WO2004038368A2 - Dispositif et procede pour surveiller la migration des leucocytes - Google Patents

Dispositif et procede pour surveiller la migration des leucocytes Download PDF

Info

Publication number
WO2004038368A2
WO2004038368A2 PCT/US2003/033177 US0333177W WO2004038368A2 WO 2004038368 A2 WO2004038368 A2 WO 2004038368A2 US 0333177 W US0333177 W US 0333177W WO 2004038368 A2 WO2004038368 A2 WO 2004038368A2
Authority
WO
WIPO (PCT)
Prior art keywords
leukocytes
channel
well
mediator
leukocyte migration
Prior art date
Application number
PCT/US2003/033177
Other languages
English (en)
Other versions
WO2004038368A3 (fr
Inventor
Gregory L. Kirk
Enoch Kim
Emanuele Ostuni
Olivier Schueller
Paul Sweetnam
Original Assignee
Surface Logix, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Surface Logix, Inc. filed Critical Surface Logix, Inc.
Priority to EP03774884A priority Critical patent/EP1558076A4/fr
Priority to AU2003282950A priority patent/AU2003282950A1/en
Priority to CA002503203A priority patent/CA2503203A1/fr
Priority to JP2004546913A priority patent/JP2006503581A/ja
Publication of WO2004038368A2 publication Critical patent/WO2004038368A2/fr
Publication of WO2004038368A3 publication Critical patent/WO2004038368A3/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5029Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on cell motility
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5025Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures for parallel transport of multiple samples
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y15/00Nanotechnology for interacting, sensing or actuating, e.g. quantum dots as markers in protein assays or molecular motors
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/06Auxiliary integrated devices, integrated components
    • B01L2300/0681Filter
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0809Geometry, shape and general structure rectangular shaped
    • B01L2300/0829Multi-well plates; Microtitration plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip

