organs-on-chips, and metabolomics to examine the complex relationship of inflammation and its effects on blood-brain barrier function ex vivo and the metabolic consequences of these responses and repair mechanisms. In this study, we pair a novel dual-chamber, organ-on-chip microfluidic device, the NeuroVascular Unit, with small-volume cytokine detection and mass spectrometry analysis to investigate how the blood-brain barrier responds to two different but overlapping drivers of neuroinflammation, lipopolysaccharide and a cytokine cocktail of IL-1β, TNF-α, and MCP1,2.ResultsIn this study, we show that (1) during initial exposure to lipopolysaccharide, the blood-brain barrier is compromised as expected, with increased diffusion and reduced presence of tight junctions, but that over time, the barrier is capable of at least partial recovery; (2) a cytokine cocktail also contributes to a loss of barrier function; (3) from this time-dependent cytokine activation, metabolic signature profiles can be obtained for both the brain and vascular sides of the blood-brain barrier model; and (4) collectively, we can use metabolite analysis to identify critical pathways in inflammatory response.ConclusionsTaken together, these findings present new data that allow us to study the initial effects of inflammatory stimulation on blood-brain barrier disruption, cytokine activation, and metabolic pathway changes that drive the response and recovery of the barrier during continued inflammatory exposure.


Journal of Physical Chemistry A | 2011

Single-nanocrystal spectroscopy of white-light-emitting CdSe nanocrystals.

Albert D. Dukes; Philip C. Samson; Joseph D. Keene; Lloyd M. Davis; John P. Wikswo; Sandra J. Rosenthal

We report the observation of broad-spectrum fluorescence from single CdSe nanocrystals. Individual semiconductor nanocrystals typically have a narrower emission spectrum than that of an ensemble. However, our experiments show that the ensemble white-light emission observed in ultrasmall CdSe nanocrystals is the result of many single CdSe nanocrystals, each emitting over the entire visible spectrum. These results indicate that each white-light-emitting CdSe nanocrystal contains all the trap states that give rise to the observed white-light emission.


Integrative Biology | 2010

Open access microfluidic device for the study of cell migration during chemotaxis

Dawit Jowhar; Gus A. Wright; Philip C. Samson; John P. Wikswo; Chris Janetopoulos

Cells sense and interpret chemical gradients, and respond by localized responses that lead to directed migration. An open microfluidic device (OMD) was developed to provide quantitative information on both the gradient and morphological changes that occurred as cells crawled through various microfabricated channels. This device overcame problems that many current devices have been plagued with, such as complicated cell loading, media evaporation and channel blockage by air bubbles. We used a micropipette to set up stable gradients formed by passive diffusion and thus avoided confounding cellular responses produced by shear forces. Two versions of the OMD are reported here: one device that has channels with widths of 6, 8, 10 and 12 μm, while the other has two large 100 μm channels to minimize cellular interaction with lateral walls. These experiments compared the migration rates and qualitative behavior of Dictyostelium discoideum cells responding to measurable cAMP and folic acid gradients in small and large channels. We report on the influence that polarity has on a cells ability to migrate when confined in a channel. Polarized cells that migrated to cAMP were significantly faster than the unpolarized cells that crawled toward folic acid. Unpolarized cells in wide channels often strayed off course, yet migrated faster than unpolarized cells in confined channels. Cells in channels farthest from the micropipette migrated through the channels at rates similar to cells in channels with higher concentrations, suggesting that cell speed was independent of mean concentration. Lastly, it was found that the polarized cells could easily change migration direction even when only the leading edge of the cell was exposed to a lateral gradient.


IEEE Transactions on Biomedical Engineering | 1983

A Low-Noise Low Input Impedance Amplifier for Magnetic Measurements of Nerve Action Currents

John P. Wikswo; Philip C. Samson; Robin P. Giffard

We have developed a low-noise solid-state current-to-voltage amplifier to measure the magnetic fields produced by nerve action currents. The amplifier senses the EMF induced in a toroidal, ferrite-core pickup coil surrounding the nerve by the circumferential magnetic field associated with the axial nerve action currents which propagate past the toroid. In order to obtain low input voltage noise, the input stage of the amplifier consists of 20 preamplifiers in parallel, each containing a discrete bipolar transistor pair. The complete amplifier has an input impedance of 1.33 ¿and a minimum noise figure of 15 dB at 100 Hz for a source resistance of 50 ¿. The amplifier has sufficient sensitivity to detect the magnetic field of the action potential propagating along a single giant axon.


Analytical Chemistry | 2013

Dynamic Dosing Assay Relating Real-Time Respiration Responses of Staphylococcus aureus Biofilms to Changing Microchemical Conditions

Jinzi Deng; Adit Dhummakupt; Philip C. Samson; John P. Wikswo; Leslie M. Shor

Bacterial biofilms are a metabolically heterogeneous community of bacteria distributed in an extracellular matrix comprised primarily of hydrated polysaccharides. Effective inhibitory concentrations measured under planktonic conditions are not applicable to biofilms, and inhibition concentrations measured for biofilms vary widely. Here, we introduce a novel microfluidic approach for screening respiration inhibition of bacteria in a biofilm array morphology. The device geometry and operating conditions allow antimicrobial concentration and flux to vary systematically and predictably with space and time. One experiment can screen biofilm respiratory responses to many different antimicrobial concentrations and dosing rates in parallel. To validate the assay, onset of respiration inhibition following NaN₃ exposure is determined optically using an O₂-sensing thin film. Onset of respiration inhibition obeys a clear and reproducible pattern based on time for diffusive transport of the respiration inhibitor to each biofilm in the array. This approach can be used for high-throughput screening of antimicrobial effectiveness as a function of microbial characteristics, antimicrobial properties, or antimicrobial dosing rates. The approach may also be useful in better understanding acquired antimicrobial resistance or for screening antimicrobial combinations.


Biomedical Microdevices | 2010

Tape underlayment rotary-node (TURN) valves for simple on-chip microfluidic flow control

Dmitry A. Markov; Steven Manuel; Leslie M. Shor; Susan R. Opalenik; John P. Wikswo; Philip C. Samson

We describe a simple and reliable fabrication method for producing multiple, manually activated microfluidic control valves in polydimethylsiloxane (PDMS) devices. These screwdriver-actuated valves reside directly on the microfluidic chip and can provide both simple on/off operation as well as graded control of fluid flow. The fabrication procedure can be easily implemented in any soft lithography lab and requires only two specialized tools—a hot-glue gun and a machined brass mold. To facilitate use in multi-valve fluidic systems, the mold is designed to produce a linear tape that contains a series of plastic rotary nodes with small stainless steel machine screws that form individual valves which can be easily separated for applications when only single valves are required. The tape and its valves are placed on the surface of a partially cured thin PDMS microchannel device while the PDMS is still on the soft-lithographic master, with the master providing alignment marks for the tape. The tape is permanently affixed to the microchannel device by pouring an over-layer of PDMS, to form a full-thickness device with the tape as an enclosed underlayment. The advantages of these Tape Underlayment Rotary-Node (TURN) valves include parallel fabrication of multiple valves, low risk of damaging a microfluidic device during valve installation, high torque, elimination of stripped threads, the capabilities of TURN hydraulic actuators, and facile customization of TURN molds. We have utilized these valves to control microfluidic flow, to control the onset of molecular diffusion, and to manipulate channel connectivity. Practical applications of TURN valves include control of loading and chemokine release in chemotaxis assay devices, flow in microfluidic bioreactors, and channel connectivity in microfluidic devices intended to study competition and predator/prey relationships among microbes.


Journal of Visualized Experiments | 2010

Window on a microworld: simple microfluidic systems for studying microbial transport in porous media.

Dmitry A. Markov; Philip C. Samson; David K. Schaffer; Adit Dhummakupt; John P. Wikswo; Leslie M. Shor

Microbial growth and transport in porous media have important implications for the quality of groundwater and surface water, the recycling of nutrients in the environment, as well as directly for the transmission of pathogens to drinking water supplies. Natural porous media is composed of an intricate physical topology, varied surface chemistries, dynamic gradients of nutrients and electron acceptors, and a patchy distribution of microbes. These features vary substantially over a length scale of microns, making the results of macro-scale investigations of microbial transport difficult to interpret, and the validation of mechanistic models challenging. Here we demonstrate how simple microfluidic devices can be used to visualize microbial interactions with micro-structured habitats, to identify key processes influencing the observed phenomena, and to systematically validate predictive models. Simple, easy-to-use flow cells were constructed out of the transparent, biocompatible and oxygen-permeable material poly(dimethyl siloxane). Standard methods of photolithography were used to make micro-structured masters, and replica molding was used to cast micro-structured flow cells from the masters. The physical design of the flow cell chamber is adaptable to the experimental requirements: microchannels can vary from simple linear connections to complex topologies with feature sizes as small as 2 microm. Our modular EcoChip flow cell array features dozens of identical chambers and flow control by a gravity-driven flow module. We demonstrate that through use of EcoChip devices, physical structures and pressure heads can be held constant or varied systematically while the influence of surface chemistry, fluid properties, or the characteristics of the microbial population is investigated. Through transport experiments using a non-pathogenic, green fluorescent protein-expressing Vibrio bacterial strain, we illustrate the importance of habitat structure, flow conditions, and inoculums size on fundamental transport phenomena, and with real-time particle-scale observations, demonstrate that microfluidics offer a compelling view of a hidden world.


Proceedings of SPIE, the International Society for Optical Engineering | 2008

Electrokinetic Delivery of Single Fluorescent Biomolecules in Fluidic Nanochannels

Lloyd M. Davis; Brian K. Canfield; Xiaoxuan Li; William H. Hofmeister; Guoqing Shen; Isaac P. Lescano-Mendoza; Bruce W. Bomar; John P. Wikswo; Dmitry A. Markov; Philip C. Samson; Claus Daniel; Zbigniew Sikorski; William N. Robinson

We describe the fabrication of sub-100-nanometer-sized channels in a fused silica lab-on-a-chip device and experiments that demonstrate detection of single fluorescently labeled proteins in buffer solution within the device with high signal and low background. The fluorescent biomolecules are transported along the length of the nanochannels by electrophoresis and/or electro-osmosis until they pass into a two-focus laser irradiation zone. Pulse-interleaved excitation and time-resolved single-photon detection with maximum-likelihood analysis enables the location of the biomolecule to be determined. Diffusional transport of the molecules is found to be slowed within the nanochannel, and this facilitates fluidic trapping and/or prolonged measurements on individual biomolecules. Our goal is to actively control the fluidic transport to achieve rapid delivery of each new biomolecule to the sensing zone, following the completion of measurements, or the photobleaching of the prior molecule. We have used computer simulations that include photophysical effects such as triplet crossing and photobleaching of the labels to design control algorithms, which are being implemented in a custom field-programmable-gate-array circuit for the active fluidic control.

Collaboration


Dive into the Philip C. Samson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leslie M. Shor

University of Connecticut

View shared research outputs
Researchain Logo
Decentralizing Knowledge