Definitions

  • the present invention relates to devices and methods for monitoring the interaction of a cell or group of cells with a substratum.
  • the present invention relates to devices and methods for monitoring leukocyte migration.
  • the present invention also relates generally to biological assays performed in gradients formed in microfluidic systems.
  • the inflammatory response is an attempt by the body to restore and maintain homeostasis after infection or injury, and is an integral part of body defense. Most of the body defense elements are located in the blood and inflammation is the means by which these elements leave the blood and enter the tissue around the injured or infected site. The primary objective of inflammation is to localize and eradicate the source of injury or infection and repair tissue surrounding the site of injury or infection.
  • phagocytes such as mast cells in the damaged tissue release a variety of cytokines and inflammatory mediators, such as histamines, leukotrienes, bradykinins, and prostaglandins. These inflammatory mediators reversibly open the junctional zones between the thin delicate cells of the inner surface of the blood vessels, known as the endothelium, that surround the damaged tissue.
  • the inflammatory mediators also cause increased blood vessel permeability and decreased blood flow velocity.
  • leukocytes which normally travel in the center of the blood vessel, move out to the periphery of the inner surface of the blood vessel to interact with the endothelium.
  • the cytokines and inflammatory mediators released by the phagocytes also induce the expression of adhesion molecules on the surface of the endothelium, resulting in an "activated" endothelium.
  • the first contact of leukocytes with the activated endothelium is known as "capture” and is thought to involve the adhesion molecules P-selectin and L- selectin, which are upregulated on endothelium after exposure to inflammatory mediators.
  • P-selectin and L-selectin belong to a family of adhesion molecules called selectins.
  • Selectins are a group of monomeric, integral membrane glycoproteins expressed on the surface of activated endothelium and leukocytes. Selectins contain an N-terminal extracellular domain with structural homology to calcium-dependent lectins, followed by a domain homologous to epidermal growth factor, and nine consensus repeats (CR) similar to sequences found in complement regulatory proteins.
  • P-selectin also known as CD62P, GMP-140, and PADGEM
  • E-selectin also known as ELAM-1
  • L-selectin also known as LECAM-1, LAM-1, Mel-14 antigen, g ⁇ 90 mel , and Leu8/TQ-1 antigen
  • All three selectins are thought to bind to selectin binding ligands, at least in part through a carbohydrate component.
  • PSGL-1 main leukocyte ligand P-selectin glycoprotein ligand-1
  • Other ligands of P-selectin include CD24 and yet uncharacterized ligands.
  • the structure of functional PSGL-1 includes a sialyl-Lewis x component.
  • L-selectin is thought to bind to its ligand on endothelial cells. L-selectin interacts with three known counter receptors or ligands, MAdCAM-1, GlyCAM-1, and CD34, although the precise ligand or counter receptor involved in capture is unknown.
  • leukocytes Once leukocytes are captured, they may transiently adhere to the endothelium and begin to "roll” along the endothelium.
  • rolling refers to the literal rolling of leukocytes along the activated endothelium in the presence of fluid drag forces arising from the relative movement between the endothelium and the leukocytes. Rolling is thought to involve P-selectin, L-selectin, and E-selectin. Bonds between P-selectin and PSGL-1 are thought to primarily mediate the "rolling" of leukocytes across the endothelium.
  • Proinflarnmatory cytokines such as interleukin-1 (LL-1), and tumor necrosis factor- ⁇ (TNF- ⁇ ) produced by cells at the injured or infected site stimulate the endothelium to produce chemokines such as interleukin-8 (IL-8) and integrin binding ligands such as intercellular adhesion molecules (ICAMs) and vascular cell adhesion molecules (VCAMs) on the the surface of the endothelial cells opposite the basal lamina.
  • chemokines are held on the surface of the endothelial cells opposite the basal lamina where the chemokines interact with chemokine receptors on the surface of the rolling leukocytes.
  • Integrins are a family of heterodimeric transmembrane glycoproteins that attach cells to extracellular matrix proteins of the basement membrane or to ligands on other cells. Integrins are composed of large ⁇ and small ⁇ subunits. Mammalian integrins form several subfamilies sharing common ⁇ subunits that associate with difference ⁇ subunits.
  • ⁇ 2 integrins include four different heterodimers: CDlla/CD18 (Lymphocyte Function- Associated Antigen- 1 (LFA-1)); CDllb/CD18 (Mac-1); CDl lc/CD18 (pl50,95), and CDlld/CD18.
  • LFA-1 Lymphocyte Function- Associated Antigen- 1
  • Mac-1 CDllb/CD18
  • CDl lc/CD18 pl50,95
  • CDlld/CD18 CDlld/CD18.
  • VLA-4 Very Late Antigen 4
  • CD49d/CD29 CD49d/CD29
  • ⁇ 4 ⁇ i Very Late Antigen 4
  • Transmigration is thought to be mediated by platelets, endothelial cell adhesion molecule- 1 (PECAM-1), junctional adhesion molecule (JAM), and possibly CD99, a transmembrane protein.
  • PECAM-1 endothelial cell adhesion molecule- 1
  • JAM junctional adhesion molecule
  • leukocytes themselves can promote tissue damage.
  • leukocytes can cause significant tissue damage by releasing toxic substances at the vascular wall or in uninjured tissue.
  • leukocytes may stick to the capillary wall or clump in venules to such a degree that the endothelium becomes lined with these cells.
  • Such a phenomenon referred to as "pavementing,” may be related to the development of arteriosclerosis and associated diseases.
  • ARDS adult respiratory distress syndrome
  • ischemia-reperfusion injury following myocardial infarction, shock, stroke, or organ transplantation
  • acute and chronic allograft rejection vasculitis
  • sepsis sepsis
  • rheumotoid arthritis inflammatory skin diseases.
  • one method involves plating a monolayer of isolated endothelial cells on the surface of microtiter plates, activating the cells with a chemoattractant and then placing labeled leukocytes in the plate.
  • a test agent such as an adhesion inhibitor, may be optionally added to the plate.
  • the number of leukocytes that remain adherent to the endothelial cell monolayer is then determined.
  • a significant disadvantage of this method is that the leukocytes are not exposed to the endothelial cells in the presence of shear flow and thus this method does not simulate physiological conditions in vivo.
  • Another method involves contacting a suspension of isolated leukocytes in a suitable medium with a human vascular tissue sample mounted on a microscope slide and then incubating the tissue with a cell suspension on a rotating table. The adhered cells are fixed and counted. Because cells are fixed, such a method precludes the observation of leukocyte migration in real time. In addition, such a method requires human vascular tissue, which can be difficult and costly to obtain.
  • Another method known in the art to study leukocyte migration involves a device consisting of two glass tubes called microslides, one microslide capable of being inserted into the other. The smaller microslide is inserted into the larger one to create a flow channel with a flat surface on which selected adhesion molecules are present.
  • This device consists of several different components that are bulky in size. As such, it requires extra handling and positioning, creating the risk of contaminating or damaging the endothelial monolayer. This device also requires the use of a large number of cells and consequently a large amount of reagents.
  • the present invention provides a system for monitoring leukocyte migration comprising a device including a housing defining a plurality of chambers therein.
  • Each of the plurality of chambers includes a first well region including at least one first well, a second well region including at least one second well, and a channel region including at least one channel connecting the first well region and the second well region with one another.
  • the system further includes a first fluid stream having a first concentration of a first substance and a second fluid stream having a second concentration of a second substance, wherein the first and second concentrations are different from one another and the first and second fluid streams are in fluid communication with at least one of the plurality of chambers.
  • the present invention also provides a method of monitoring leukocyte migration comprising disposing endothelial cells on a surface, passing a fluid along the surface under conditions of substantially laminar flow wherein the fluid comprises a concentration gradient of at least one substance. The concentration gradient is substantially perpendicular to the direction of flow. The method further includes exposing a sample comprising leukocytes to the surface, and observing the interaction between the leukocytes and the endothelial cells. [001 ] The present invention moreover provides a method of monitoring leukocyte migration comprising providing a device including a housing defining a plurality of chambers therein.
  • Each of the plurality of chambers includes a first well region including at least one first well, a second well region including at least one second well, and a channel region including at least one channel connecting the first well region and the second well region with one another.
  • the method further comprises disposing at least one leukocyte migration mediator, or endothelial cells in the at least one channel, delivering a sample comprising leukocytes to the at least one channel by laminar flow, and observing the interaction between the leukocytes and the at least one leukocyte migration mediator or the interaction between the leukocytes and the endothelial cells.
  • Figure 1 is a perspective view of an embodiment of a device adapted to be used in a method for monitoring leukocyte migration according to the present invention.
  • Figure 2 is a cross-sectional view of the device of Fig. 1 along lines II-LT.
  • Figure 3 is a top plan view of the device of Fig. 1.
  • Figure 4 is a top plan view of an alternative embodiment of a chamber defined in a housing of a device adapted to be used in a method for monitoring leukocyte migration according to the present invention.
  • Figure 5 is a top plan view of a plurality of chambers such as the chamber of Fig. 4 disposed in a predetermined relationship with respect to one another.
  • Figure 6 is a top, perspective view of an alternative embodiment of a device adapted to be used in a method for monitoring leukocyte migration according to the present invention, where the device displays the dimensions and pitch of a standard 96-well microtiter plate.
  • Figure 6A is a top enlarged view of an individual well of an alternative embodiment of the device according to the present invention.
  • Figure 7 is a bar graph comparing the velocity of shear flow under different cell suspension volumes.
  • Figure 8 is a graph comparing the number of cells rolling under different dilutions of P-selectin antibody.
  • Figure 9 is a graph and time-lapsed still photographs of cells rolling and adhering under different dilutions of P-selectin antibody.
  • Figure 10 is a graph and time-lapsed still photographs of cells rolling and adhering under different dilutions of E-selectin antibody.
  • Figure 11 is a graph and time-lapsed still photographs of cells adhering to endothelium in the presence of antibodies to E-selectin, P-selectin, and VCAM-1.
  • FIG. 12 depicts the results of an experiment involving the creation of a concentration gradient of TNF- ⁇ via lammar flow.
  • the TNF- ⁇ was delivered to a confluent "lawn" of endothelial cells.
  • the endothelial cells that were contacted by the TNF- ⁇ were activated and thus are able to bind the leukocytes.
  • Leukocytes were then delivered to the endothelial cells.
  • the leukocytes bound to the area of the endothelial cells that received high concentrations of TNF- ⁇ whereas those areas not exposed to TNF- ⁇ or exposed to very little TNF- ⁇ did not bind leukocytes.
  • Figure 13 depicts an exemplary microfluidic device for creating a laminar flow gradient.
  • leukocytes refers to granulocytes including neutrophils, eosinophils, basophils, monocytes, and lymphocytes including B cells and T cells and unless otherwise specified, platelets.
  • leukocytes includes leukocytes obtained from both normal blood samples and pathological blood samples.
  • leukocyte migration cascade refers to the cascade of sequential events involving a leukocyte's migration along the endothelium lining a blood vessel.
  • the leukocyte migration cascade includes the capture, rolling, arrest, and transmigration of a leukocyte on, along, or through the endothelium.
  • the term "leukocyte migration mediator” as used herein refers to any molecule that mediates the migration of leukocytes along the endothelium lining a blood vessel.
  • the term “mediates” as used in the context of a “leukocyte migration mediator” means influencing the migration of a leukocyte by, for example, binding to the ligand or counter-receptor of the leukocyte migration mediator.
  • the term “leukocyte migration mediator” refers to any molecule involved in the leukocyte migration cascade.
  • a leukocyte migration mediator includes a leukocyte capture mediator, a leukocyte rolling mediator, a leukocyte arrest mediator, a leukocyte transmigration mediator, or any combination thereof.
  • capture refers to a step in the leukocyte migration cascade characterized by the tethering or first contact of leukocyte with the endothelium of a blood vessel so that the motion of the leukocyte along the endothelium is temporarily delayed relative to the flow of fluid containing free flowing leukocytes.
  • the term "leukocyte capture mediator” as used herein refers to a leukocyte migration mediator that mediates the capture of a leukocyte on the endothelium of a blood vessel.
  • Non- limiting examples of leukocyte capture mediators are P-selectin and L-selectin binding ligands.
  • the term "capture mediator binding partner” refers to any ligand or counter-receptor that binds a leukocyte capture mediator.
  • Non-limiting examples of capture mediator binding partners are PSGL-1 and L-selectin.
  • the term "rolling” as used herein refers to a step in the leukocyte migration cascade, and is characterized by the rolling of a leukocyte along the endothelium of a blood vessel from receptor to receptor on the endothelium further characterized by leukocytes forming and breaking adhesive bonds with endothelial ligands or counter-receptors.
  • leukocyte rolling mediator refers to any leukocyte migration mediator that mediates the rolling of a leukocyte along the endothelium of a blood vessel.
  • leukocyte rolling mediators are P-selectin, E-selectin, and L-selectin binding ligands.
  • rolling mediator binding partner refers to any ligand or counter-receptor that binds to a leukocyte rolling mediator.
  • rolling mediator binding partners are PSGL-1, E-selectin binding ligand, and L-selectin.
  • arrest refers to a step in the leukocyte migration cascade characterized by the adherence of leukocytes to the endothelium of a blood vessel.
  • leukocyte arrest mediator refers to any leukocyte migration mediator that mediates the arrest of a leukocyte on the endothelium of a blood vessel.
  • arrest mediators are integrin binding ligands, such as ICAM-1, ICAM-2, and VCAM-1 that bind integrins expressed on the surface of leukocytes.
  • arrest mediator binding partner refers to any ligand or counter- receptor that binds to a leukocyte arrest mediator.
  • arrest mediator binding partners are integrins including LFA-1, Mac-1, pl50,95, VLA-4, and VLA-5.
  • transmigration refers to a step in the leukocyte migration cascade characterized by the exit of leukocytes from a blood vessel to surrounding tissue through passage between cells of the endothelium of the blood vessel.
  • leukocyte transmigration mediator refers to any leukocyte migration mediator that mediates the transmigration of a leukocyte through the endothelium of a blood vessel.
  • leukocyte transmigration mediators are PECAM-1 and JAM.
  • transmigration binding partner refers to any ligand or counter-receptor that binds to a leukocyte transmigration mediator.
  • physiological shear flow includes shear flow under normal and pathological conditions. Physiological shear flow rate under normal conditions is about 0.1 to about 20 dynes/cm. 2
  • test agent refers to any chemical molecule or compound to be tested in the present invention to determine its ability to interact with another chemical molecule or compound.
  • the test agent may be tested to determine whether it inhibits or promotes leukocyte migration by inhibiting or promoting capture, rolling, arrest, or transmigration.
  • pitch refers to the distance between respective vertical centerlines between adjacent wells in the test orientation of the device.
  • well region refers to a region that comprises one or a plurality of wells.
  • well as used herein is meant to indicate any cavity that is able to receive a fluid therein.
  • channel region refers to a region that comprises one or a plurality of channels therein, while “channel” refers to any passageway.
  • conformal contact is meant to designate a substantially fluid-tight, form-fitting contact with a planar or non-planar surface
  • reversible conformal contact is meant to designate a conformal contact that may be interrupted without compromising a structural integrity of the members making the conformal contact.
  • test orientation of the device is meant to refer to a spatial orientation of the device during the monitoring of leukocyte migration and the terms “top,” “bottom,” “upper” and “side” are defined relative to the test orientation of the device.
  • the test orientation of the device for use in a method of monitoring leukocyte migration contemplates the orientation of the device such that a migration path along the channel region of any cells occurs in a substantially horizontal plane.
  • the test orientation of the device for use in monitoring leukocyte migration contemplates the orientation of the device such that a migration path along the channel region of any cells occurs in a substantially vertical plane.
  • the present invention generally provides devices and methods for in vitro monitoring the interaction of cells with a substratum.
  • cell types that may be monitored by the devices and methods of the present invention include leukocytes, red blood cells, platelets, non-blood cells, and tumor cells.
  • types of substratum that may interact with the cells include the endothelium, immobilized ligands, physisorbed adhesion and rolling molecules and basal lamina or basal lamina mimic.
  • the present invention provides a device and method for in vitro monitoring of leukocyte migration in the presence of shear flow in order to study the cascade of events involved in the inflammatory response in vivo.
  • the present invention also provides a device and method for the high-throughput screening of test agents that potentially target these events.
  • the present invention is directed to study and target the capture, rolling, arrest, and transmigration of a leukocyte on, along, or through the endothelium (such events collectively referred to as the "leukocyte migration cascade").
  • device 10 generally includes a housing 12 defining a plurality of chambers 14 therein, such as, by way of example, embodiments of chamber 14 depicted in Figs. 1-6.
  • Each chamber 14 includes: a first well region 16 including at least one first well 18 and a second well region 20 including at least one second well 22.
  • the chamber 14 further includes a channel region 24 including at least one channel 26 connecting the first well region 16 and the second well region 20 with one another.
  • the first well regions 16 and the second well regions 20 of the respective ones of the plurality of chambers are disposed relative to one another to match a pitch of a standard microtiter plate.
  • first well 18 and second well 22 are adapted to receive a sample comprising leukocytes and channel 26 is adapted to receive endothelial cells or leukocyte migration mediators thereon and is configured to support physiological shear flow therealong.
  • channel 26 contains endothelial cells disposed therein.
  • the endothelial cells may be activated prior to exposure to channel 26 or may have chemokines immobilized on the surface opposite the basal lamina therein upon exposure to channel 26.
  • Various cytophilic substances may be disposed in channel 26 to assist in the attachment of endothelial cells. Cytophilic substances are generally substances that have an affinity for cells or substances that promote cell attachment to the surface and include, for example, gelatin, collagen, fibronectin, fibrin, basal lamina, including, but not limited to MATRIGELTM or other hydrogels.
  • channel 26 includes a plurality of leukocyte migration mediators disposed therein.
  • the plurality of leukocyte migration mediators comprises at least one first leukocyte migration mediator and at least one second leukocyte migration mediator, wherein the at least one first and the at least one second leukocyte migration mediators are different from one another.
  • the leukocyte migration mediators are disposed in channel 26 so as to form a surface concentration gradient along a longitu dinal axis of chamber 14 in increasing concentration from first well 20 to second well 22.
  • channel 26 includes chemokines disposed therein to interact with chemokine receptors on the surface of rolling leukocytes.
  • the present invention also contemplates a method of monitoring leukocyte migration.
  • a sample including leukocytes is placed in first well 18 (or second well 22), the sample is allowed to flow along channel 26, the interaction (such interaction including a lack thereof) between the leukocytes and the endothelial cells is observed, and the sample including leukocytes is collected in second well 22 (or the first well 18) as the leukocytes exit channel 26.
  • a chemoattractant may be added to channel 26 to activate the endothelial cells before a sample containing leukocytes is added to first well 18 (or second well 22).
  • a test agent is placed in channel 26 and the interaction between the leukocytes and endothelial cells in the presence of the test agent is observed.
  • channel 26 contains a leukocyte migration mediator disposed therein
  • a sample including leukocytes is placed in first well 18 (or second well 22), the sample is allowed to flow along channel 26, the interaction (such interaction including a lack thereof) between the leukocytes and the leukocyte migration mediator is observed, and the sample including leukocytes is collected in second well 22 (or the first well 18) as the leukocytes exit channel 26.
  • a test agent is placed in channel 26 and the interaction between leukocytes and the leukocyte migration mediator in the presence of the test agent is observed.
  • device 10 may match the footprint of an industry standard microtiter plate
  • an advantage of device 10 is that device 10 may be used to conduct multiple assays simultaneously in the same device, and to high throughput screen various test agents.
  • the first well regions 16 and the second well regions 20 of the respective ones of the plurality of chambers 14 are disposed relative to one another to match a pitch of a standard microtiter plate.
  • the wells may be disposed relative to one another to match a pitch of one of a 24-well microtiter plate, a 96-well microtiter plate, a 384- well microtiter plate, 768-well microtiter plate and a 1536-well microtiter plate.
  • pitch P will be set to about 9 mm.
  • device 10 itself fits in the footprint of an industry standard microtiter plate. As such, device 10 preferably has the same outer dimensions and overall size of an industry standard microtiter plate.
  • device 10 comprises 48 chambers designed in the format of a standard 96-well plate, such that the respective wells 18/22 are disposed relative to one another to match a pitch of a standard 96-well microtiter plate with each well fitting in the space of each well of the plate.
  • 48 experiments can be conducted.
  • chambers 14 may be disposed relative to one another to match a pitch of a standard microtiter plate.
  • chambers 14 are sized so that a chamber 14 fits in the area normally required for a single well of a standard microtiter plate.
  • device 10 designed in the footprint of a 96-well microtiter plate configuration, has 96 chambers and therefore allows 96 experiments to be performed.
  • embodiments of device 10 would advantageously fit into existing infrastructures of fluid handling, storage, registration and detection.
  • Device 10 is also conducive to high throughput screening as it allows robotic fluid handling and automated detection and data analysis.
  • the use of robotic and automated systems also decreases the amount of time to prepare and perform the assays and analyze the results of the assays.
  • the use of device 10 decreases the occurrence of human error in preparing and performing assays and analyzing data.
  • the present invention also contemplates a method of screening a plurality of test agents.
  • the method of screening test agents includes providing a device comprising a housing defining a plurality of chambers.
  • Each chamber includes: a first well region including at least one first well; a second well region including at least one second well; and a channel region including at least one channel connecting the first well region and the second well region with one another.
  • the at least one channel includes at least one leukocyte migration mediator disposed therein.
  • the at least one channel includes endothelial cells disposed therein.
  • At least one of the plurality of chambers on the one hand, and the first well regions and the second well regions of respective ones of the plurality of chambers on the other hand are disposed relative to one another to match a pitch of a standard microtiter plate.
  • the method of screening test agents further includes providing leukocytes in each of the channels of respective ones of the plurality of chambers; placing at least one of a plurality of test agents in each of the channels of respective ones of the plurality of chambers; and observing the interaction between the leukocytes and the endothelial cells or the interaction between the leukocytes and the at least one leukocyte migration mediator in the presence of the test agents.
  • test agents it can be determined whether the test agents have an effect on the number of leukocytes that are captured, arrested, or have transmigrated as well as whether the test agents have an effect on velocity and number of leukocytes that roll along chamiel 26.
  • the test agent may include any desired biological, chemical, or electrical substance, including but not limited to, an inhibitor of leukocyte migration, a promoter of leukocyte migration, or any other therapeutic agent.
  • Further examples of test agents include proteins, nucleic acids, peptides, polypeptides, carbohydrates, lipids, hormones, enzymes, small molecules or pharmaceutical agents. This method is particularly useful in the area of drug discovery where a plurality of test agents may be screened in a single device 10.
  • each of the test agents is different from one another and a single test agent is placed in each channel.
  • a single test agent is placed in each channel.
  • two or more test agents of the plurality of test agents may be placed in each channel of each of the plurality of chambers.
  • the device of the present invention may also be used to monitor the steps of the leukocyte migration cascade under a normal or pathological physiological shear flow condition.
  • a normal physiological flow condition refers to the shear flow rate during a non-pathological state and is in the range of about 0.1 dynes/cm 2 to about 20 dynes/cm.
  • a pathological physiological flow condition refers to the shear flow rate during the inflammatory response and is generally varied depending on the disease state.
  • the physiological shear flow is preferably produced by hydrostatic pressure, or microcapillary action, the flow can be produced by any means known in the art.
  • physiological shear flow can be created by applying pressure through a vacuum adjacent to second well 22 or by applying pressure through a syringe pump adjacent to first well 18.
  • the shear flow may be manipulated by altering the dimensions of the channels or modifying the degree of pressure applied through the vacuum or syringe pump.
  • the leukocytes can be introduced into chamiel 16 in a pulsatile manner.
  • channel 26 may have endothelial cells disposed therein or leukocyte migration mediators disposed therein.
  • the endothelial cells may be disposed on any surface or surfaces of channel 26. In a preferred embodiment, the endothelial cells are on the bottom and side surfaces of channel 26.
  • the endothelial cells may be disposed uniformly in channel 26, may be disposed in discrete patches, or may be disposed along a concentration gradient such that the concentration of endothelial cells decreases from first well 18 to second well 22.
  • the endothelial cells may be grown on channel 26 in the presence or absence of shear flow.
  • channel 26 has endothelial cells disposed therein
  • several different assays may be performed to observe the interaction between leukocytes and the endothelial cells during the leukocyte migration cascade.
  • a sample containing leukocytes is introduced into channel 26 via first well 18 or second well 22.
  • the number of leukocytes rolling as well as the rolling velocity of the leukocytes may then be determined.
  • Assays measuring the inhibition of rolling may also be performed by adding to channel 26, for example, inhibitors that block the interaction between leukocytes and endothelial cells.
  • assays measuring the enhancement of rolling maybe performed by adding to channel 26, for example, promoters that promote the interaction between leukocytes and endothelial cells.
  • a test agent could also be added to channel 26 to determine the effect of the test agent on the interaction between leukocytes and endothelial cells.
  • a chemoattractant is introduced into channel 26 in order to "activate" the endothelium.
  • the chemoattractants may be any molecule suitable to stimulate the endothelium to express integrin binding ligands such as ICAMs and VCAMs.
  • Non-limiting examples of chemoattractants include cytokines such as IL-1 and TNF- ⁇ .
  • a sample including leukocytes is then introduced in channel 26 via first well 18 or second well 22.
  • the sample including leukocytes is preincubated with a chemoattractant capable of triggering the activation of arrest mediator binding partners, for example integrins, on the surface of leukocytes.
  • a chemoattractant capable of triggering the activation of arrest mediator binding partners, for example integrins, on the surface of leukocytes.
  • the chemoattractant is any suitable substance capable of triggering integrin expression by leukocytes and includes, for example, formyl peptides, intercrines, IL-8, GRO/MGSA, NAP2, ENA-78, MCP-1/MCAF, RANTES, 1-309, other peptides, platelet activating factor (PAF), lymphokines, collagen, fibrin, and histamines.
  • the number of arrested cells may then be determined.
  • Assays measuring the inhibition of arrest may also be performed, for example, by adding inhibitors that block the interaction between chemoattractants and chemoattractant receptors on the surface of the leukocytes or the endothelium, or that block the interaction between leukocyte arrest mediators and arrest mediator binding partners.
  • a test agent could also be added to channel 26 to determine the interaction between the leukocytes and the endothelial cells in the presence of the test agent.
  • a layer of endothelial cells is placed in channel 26.
  • channel 26 may first be coated with a layer of fibronectin or any other basement membrane mimic before adding the endothelial cells to channel 26.
  • the endothelial cells are exposed to eotaxins or chemokines, including RANTES or monocyte chemoattractant protein (MCP-3 or MCP-4) prior to introduction of the sample containing leukocytes.
  • the sample including leukocytes is then introduced into channel 26 via first well 18 or second well 22.
  • the leukocytes are preincubated with a chemoattractant capable of triggering the activation of arrest mediator binding partners, for example integrins, on the surface of leukocytes.
  • Transmigrated cells may be characterized by appearing flattened and phase-dark under a microscope. Flattened, phase-dark cells may be confirmed as being under the endothelial cell monolayer by observing the focal plane of the leukocytes and the endothelial cells using a microscope. A cell may be considered transmigrated if, for example, greater than 50 % of the cell is under the endothelial cell monolayer at the point of quantification. Transmigration may be expressed as the number of transmigrated cells divided by the total cells counted.
  • Inhibition of transmigration may also be examined by blocking, for example, the receptor on endothelium cells that binds the chemoattractant responsible for activating the endothelium and then determining the number of cells that transmigrate across the endothelium.
  • a test agent may also be introduced in channel 26 to determine the interaction between the leukocytes and the endothelial cells in the presence of the test agent.
  • the endothelial cells disposed in channel 26 have been altered or modified through known techniques in molecular biology.
  • the cells may be modified to overexpress particular genes or to not express particular genes coding for the various leukocyte migration mediators responsible for the leukocyte migration cascade.
  • Such an embodiment affords control over the expression of precise leukocyte migration mediators and allows greater manipulation of the mediator involved in the leukocyte migration cascade.
  • the endothelial cells may be genetically modified to reduce or inhibit the expression of a gene believed to encode a protein involved in the leukocyte migration cascade to assist in the elucidation of the proteins involved in leukocyte migration cascade. Methods for genetically modifying a cell are known in the art.
  • the endothelial cells may be transfected with a vector to genetically modify a protein expressed by the endothelial cells.
  • the endothelial cells maybe modified to express a variant of the protein to be tested.
  • the gene expressing the particular protein can be modified to express a variant. Then using the device and assays of the present invention, the effect of this variant on the various parts of the cascade can be monitored.
  • the variant can be created using techniques known in the art by making deletions, additions or substitutions in the sequence encoding the protein.
  • a "variant" of a polypeptide is defined as an amino acid sequence that is altered by one or more amino acids. Similar minor variations can also include amino acid deletions or insertions, or both. Guidance in determining which and how many amino acid residues may be substituted, inserted or deleted without abolishing biological or immunological activity can be found using computer programs well known in the art, for example, DNAStar software.
  • a “deletion” is defined as a change in either amino acid or nucleotide sequence in which one or more amino acid or nucleotide residues, respectively, are absent as compared to an amino acid sequence or nucleotide sequence of a naturally occurring polypeptide.
  • An “insertion” or “addition” is that change in an amino acid or nucleotide sequence which has resulted in the addition of one or more amino acid or nucleotide residues, respectively, as compared to an amino acid sequence or nucleotide sequence of a naturally occurring polypeptide.
  • substitution results from the replacement of one or more amino acids or nucleotides by different amino acids or nucleotides, respectively as compared to an amino acid sequence or nucleotide sequence of a naturally occurring polypeptide.
  • the variant can have "conservative" changes, wherein a substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine. More rarely, a variant can have "nonconservative" changes wherein a substituted amino acid does not have similar structural or chemical properties such as replacement of a glycine with a tryptophan.
  • an endothelial cell expressing a leukocyte rolling mediator such as P-selectin can be genetically modified such that the expression of the P-selectin is reduced or inhibited using a homologous recombination gene "knock-out" method (see, for example, Capecchi, Nature, 344:105 (1990) and references cited therein; Koller et al, Science, 248:1227-1230 (1990); Zijlstra et al., Nature, 342:435-438 (1989), each of which is incorporated herein by reference; see, also, Sena and Zarling, Nat.
  • a homologous recombination gene "knock-out” method see, for example, Capecchi, Nature, 344:105 (1990) and references cited therein; Koller et al, Science, 248:1227-1230 (1990); Zijlstra et al., Nature, 342:435-438 (1989), each of which is incorporated herein by reference; see
  • a “knock- out” of a target gene means an alteration in the sequence of the gene that results in a decrease of function of the target gene, preferably such that target gene expression is undetectable or insignificant.
  • a knock-out of a gene means that function of the gene has been substantially decreased so that protein expression is not detectable or only present at insignificant levels.
  • a “knock-in” of a target gene means an alteration in a host cell genome that results in altered expression or increased expression of the target gene, e.g., by introduction of an additional copy of the target gene, or by operatively inserting a regulatory sequence that provides for enhanced expression of an endogenous copy of the target gene.
  • the expression of the leukocyte migration mediator by an endothelial cell also can be reduced or inhibited by providing in the endothelial cell an antisense nucleic acid sequence, which is complementary to a nucleic acid sequence or a portion of a nucleic acid sequence encoding a leukocyte migration mediator.
  • an antisense nucleic acid sequence which is complementary to a nucleic acid sequence or a portion of a nucleic acid sequence encoding a leukocyte migration mediator.
  • leukocyte migration mediator Another embodiment creating control over the precise leukocyte migration mediators to be studied, including control over the type and amount of leukocyte migration mediator expressed, involves an embodiment of device 10 wherein at least one leukocyte migration mediator is disposed in channel 26 therein.
  • the term "leukocyte migration mediator” used herein necessarily refers to at least one leukocyte migration mediator, unless otherwise specified.
  • Disposing a leukocyte migration mediator in channel 26 also allows for the precise targeting of the ligand/receptor interactions underlying the individual events of the leukocyte migration cascade.
  • device 10 may be used to examine the capture of a leukocyte wherein the leukocyte migration mediator disposed in channel
  • a leukocyte capture mediator As a consequence of the initial immune response to infection, inflammatory mediators induce the expression of adhesion molecules on the surface of the endothelium, resulting in an "activated endothelium.”
  • the first contact of a leukocyte with the activated endothelium is known as “capture” and is thought to involve a capture mediator P-selectin and a capture mediator binding partner L-selectin.
  • P-selectin is thought to be the primary adhesion molecule involved the capture process and the binding of P-selectin to its main capture mediator binding partner, PSGL- 1 , is strongly implicated in this process.
  • the leukocyte capture mediator disposed in channel 26 may comprise, for example, P-selectin and/or an L-selectin binding ligand.
  • device 10 may be used to examine the rolling of a leukocyte wherein the leukocyte migration mediator may comprise a leukocyte rolling mediator. Once leukocytes are captured, they may transiently adhere to the endothelium and begin to roll along the endothelium.
  • the rolling of leukocytes is thought to mvolve: a rolling mediator, P-selectin; a rolling mediator binding partner, L-selectin; and a rolling mediator, E-selectin, although P- selectin is considered the primary adhesion molecule involved in this process.
  • the leukocyte rolling mediator disposed in channel 26 may include, for example P-selectin, E-selectin, and/or an L-selectin ligand.
  • device 10 may be used to examine the arrest of a leukocyte wherein the leukocyte migration mediator may comprise a leukocyte arrest mediator.
  • chemoattractants such as IL-1 and TNF- ⁇ produced by cells at the injured site.
  • chemoattractants such as IL-1 and TNF- ⁇ produced by cells at the injured site.
  • chemoattractants stimulate the endothelium to produce chemokines and arrest mediators on the surface of the endothelium opposite the basal lamina.
  • the arrest mediators comprise, for example, integrin binding ligands such as ICAMs, including ICAM-1, ICAM-2, or/and ICAM-3 and VCAMs, including VCAM-1 and/or VCAM-2.
  • the chemokines interact with chemokine receptors on the surface of the rolling leukocytes, which triggers the activation of arrest mediator binding partners on the surface of leukocytes.
  • Arrest mediator binding partners include integrins, such as, for example, LFA-1, Mac-1, and pl50,95, and VLA-4. Activation of these arrest mediator binding partners is thought to cause the slowly rolling leukocytes to "arrest” and strongly bind to the arrest mediators, such as ICAM-1, VCAM-1, and other integrin binding ligands such as collagen, fibronectin, and fibrinogen, on the endothelium.
  • the leukocyte arrest mediator disposed in channel 26 may include at least one integrin binding ligand.
  • device 10 may be used to examine the transmigration of a leukocyte wherein the leukocyte migration mediator disposed in channel 26 comprises a leukocyte transmigration mediator.
  • the leukocytes Once bound to the endothelium, the leukocytes flatten and squeeze between the endothelium to leave the blood vessel and enter the damaged tissue. The leukocytes follow a chemotactic gradient of chemoattractants released by cells in the damaged tissue area.
  • transmigration is thought to be mediated by platelets and endothelial cell adhesion molecule-1 (PECAM-1).
  • PECAM-1 endothelial cell adhesion molecule-1
  • the leukocyte transmigration mediator disposed in channel 26 may include at least one of the aforementioned adhesion molecules or any other molecule determined to be implicated in transmigration.
  • Device 10 of the present invention may be used to study each aforementioned step in the leukocyte migration cascade in isolation, a combination of two or more steps in the leukocyte migration cascade, or the leukocyte migration cascade in its entirety.
  • leukocyte rolling mediators may be disposed in channel 26.
  • both rolling and arrest of leukocytes are desired to be studied, then both rolling and arrest mediators may be disposed in channel 26.
  • capture mediators, rolling mediators, arrest mediators, and transmigration mediators may be disposed in channel 26.
  • a leukocyte migration mediator comprising a capture mediator is disposed in channel 26.
  • a sample comprising leukocytes is introduced into channel 26 via first well 18 or second well 22.
  • Capture events are defined as adhesive interactions of those freely flowing leukocytes moving closest to the surface of channel 26 containing the capture mediators and that are therefore the only leukocytes potentially capable of interacting with the capture mediators on channel 26.
  • Different types of initial leukocyte capture can be characterized, observed, and monitored. For example, transient capture involving leukocytes only attaching briefly to channel 26 without initiating rolling motions, and rolling capture involving leukocytes that remain rolling on channel 26, can be determined.
  • the number of each type of captured leukocyte can be divided by the total number of free flowing leukocytes to determine the frequency of initial capture of leukocytes.
  • the leukocytes can also be observed via any method known in the art and via methods disclosed in co-pending application entitled “Test Device and Method of Making Same," which is herein incorporated by reference in its entirety. Briefly, the leukocytes may be observed by using a microscope, including phase-contrast, fluorescence, luminescence, differential-interference contrast, dark field, confocal laser-scanning, digital deconvolution, and video microscopes; a high-speed video camera; and an array of individual sensors.
  • a digital movie camera may be used to monitor leukocyte activity under continuous flow conditions or a camera may be used to obtain still photographic images at particular points in time. Such observations reveal the interaction between the capture mediator binding partner expressed by the leukocytes and the capture mediator expressed by the endothelium.
  • the cells may be incubated with staining agents and then detected based upon color or intensity contrast using any suitable microscopy technique(s).
  • fluorescence-labeling may be used to detect whether capture mediator binding partners bind to capture mediators.
  • non-labeled cells may be used to monitor migration.
  • a heterogeneous mixture of multiple cell types may be introduced into channel 26 with only one cell type capable of interacting with the capture mediators in channel 26. After the cells have been introduced into channel 26, an antibody specific to any antigen on the surface of this cell type may be used to label this cell type. If a multiple number of cell types can interact with the capture mediators, antibodies labeled with specific fluorophores can be used to distinguish different cell types.
  • a leukocyte migration mediator comprising a rolling mediator is placed in channel 26. A sample comprising leukocytes is introduced into channel 26 via first well 18 or second well 22. The number of leukocytes rolling and the rolling velocity of the leukocytes can be determined.
  • a camera is operatively linked to device 10 to obtain images of leukocytes rolling along channel 26 during predetermined intervals over a predetermined period of time.
  • the rolling velocity of the cells is determined by measuring the length the cells traveled (l frame ) in an image obtained by the camera and determining the exposure time of the image (t eX p os ure)-
  • V the rolling velocity
  • a leukocyte migration mediator comprising a first leukocyte migration mediator and a second leukocyte migration mediator, the first and second leukocyte migration mediators being different from one another is utilized.
  • first leukocyte migration mediator used herein necessarily refers to at least one first leukocyte migration mediator and the term “second leukocyte migration mediator” used herein necessarily refers to at least one second leukocyte migration mediator.
  • first leukocyte migration mediator comprising a rolling mediator
  • second leukocyte migration mediator comprising an arrest mediator are placed in channel 26.
  • a fluid sample comprising leukocytes is preincubated with a chemoattractant capable of triggering the activation of arrest mediator binding partners, for example, integrins, on the surface of the leukocytes.
  • the chemoattractant is any suitable substance capable of triggering integrin expression by leukocytes and includes, for example, a formyl peptide, intercrines, IL-8 GRO/MGSA,NAP-2, ENA-78, MCP-1/MCAF, RANTES, 1-309, other peptides, platelet activating factor (PAF), lymphokines, collagen, fibrin and histamines.
  • the number of arrested cells can then be determined and assays similar to those performed with only rolling mediators can be performed.
  • test agents may comprise any biological, chemical or electrical substance that includes, but is not limited to potential inhibitors of the leukocyte migration mediator or potential promoters of migration mediated by the leukocyte migration mediator.
  • test agents include proteins, peptides, polypeptides, enzymes, hormones, lipids, carbohydrates, small molecules, and pharmaceutical agents.
  • the device may be used to identify an inhibitor or promoter that competitively or noncompetitively inhibits or promotes a capture mediator and capture mediator binding partner interaction; rolling mediator and rolling mediator binding partner interaction; arrest mediator and arrest mediator binding partner interaction; and/or transmigration mediator and transmigration mediator binding partner interaction.
  • the leukocyte migration mediator comprises a first leukocyte migration mediator and a second leukocyte migration mediator, the first and second leukocyte migration mediators beings different from one another.
  • the test agent comprises a potential inhibitor of the first leukocyte migration mediator, the second leukocyte migration mediator, or both.
  • the test agent comprises a potential promoter of migration mediated by the first leukocyte migration mediator, the second leukocyte migration mediator, or both.
  • a potential promoter of migration mediated by the first leukocyte migration mediator, the second leukocyte migration mediator, or both.
  • these inhibitors and promoters can be tested for efficacy in vivo and ultimately utilized as therapeutic agents.
  • a leukocyte migration mediator comprising a capture mediator is disposed in channel 26. After the potentially inhibitory test agent is incubated with a fluid sample comprising leukocytes, the sample is introduced into channel 26 via first well 18 or second well 22.
  • Capture events are defined as adhesive interactions of those freely flowing leukocytes moving closest to the surface of channel 26 containing the capture mediators and that are, therefore, the only leukocytes potentially capable of interacting with the capture mediators on channel 26.
  • Different types of initial leukocyte capture can be characterized, observed, and monitored. For example, transient capture involving leukocytes only attaching briefly to channel 26 without initiating rolling motions, and rolling capture involving leukocytes that remain rolling on channel 26 can be determined.
  • the number of each type of captured leukocyte can be divided by the total number of free flowing leukocytes to determine the frequency of initial capture of leukocytes incubated with the potential inhibitory test agent and this frequency can be compared to the frequency of initial leukocyte capture in the absence of the potential inhibitory test agent.
  • a leukocyte migration mediator comprising a rolling mediator is placed in channel 26.
  • the potentially inhibitory test agent is incubated with a fluid sample comprising leukocytes, the sample is introduced into channel 26 via first well 18 or second well 22.
  • the potentially inhibitory test agent is introduced into the fluid sample during passage of the fluid sample in channel 26 when leukocytes have begun rolling. A decrease in rolling (e.g.
  • a leukocyte migration mediator comprising a first leukocyte migration mediator and a second leukocyte migration mediator, the first and second leukocyte migration mediators being different from one another may be utilized.
  • the first leukocyte migration mediator comprising a rolling mediator and the second leukocyte migration mediator comprising an arrest mediator are placed in channel 26.
  • a fluid sample comprising leukocytes is preincubated with a chemoattractant capable of triggering the activation of arrest mediator binding partners, for example, integrins, on the surface of the leukocytes.
  • a chemoattractant capable of triggering the activation of arrest mediator binding partners, for example, integrins
  • the sample is introduced into channel 26 via first well 18 or second well 22.
  • the potentially inhibitory test agent is introduced into the fluid sample during passage of the fluid sample in channel 26 when leukocytes have begun rolling.
  • Device 10 can also be used to identify whether a test agent acts as a promoter of the inflammatory response by increasing the efficiency of the leukocyte migration cascade or by acting as a functional component thereof (e.g. a capture mediator, a rolling mediator, an arrest mediator, or a transmigration mediator).
  • a test agent acts as a promoter of the inflammatory response by increasing the efficiency of the leukocyte migration cascade or by acting as a functional component thereof (e.g. a capture mediator, a rolling mediator, an arrest mediator, or a transmigration mediator).
  • Such a functional component may be detected by its ability to promote capture, rolling, arrest or transmigration of a leukocyte where such action was previously lacking (due to lack of appropriate cellular specificity of a rolling mediator or arrest mediator previously present in channel 26 of chamber 14 or lack of any rolling mediator or arrest mediator).
  • device 10 comprising a first leukocyte migration mediator comprising a rolling mediator and second leukocyte migration mediator comprising an arrest mediator disposed in channel 26 may be used to identify an arrest mediator functional in leukocyte migration.
  • device 10 comprising an arrest mediator and/or a rolling mediator disposed in channel 26 may be used to identify a rolling mediator functional in leukocyte migration.
  • rolling mediators are disposed in channel 26 that have rolling binding partners present on the surface of leukocytes in a fluid sample to be introduced into channel 26 through first well 18 or second well 22.
  • One or more chemoattractants capable of activating the leukocytes to express arrest mediator binding partners are preincubated with the fluid sample comprising leukocytes.
  • a test agent to be tested for arrest mediating function is disposed in channel 26. After the fluid sample comprising leukocytes is introduced into channel 26 via first well 18 or second well 22 and the sample passes along channel 26, it is determined whether any leukocytes have arrested on channel 26. Arrest of leukocytes indicates that the test agent may be an arrest mediator that recognizes an arrest mediator binding partner on the surface of the same leukocytes that express the rolling mediator binding partner.
  • the test agent to be tested for rolling mediator function is disposed in channel 26 and the fluid sample comprising leukocytes is introduced into channel 26 via the first well 18 or second well 22 and the sample is allowed to flow along channel 26. Rolling of the leukocytes along channel 26 indicates that the test agent has rolling mediator function and that the leukocytes express a binding partner for the rolling mediator.
  • a rolling mediator is also identified by disposing the test agent to be tested for rolling mediator function in channel 26 and also disposing an arrest mediator in channel 26.
  • One or more chemoattractants capable of activating the leukocytes to express arrest mediator binding partners, such as integrins, are preincubated with the fluid sample comprising leukocytes.
  • the fluid sample comprising leukocytes is then introduced into channel 26 via the first well 18 or second well 22 and the sample is allowed to flow along channel 26.
  • Arrest of leukocytes indicates that the test agent has rolling mediator function and that the leukocytes that express the arrest mediator binding partner and the chemoattractant receptor also express a binding partner for the test agent.
  • a test agent may also be identified as a functional component in the processes of leukocyte rolling or rolling and arrest, or an enhancer thereof, by the aforementioned methods in which an increase in number or percentage of leukocytes rolling or arrested is detected relative to the number or percentage of such leukocytes in the absence of the test agent.
  • the migration of the leukocytes may be observed, monitored, recorded, and analyzed by any method known in the art and via the methods disclosed in co-pending application, "Test Device and Method of Making Same" referred to above.
  • the present invention also provides a kit to conduct the aforementioned assays.
  • the kit comprises a device including a housing 12 defining a plurality of chambers 14.
  • Each of the plurality of chambers 14 includes a first well region 16 including at least one first well 18; a second well region 20 including at least one second well 22; and a channel region 24 including at least one channel 26 connecting the first well region 16 and the second well region 20 with another.
  • the first well regions 16 and the second well regions 20 of the respective ones of the plurality of chambers 14 are preferably disposed relative to one another to match a pitch of a standard microtiter plate, thus advantageously allowing for high throughput screening of tests agents.
  • the kit further includes a first leukocyte migration mediator.
  • the kit may also contain a sample comprising leukocytes.
  • the first leukocyte migration mediator and the sample comprising leukocytes may be packaged in the kit in any manner known in the art.
  • the first leukocyte migration mediator may be contained in a vial or container and the sample comprising leukocytes may similarly be contained in a separate vial or container.
  • the kit may further include a second leukocyte migration mediator different from the first leukocyte migration mediator.
  • the kit may additionally include an inhibitor adapted to inhibit the first leukocyte migration mediator, the second leukocyte migration mediator, or both.
  • the kit may further include a promoter adapted to promote migration mediated by the first leukocyte migration mediator, the second leukocyte migration mediator, or both.
  • the housing 12 of device 10 comprises a support member 28, and a top member 30 mounted to the support member 28, wherein the support member 28 and the top member 30 are configured such that they together define the plurality of chambers 14.
  • the housing is also sized to match dimensions of a standard microtiter plate, for example, the dimensions of a 24-well microtiter plate, a 96-well microtiter plate, a 384- well microtiter plate, 768-well microtiter plate and a 1536-well microtiter plate.
  • the top member may be made of any suitable material known in the art including glass, plastic, or an elastomeric material such as polydimethylsiloxane (PDMS).
  • the support member may be made of glass, polystyrene, polycarbonate, polyacrylates, polymethyl methacrylate (PMMA), PDMS and other plastics.
  • top member 30 is in conformal contact with support member 28.
  • device 10 comprises a support member 28; and top member 30, the top member 30 mounted to the support member 28 by being placed in conformal contact with the support member 28.
  • the support member 28 and the top member 30 are configured such that they together define at least one chamber 14.
  • the at least one chamber 14 includes a first well region 16 including at least one first well 18; a second well region 20 including at least one second well 22; and a channel region 24 including at least one channel 26 connecting the first well region 16 and the second well region 20 with one another.
  • the at least one channel 26 includes at least one leukocyte migration mediator disposed therein.
  • the at least one channel 26 includes endothelial cells disposed therein.
  • the top member 30 is configured to be placed in reversible, conformal contact with the support member 28.
  • top member 30 is preferably made of a material that is adapted to effect conformal contact, preferably reversible conformal contact, with support member 28.
  • the flexibility of such a material allows top member 30 to form-fittingly adhere to support member 28 in such a way as to form a substantially fluid-tight seal therewith.
  • the conformal contact should preferably be strong enough to prevent slippage of top member 30 on support member 28.
  • top member 30 may be made of a material having the structural integrity to allow top member 30 to be removed by a simple peeling process.
  • top member 30 to be removed from support member 28 after experimentation, properly cleansed, and then reused for future assays.
  • the peeling process does not disturb any surface treatment, such as leukocyte migration mediators or endothelial cells, on support member 28.
  • the substantially fluid-tight seal effected between top member 30 and support member 28 by virtue of the conformal contact of top member 30 with support member 28 prevents fluid from leaking from one chamber to an adjacent chamber, and also prevents contaminants from entering the wells.
  • the seal preferably occurs essentially instantaneously without the necessity to maintain external pressure.
  • the conformal contact obviates the need to use a sealing agent to seal top member 30 to support member 28.
  • the top member 30 is made of a material that does not degrade and is not easily damaged by virtue of being used in multiple tests, and that affords considerable variability in the top member's configuration during manufacture of the same. More preferably, the material may be selected for allowing the top member 30 to be made using photolithography.
  • the material comprises an elastomer such as silicone, natural or synthetic rubber, or polyurethane. In a more preferred embodiment, the material is PDMS.
  • Support member 28 provides a support upon which top member 30 rests, and may be made of any material suitable for this function. Suitable materials are known in the art such as glass, polystyrene, polycarbonate, PMMA, polyacrylates, PDMS, and other plastics.
  • well regions 16 and 20 are vertically offset with respect to one another is a test orientation of device 10.
  • well regions 16 and 20 are horizontally offset with respect to one another is a test orientation of device 10.
  • Wells 18 and 22 of respective well regions 16 and 20 of each chamber 14 are not limited in their configuration to any particular three dimensional contour, it being only required that they be adapted to receive a fluid therein, preferably a sample comprising leukocytes.
  • wells 18 and 22 are configured such that they substantially define circles in top plan views thereof, as shown by way of example in Figs. 1-6.
  • other contours in the top plan view of a given well is within the scope of the present invention, as readily recognized by one skilled in the art.
  • the pitch P is set to be equal to about 9 mm
  • the diameter D w of a top plan contour of the wells is set to be equal to about 6 mm.
  • length L of each channel 26 is equal to about 3 mm.
  • wells 18 and 22 are defined in part by respective through-holes 18 and 22 in top member 30, and in part by an upper surface U of support member 28.
  • the sides of each well 18 and 22 are defined by respective walls of the tlirough holes 18 and 22 in the top member 30, and the bottoms of wells 18 and 22 are defined by a corresponding portion of the upper surface U of support member 28.
  • a length L of a channel 26 is defined in a direction of the longitudinal axis thereof.
  • depth D of a channel 26 is defined in a direction normal to a top surface of housing 12; and a width W of a channel region 26 is defined in a direction normal to length L and depth D.
  • channel region 24 comprises a plurality of rectilinear, parallel channels 26 extending between well regions 16 and 20.
  • channels 26 have lengths L that are substantially identical, as shown schematically by way of example in Fig. 4. More preferably, the plurality of channels 26 comprises eight channels. By using multiple channels, multiple assays can be performed simultaneously using one sample comprising leukocytes.
  • Channels 26 preferably each have a width W of 50 ⁇ m to 5 mm; a length L of about 1-10 mm; and a height H of about 10-100 ⁇ m. More preferably, the channels 26 each have a width W of about 100 microns, a length L of about 3 mm and a height of about 50 ⁇ m to about 80 ⁇ m.
  • the dimensions of the channels 26 of channel region 24 should be configured to support the migration of leukocytes under conditions simulating such migration during an inflammatory response. As such, the channel region should be • adapted to support the migration of leukocytes under shear flow and to support at least one leukocyte migration mediator disposed therein.
  • Device 10 of the present invention can be fabricated, according to a preferred embodiment of a method of the present invention, by standard photolithographic procedures.
  • Photolithographic procedures can be used to produce a master that is the negative image of any desired configuration of top member 30.
  • the dimensions of chamber 14, such as the size of well region 16 and 18, or the length of channel region 24, can be altered to fit any advantageous specification.
  • the material for top member 30 is either spin cast, injected, or poured over the master and cured. Once the mold is created, this process can be repeated as often as necessary. This process not only provides great flexibility in the top member's design, it also allows the top members to be massively replicated.
  • leukocyte migration mediators can be disposed in channel 26.
  • the leukocyte migration mediators can be disposed in channel 26 by affixing them or physioadsorbing them directly on the upper surface U of support member 28, or by coating a solution or suspension comprising the leukocyte migration mediators on the upper surface U of support member 28, as long as the mediators are accessible to leukocytes flowing by the upper surface U.
  • the leukocyte migration mediators are either covalently or non- covalently affixed directly to upper surface U by techniques such as covalent bonding via an amide, ester or lysine side chain linkage or adsorption.
  • the present invention also provides a device comprising a housing 12; means associated with the housing defining a plurality of chambers 14 in the housing 12.
  • Each of the plurality of chambers 14 includes: an inlet means for receiving a sample comprising leukocytes; an outlet means in flow communication with the inlet means for receiving the sample comprising leukocytes from the inlet means; and connection means connecting the inlet means and the outlet means to one another, the connection means including at least one leukocyte migration mediator disposed therein.
  • an example of means associated with the housing defining a plurality of chambers in the housing comprises a top member mounted to a support member as shown in Fig. 6.
  • the above means have been substantially shown and described in relation to the embodiments of the Fig. 1-6. Other such means would be within the knowledge of persons skilled in the art.
  • the present invention provides methods of assaying and studying biological phenomenon that either depend on or react to gradient formation and/or flow conditions. Such biological phenomenon include many of the processes in the body such as cell-surface interactions such as that occurring during leukocyte adhesion and rolling. In addition, studies involving chemotaxis, haptotaxis and cell migration will be better served with assays that are able to study such cell movement in the presence of gradients and/or flow conditions.
  • Various types of gradients are useful in the study of biological systems.
  • Such useful gradients include static gradients, which have concentrations that are fixed, or set or substantially fixed or set.
  • a static gradient is a gradients of immobilized molecules on a surface.
  • static gradients include the use of differing concentrations of immobilized biomolecules (proteins, antibodies, nucleic acids, and the like) or immobilized chemical moieties (drugs and small molecules).
  • Other useful gradients include dynamic gradients, which have concentrations that may be varied.
  • One example of a dynamic gradient is a gradient of fluid streams having molecules in varying concentrations.
  • fluid gradients include the use of fluid streams containing biomolecules such as growth factors, toxins, enzymes, proteins, antibodies, carbohydrates, drugs or other chemical and small molecules in varying concentrations.
  • a dynamic/solution based gradient is created by laminar flow technology.
  • Laminar flow technology typically involves two or more fluid streams from two or more different sources. These fluid streams are brought together into a single stream and are made to flow parallel to each other without turbulent mixing. Fluids with different characteristics such as varying low Reynolds numbers will flow side by side and will not mix in the absence of turbulence. Since the fluids do not mix, they create pseudo-channels (pseudo by the fact that there are no physical separation between the fluids). The generation of solution and surface gradients is discussed in U.S. patent application 2002/0113095 and an article, Jeon, Noo Li, et al., Langmuir, 16, 8311-8316 (2000).
  • the devices of the present invention By combining the devices of the present invention with the formation of a dynamic gradient, a vast number of assay parameters can be generated by altering any portion of the device. For example, by combining the device as disclosed herein with cell patterning techniques, along with the introduction of a dynamic gradient, various conditions can be created to test numerous biological interactions. Further, the device and assays may be useful in drug discovery and drug testing as many cells and biological materials behave differently ex vivo when not exposed to gradients than compared to when the cells or biological materials are present in vivo and thus exposed to gradients and flow conditions.
  • cells can be patterned across channel 26.
  • Cell patterning can be achieved by methods known in the art, as well as disclosed in the present invention (such as, but not limited to, microcontact printing or by the use of elastomeric stencils).
  • a solution containing any desired biomolecule or chemical/drug can then be flowed across the patterned cells.
  • the cells could be first treated by a biomolecule such as an activator to more closely recreate a biological system, and then be subsequently exposed to a chemical or drug.
  • a biomolecule such as an activator
  • a chemical or drug By creating a gradient, such as by laminar flow, different amounts of biomolecules or chemicals/drugs can be delivered to the patterned cells and thus the effect of concentration of each biomolecule or chemical/drug be tested simultaneously against each other.
  • immobilized cells or other immobilized biomolecules such as proteins, antibodies, nucleic acids, etc.
  • a single type of cell is immobilized throughout the entire chamiel region 24.
  • a mixture of cell types are immobilized, one cell type per region 24.
  • a mixture of cell types is immobilized throughout the entire chamiel region 24. This may be advantageous in monitoring cell-cell interactions.
  • different cell types are immobilized in each different region 24.
  • leukocyte migration mediators or other biomolecules present in channels 24 are present in one embodiment.
  • one type of leukocyte migration mediator is present in each channel 24 at the same concentration.
  • one type of leukocyte migration mediator is present in each channel 24 at differing concentrations.
  • different leukocyte migration mediators are present in each channel 24.
  • Each channel 24 may have the same mixture or a different mixture. When the mixture is the same, the ratios or concentrations of the different leukocyte migration mediators may be different in each channel 24.
  • the present invention provides flexibility in assay design. For example, in one embodiment a single compound is present in all channels 24 at the same concentration throughout. In another embodiment, the same compound is present in all the channels 24 but each channel 24 has a different concentration of that compound. In another embodiment, each channel 24 has a different compound, hi another embodiment, there is more than one compound. When there is more than one compound, each channel 24 may have the same mixture of compounds or may have a different mixture of compounds. Further, when the mixtures of the compounds are the same, each channel 24 may receive a different concentration of that mixture. Yet, even further, each channel 24 may receive the mixture of the compounds, with each channel 24 having a different ratio of compounds to each other.
  • Such assay systems can be used to test among many numerous biological interactions, the effects of chemical or drugs on cells or other biomolecules.
  • a master of the device according to the present invention is made using photolithography.
  • a silicon substrate is patterned based on a negative pattern of the top member using a suitable photoresist.
  • PDMS polydimethyl siloxane
  • the thus poured PDMS layer is allowed to cure in an oven at about 30°C for about 17 hours.
  • the device is washed thoroughly with 2% Micro-90 (a product of International Products Corp.), rinsed for 10 minutes at 70° C in "Sonic Bath,” and rinsed with de-ionized water, followed by a rinsing with 100% ethanol.
  • the PDMS layer is then dried under nitrogen.
  • a pre-cleansed glass slide such as a rectangular one having dimensions of about 4.913 +/- 0.004 inches (in.) by about 3.247 +/-0.004 in. and a thickness of about 1.75 millimeters (mm), mm, is washed three times with ethanol and twice with methanol.
  • the surfaces of the PDMS layer and the glass slide to be bound together are both plasma oxidized for about 84 seconds.
  • the PDMS layer and the glass slide are then pressed together using forceps to squeeze out air pockets therebetween. In this manner, a fluid-tight, conformal contact is established between the PDMS layer as top member and the glass slide and support member.
  • Neutrophils are isolated from a volume of 5 milliliters (ml) of human blood from a healthy volunteer.
  • the 5 ml of blood is diluted with Hanks Balance Salt Solution (HBSS) in a 1 :2 ratio thereby increasing the total volume of blood to equal 15 ml.
  • HBSS Hanks Balance Salt Solution
  • the whole blood dilution is layered over 10ml of Ficoll-Paque Plus (obtained from Amersham Pharmacia Biotech AB, catalog # 17-1440-02).
  • the blood is then centrifuged for 30 minutes at 400g at room temperature. The supernatant is aspirated off without disturbing the pellet.
  • the pellet is resuspended on 10 ml of HBSS and 150 ⁇ l of 6% dextran to make up a 1% solution.
  • the red blood cells are allowed to settle for at least one hour at room temperature.
  • the neutrophils remain inside the supernatant while the red blood cells mostly settle down forming a pellet.
  • the supernatant is pipetted out and diluted in a 1 :2 ratio using HBSS.
  • This suspension is centrifuged for 10 minutes at a velocity of 600g.
  • the supernatant is aspirated and the pellet is dissolved in 19 ml of deionized water.
  • the pellet is resuspended in 1 ml of 10X PBS. This suspension is centrifuged at 400g for 10 minutes.
  • the red blood cells are lysed in this process and the remaining cells are mostly neutrophils.
  • the resulting pellet may be dissolved in media containing BSA in order to avoid the clumping of cells after a prolonged period of time at room temperature.
  • the cell density is determined by counting the number of cells using a hemocytometer.
  • PBS Phosphate Buffer Saline
  • neutrophils obtained from the method described in part A in 60 ⁇ l of media are added to the first well of each chamber (about 10 3 to about 10 6 cells per well of a 24 well plate, in volume of 60 ⁇ l of media per well) (non-labeled and fluorescently labeled monocytic cell lines-U937 (obtained from ATCC, catalog # TLB-202 and THP-1 (obtained from ATCC, catalog # CRL-1593.2) as well as other primary leukocytes may also be used.
  • V c(lti m e /te ⁇ posure)
  • c conversation factor for determining the actual distance the cells traveled. This factor may vary from image to image.
  • ltime the length of the leukocytes migration in the captured image.
  • texposure the exposure time of the image.
  • t eX posure is 100 milliseconds (ms) when the flow rate is about 0.1 dynes/cm to about 20 dynes/cm .
  • Neutrophils are isolated according to the method disclosed in section II, part A.
  • the endothelial cells are prepared for seeding.
  • Cells are obtained from Clonetics at Bio-Whittaker in cryogenic vials. They are grown in T75 flasks until ready to be split using 0.025% Trypsin EDTA. The cells are seeded on the channel at a density of lxlO 5 cells per 5 ⁇ l of media per assay for approximately two days to form a confluent monolayer of endothelial cells. During these two days, the endothelial cells are replenished with 40 ⁇ L of fresh media added into each well.
  • the endothelial cells are exposed to a concentration of 1 ng/ml of TNF- ⁇ (other chemokines may alternatively be used) for a period of four hours at 37°C.
  • TNF- ⁇ other chemokines may alternatively be used
  • the TNF- ⁇ is washed using 60 ⁇ L of fresh media twice. The volume of media inside each well is replaced with 15 ⁇ L of fresh media.
  • Neutrophils isolated from Section II, part A in 60 ⁇ l of media are added to the first well of chamber the (about 10 to about 10 cells per well of a 24 well plate, in volume of 60 ⁇ l of media per well) (non-labeled and fluorescently labeled monocytic cell lines-U937 and THP-1 as well as primary leukocytes may also be used.) If a monocytic cell line is used, the cells are fluorescence labeled using cell tracker probes (obtained from Molecular Probes, catalog #s C-2925 and C-2927). The cells are incubated with a l ⁇ M concentration of probes for 30 minutes at 37°C. The media is then changed and the cells are placed inside an incubator for an additional 30 minutes.
  • V C ⁇ time /texposure)
  • c conversation factor for determining the actual distance the cells traveled. This factor may vary from image to image.
  • l t i me the length of the leukocytes migration in the captured image.
  • te x posure the exposure time of the image.
  • t eX posure is 100 ms when the flow rate is about 0.1 dynes/cm 2 to about 20 dynes/cm 2 .
  • Neutrophils are isolated according to the method disclosed in section II, part A.
  • B. Placement of Leukocytes, P-selectin, and P-selectin Antibodies in the Chamber 20 ⁇ l of 0.1% BSA are pipetted in the first well of each chamber of the device fabricated according to the method described in Section I. Microcapillary action draws water into the channels. After ensuring no air bubbles are inside the channels, an additional 10 ⁇ l of BSA are pipetted in the second well of each chamber.
  • P-Selectin 50 ⁇ g/mL
  • ICAM-1 50 ⁇ g/mL
  • the device is incubated for two hours at room temperature in a dish with a cover in order to keep the wells from drying out. After the incubation, the channels of each well are washed four times using 0.1% Bovine Serum Albumin (BSA) in Phosphate Buffer Saline (PBS).
  • BSA Bovine Serum Albumin
  • PBS Phosphate Buffer Saline
  • 100 ng/mL of P- selectin antibody is pipetted into the first well of chamber #1; 10 ng/mL of P- selectin antibody is pipetted into first well of chamber #2; and lng/mL of P-selectin antibody is pipetted into the first well of chamber #3; 100 ⁇ g/mL of P-selectin antibody is pipetted into the first well of chamber #4; and 0.1% BSA in PBS is pipetted into the first well of chamber #5.
  • the device is incubated for thirty minutes at room temperature in a dish with a cover in order to keep the wells from drying out.
  • the channels are washed first with 20 ⁇ l of BSA, then with lO ⁇ l of BSA and then 0.1% BSA in PBS.
  • Neutrophils in 20 ⁇ l of media are added to the first well of each chamber (about 10 to about 10 per well of a 24 well plate, in volume of 20 ⁇ l of media per well) (non-labeled and fluorescently labeled monocytic cell lines-U937 and THP-1 as well as primary leukocytes may be used).
  • Digital images are taken on a Zeiss inverted microscope using AXIOCAMTM beginning 15 seconds after the sample comprising leukocytes is added to the first well. Data is analyzed on AXIOVISIONTM software. Time-lapsed images are taken every 30 seconds for 5 minutes and 15 seconds.
  • 10X objective lens is used to view and record the number of cells rolling after the treatment with P-selectin antibody.
  • a lOOng/mL dilution of the antibody is a preferred concentration to inhibit the rolling of the cells.
  • the number of leukocytes that roll and adhere to the endothelium are reduced in the presence of anti-P selectin.
  • E-Selectin 50 ⁇ g/mL
  • ICAM-1 50 ⁇ g/mL
  • the device is incubated for two hours at room temperature in a dish with a cover in order to keep the wells from drying out. After the incubation, the channels of each well are washed four times using 0.1% Bovine Serum Albumin (BSA) in Phosphate Buffer Saline (PBS).
  • BSA Bovine Serum Albumin
  • PBS Phosphate Buffer Saline
  • E- selectin antibody is pipetted into the first well of chamber #1; 10 ng/mL of E- selectin antibody is pipetted into first well of chamber #2; and Ing/mL of E-selectin antibody is pipetted into the first well of chamber #3; 100 ⁇ g/mL of E-selectin antibody is pipetted into the first well of chamber #4; and 0.1 % BSA in PBS is pipetted into the first well of chamber #5.
  • the device is incubated for thirty minutes at room temperature in a dish with a cover in order to keep the wells from drying out. After incubation, the channels are washed four times with 0.1% BSA in PBS.
  • Neutrophils in 20 ⁇ l of media are added to the first well of each chamber (about 10 3 to about 10 6 cells per well of a 24 well plate, in volume of 20 ⁇ l of media per well) (non-labeled and fluorescently labeled monocytic cell lines-U937 and THP-1 as well as primary leukocytes may be used).
  • Digital images are taken on a Zeiss inverted microscope using AXIOCAMTM beginning 15 seconds after the sample comprising leukocytes is added to the first well. Data is analyzed on AXIOVISIONTM software. Time-lapsed images are taken every 30 seconds for 5 minutes and 15 seconds. 10X objective lens is used to view and record the number of cells rolling after the treatment with E-selectin antibody.
  • a lOOng/mL dilution of the antibody is a preferred concentration to inhibit the rolling of the cells.
  • the number of leukocytes that roll and adhere to the endothelium are reduced in the presence of anti-E selectin.
  • Neutrophils are isolated according to the method disclosed in section IV, part A.
  • Endothelial cells are placed and activated in four different channels of four chambers (#l-#4) according to the method disclosed in section IV, part B. With respect to a fifth (#5) chamber, endothelial cells are placed in the channel, but are not activated.
  • 100 ⁇ g/ml of P- selectin antibody is pipetted into the first well of chamber #1; 100 ⁇ g/ml of E- selectin antibody is pipetted into the first well of chamber #2; 100 ⁇ g/ml of VCAM- 1 antibody is pipetted into the first well of chamber #3; and 100 ⁇ g/ml of BSA in PBS is pipetted into the first well of chamber #4.
  • the device is incubated for thirty minutes at room temperature in a dish with a cover in order to keep the wells from drying out. After incubation, the channels are washed four times with 0.1% BSA in PBS.
  • Neutrophils in 20 ⁇ l of media are added to the first well of each chamber (about 10 to about 10 cells per well of a 24 well plate, in volume of 20 ⁇ l of media per well) (non-labeled and fluorescently labeled monocytic cell lines-U937 and THP-1 as well as primary leukocytes may be used).
  • Digital images are taken on a Zeiss inverted microscope using AXIOCAMTM beginning 15 seconds after the sample comprising leukocytes is added to the first wells. Data is analyzed on AXIO VISIONTM software. Time-lapsed images are taken every 30 seconds for 5 minutes and 15 seconds. 10X objective lens is used to view and record the number of cells rolling after the treatment with the antibodies as seen in Figure 10.
  • TNF- ⁇ was delivered to the lawn of endothelial cells via laminar flow to "activate" the endothelial cells.
  • Each stream of solutions containing TNF- ⁇ were at different concentrations, thus creating a gradient perpendicular to the channel. This gradient effectively delivered TNF- ⁇ to the lawn of endothelial cells at different concentrations at different positions on the lawn of cells.
  • Leukocytes were then flowed over the lawn of activated endothelial cells. Referring to Figure 12, only those endothelial cells that were activated by TNF- ⁇ provide suitable "attachment" sites for the leukocytes.
  • the leukocytes did not attach equally to the entire lawn, but attached to the areas of the endothelial cell lawn that had been exposed to high concentrations of TNF- ⁇ and did not attach to those areas of the lawn that had been exposed to low concentrations of TNF- ⁇ , or those areas not exposed to TNF- ⁇ at all. These results indicate that there was indeed a creation of a concentration gradient of TNF- ⁇ by the laminar flow.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nanotechnology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pathology (AREA)
  • Clinical Laboratory Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Food Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Composite Materials (AREA)
  • Condensed Matter Physics & Semiconductors (AREA)
  • Materials Engineering (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

L'invention concerne un dispositif utilisé pour surveiller la migration des leucocytes, ainsi qu'un procédé d'utilisation de ce dispositif pour surveiller la migration des leucocytes en présence d'un flux de cisaillement physiologique et donc pour simuler les conditions physiologiques d'un vaisseau sanguin in vivo. L'invention concerne également un procédé d'utilisation de ce dispositif pour le criblage à haut débit d'une pluralité d'agents de test. De plus, l'invention concerne un système d'essai flexible et de nombreux essais pouvant être utilisés pour tester des interactions biologiques et des systèmes. Des gradients de flux laminaires sont utilisés pour reproduire des situations de gradient existant in vivo.
PCT/US2003/033177 2002-10-22 2003-10-21 Dispositif et procede pour surveiller la migration des leucocytes WO2004038368A2 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP03774884A EP1558076A4 (fr) 2002-10-22 2003-10-21 Dispositif et procede pour surveiller la migration des leucocytes
AU2003282950A AU2003282950A1 (en) 2002-10-22 2003-10-21 Device and method for monitoring leukocyte migration
CA002503203A CA2503203A1 (fr) 2002-10-22 2003-10-21 Dispositif et procede pour surveiller la migration des leucocytes
JP2004546913A JP2006503581A (ja) 2002-10-22 2003-10-21 白血球遊走をモニタリングするための装置及び方法

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US41997602P 2002-10-22 2002-10-22
US41998002P 2002-10-22 2002-10-22
US60/419,976 2002-10-22
US60/419,980 2002-10-22

Publications (2)

Publication Number Publication Date
WO2004038368A2 true WO2004038368A2 (fr) 2004-05-06
WO2004038368A3 WO2004038368A3 (fr) 2004-07-29

Family

ID=32179774

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2003/033177 WO2004038368A2 (fr) 2002-10-22 2003-10-21 Dispositif et procede pour surveiller la migration des leucocytes
PCT/US2003/033146 WO2004038367A2 (fr) 2002-10-22 2003-10-21 Tests biologiques utilisant des gradients formes dans des systemes microfluidiques

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2003/033146 WO2004038367A2 (fr) 2002-10-22 2003-10-21 Tests biologiques utilisant des gradients formes dans des systemes microfluidiques

Country Status (5)

Country Link
EP (2) EP1558076A4 (fr)
JP (2) JP2006503581A (fr)
AU (2) AU2003282950A1 (fr)
CA (2) CA2503186A1 (fr)
WO (2) WO2004038368A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013158939A1 (fr) * 2012-04-18 2013-10-24 Hemoshear, Llc Modèle in vitro pour conditions pathologiques ou physiologiques
US9121847B2 (en) 2008-04-08 2015-09-01 Massachussetts Institute Of Technology Three-dimensional microfluidic platforms and methods of use thereof
WO2016065125A1 (fr) * 2014-10-22 2016-04-28 The General Hospital Corporation Procédé et appareil de visualisation d'objets à l'aide d'un rétroéclairage oblique
US9617521B2 (en) 2013-10-21 2017-04-11 Hemoshear, Llc In vitro model for a tumor microenvironment
CN114887034A (zh) * 2022-07-14 2022-08-12 中山莱博瑞辰生物医药有限公司 LLP2A-Ale在制备用于治疗和/或预防外周血淋巴细胞减少症的药物中的用途

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005123950A2 (fr) * 2004-05-19 2005-12-29 Massachusetts Institute Of Technology Modeles de maladie comprenant des tissus/cellules perfuses a trois dimensions
US7351423B2 (en) 2004-09-01 2008-04-01 Depuy Spine, Inc. Musculo-skeletal implant having a bioactive gradient
KR100873383B1 (ko) 2007-11-13 2008-12-10 이화여자대학교 산학협력단 선충의 주화성 연구용 미세 유체 소자
DE102009035502A1 (de) 2009-07-30 2011-02-03 Universitätsklinikum Jena Verfahren und Vorrichtung zur Erfassung der Bewegung und Anlagerung von Zellen und Partikeln an Zell-, Gewebe- und Implantatschichten bei der Simulation von Flussbedingungen
DE102009038520B4 (de) * 2009-08-25 2012-07-05 Universität Rostock Verfahren und Vorrichtung zur Zellprobendiagnostik
CN102580798B (zh) * 2012-02-28 2014-03-05 东北大学 一种微流控芯片溶液梯度稀释装置
JP5910282B2 (ja) * 2012-04-20 2016-04-27 大日本印刷株式会社 細胞遊走解析方法および細胞培養物作成方法
KR101433091B1 (ko) * 2012-04-26 2014-08-25 한국과학기술원 박테리아의 주화성 분석용 마이크로 플루이딕 장치, 제조방법 및 이를 이용한 박테리아의 주화성 분석 방법
CN105080627A (zh) * 2015-08-25 2015-11-25 辽宁中医药大学 一种用于药物筛选的集成化微流控芯片及其应用方法
CN106497788A (zh) * 2016-11-28 2017-03-15 南方医科大学珠江医院 体外肝细胞异质性培养装置及其培养方法
WO2019045926A1 (fr) * 2017-08-30 2019-03-07 Imprimed, Inc. Dispositifs et procédés de criblage à haut débit de composés chimiques et biochimiques
CN111019828B (zh) * 2019-12-26 2021-12-21 武汉大学 一种开放式高通量微流控卵母细胞动态三维培养芯片及其应用
WO2023144747A1 (fr) * 2022-01-28 2023-08-03 Glaxosmithkline Intellectual Property Development Limited Dosage à haut débit pour migration cellulaire et chimiotaxie et fonction associée

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5460945A (en) * 1991-05-30 1995-10-24 Center For Blood Research, Inc. Device and method for analysis of blood components and identifying inhibitors and promoters of the inflammatory response
US20020168757A1 (en) * 2000-11-08 2002-11-14 Gregory Kirk Test device and method of making same
US6632619B1 (en) * 1997-05-16 2003-10-14 The Governors Of The University Of Alberta Microfluidic system and methods of use

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5284753A (en) * 1991-03-20 1994-02-08 Neuro Probe, Inc. Multiple-site chemotactic test apparatus and method
EP0981408B1 (fr) * 1997-05-16 2004-04-21 Alberta Research Council Systeme microfluidique et ses utilisations
AU5291600A (en) * 1999-06-01 2000-12-18 Caliper Technologies Corporation Microscale assays and microfluidic devices for transporter, gradient induced, and binding activities
US7326563B2 (en) * 2000-11-08 2008-02-05 Surface Logix, Inc. Device and method for monitoring leukocyte migration
AU2003213834A1 (en) * 2002-03-12 2003-09-29 Surface Logix, Inc. Cell motility and chemotaxis test device and methods of using same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5460945A (en) * 1991-05-30 1995-10-24 Center For Blood Research, Inc. Device and method for analysis of blood components and identifying inhibitors and promoters of the inflammatory response
US6632619B1 (en) * 1997-05-16 2003-10-14 The Governors Of The University Of Alberta Microfluidic system and methods of use
US20020168757A1 (en) * 2000-11-08 2002-11-14 Gregory Kirk Test device and method of making same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
See also references of EP1558076A2 *
TEMPELMAN L.A. ET AL.: 'Motion of model leukocytes near a wall in simple shear flow' BIOTECHNOL. PROG. vol. 10, 1994, pages 97 - 108, XP002977146 *
VEREYCKEN-HOLLER V. ET AL.: 'Radiation effects on circulating and endothelial cell interactions studied by quantitative real-time videomicroscopy' INT. J. RADIATION BIOL. vol. 78, no. 10, September 2002, pages 923 - 930, XP002977145 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9121847B2 (en) 2008-04-08 2015-09-01 Massachussetts Institute Of Technology Three-dimensional microfluidic platforms and methods of use thereof
WO2013158939A1 (fr) * 2012-04-18 2013-10-24 Hemoshear, Llc Modèle in vitro pour conditions pathologiques ou physiologiques
US9500642B2 (en) 2012-04-18 2016-11-22 Hemoshear, Llc In vitro model for pathological or physiologic conditions
US9658211B2 (en) 2012-04-18 2017-05-23 Hemoshear, Llc In vitro model for pathological or physiologic conditions
US10514374B2 (en) 2012-04-18 2019-12-24 Hemoshear, Llc In vitro model for pathological or physiologic conditions
US10837957B2 (en) 2012-04-18 2020-11-17 Hemoshear, Llc In vitro model for pathological or physiologic conditions
US11061016B2 (en) 2012-04-18 2021-07-13 Hemoshear, Llc In vitro model for pathological or physiologic conditions
US9617521B2 (en) 2013-10-21 2017-04-11 Hemoshear, Llc In vitro model for a tumor microenvironment
US10221394B2 (en) 2013-10-21 2019-03-05 Hemoshear, Llc In vitro model for a tumor microenvironment
US11008549B2 (en) 2013-10-21 2021-05-18 Hemoshear, Llc In vitro model for a tumor microenvironment
WO2016065125A1 (fr) * 2014-10-22 2016-04-28 The General Hospital Corporation Procédé et appareil de visualisation d'objets à l'aide d'un rétroéclairage oblique
CN114887034A (zh) * 2022-07-14 2022-08-12 中山莱博瑞辰生物医药有限公司 LLP2A-Ale在制备用于治疗和/或预防外周血淋巴细胞减少症的药物中的用途

Also Published As

Publication number Publication date
CA2503186A1 (fr) 2004-05-06
JP2006503581A (ja) 2006-02-02
AU2003287170A1 (en) 2004-05-13
WO2004038367A2 (fr) 2004-05-06
CA2503203A1 (fr) 2004-05-06
EP1578194A4 (fr) 2007-03-21
JP2006507815A (ja) 2006-03-09
AU2003282950A1 (en) 2004-05-13
EP1558076A4 (fr) 2007-03-21
WO2004038368A3 (fr) 2004-07-29
EP1558076A2 (fr) 2005-08-03
WO2004038367A3 (fr) 2004-11-11
EP1578194A2 (fr) 2005-09-28

Similar Documents

Publication Publication Date Title
WO2004038368A2 (fr) Dispositif et procede pour surveiller la migration des leucocytes
US20070038384A1 (en) Devices and assays for monitoring/measuring cellular dynamics to create subject profiles from primary cells
JP7416749B2 (ja) 細胞分泌物の分析のための装置及び方法
US7374906B2 (en) Biological assays using gradients formed in microfluidic systems
US8921122B2 (en) System and method for quantitative assessment of biological migration behavior
US7326563B2 (en) Device and method for monitoring leukocyte migration
WO2003091730A1 (fr) Dispositif et procede destines a controler la migration des leucocytes
US20040178071A1 (en) Microfluidic system and methods of use
KR20120026551A (ko) 세포 집단 및 혼합된 세포 집단에서의 변화의 검출 방법
Muller et al. Assays of transendothelial migration in vitro
Bongrand et al. Studying cell adhesion
Müller Novel tools for the study of cell type-specific exosomes and microvesicles
Caserta et al. A methodology to study chemotaxis in 3‐D collagen gels
AU2006292757A1 (en) Cell migration assay
Shortman Separation methods for lymphocyte populations
US7419823B2 (en) Device and method for monitoring leukocyte migration
US20120058507A1 (en) Clonal Derivation and Cell Culture quality Control Screening Methods
US20030044766A1 (en) Methods and devices for detecting cell-cell interactions
US20030044879A1 (en) Method and device for forming a concentration gradient for chemotactic evaluation through use of laminar flow
WO2006132326A1 (fr) Procédé d’immobilisation d’une cellule vitale et capteur destiné à mesurer la fonction d’activation de la cellule vitale
Francis Direct measurements of cell adhesion forces
Hallila Experimental work: MONITORING DRUG-CELL INTERACTIONS WITH SURFACE PLASMON RESONANCE TECHNIQUE
Usman Quantification of affinity mediated cell/surface interactions

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2503203

Country of ref document: CA

Ref document number: 2004546913

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003282950

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003774884

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003774884

Country of ref document: EP