WO2009061392A1 - Formation de structures de gel à l'aide de canaux microfluidiques - Google Patents

Formation de structures de gel à l'aide de canaux microfluidiques Download PDF

Info

Publication number
WO2009061392A1
WO2009061392A1 PCT/US2008/012445 US2008012445W WO2009061392A1 WO 2009061392 A1 WO2009061392 A1 WO 2009061392A1 US 2008012445 W US2008012445 W US 2008012445W WO 2009061392 A1 WO2009061392 A1 WO 2009061392A1
Authority
WO
WIPO (PCT)
Prior art keywords
gel
fluid
channel
cells
microfluidic channel
Prior art date
Application number
PCT/US2008/012445
Other languages
English (en)
Inventor
Amy P. Wong
Raquel Perez-Castillejos
J. Christopher Love
George M. Whitesides
Original Assignee
President And Fellows Of Harvard College
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 President And Fellows Of Harvard College filed Critical President And Fellows Of Harvard College
Publication of WO2009061392A1 publication Critical patent/WO2009061392A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0062General methods for three-dimensional culture
    • 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
    • B01L3/502707Containers 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 characterised by the manufacture of the container or its components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/16Microfluidic devices; Capillary tubes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M25/00Means for supporting, enclosing or fixing the microorganisms, e.g. immunocoatings
    • C12M25/14Scaffolds; Matrices
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/06Fluid handling related problems
    • B01L2200/0636Focussing flows, e.g. to laminate flows
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/06Fluid handling related problems
    • B01L2200/0647Handling flowable solids, e.g. microscopic beads, cells, particles
    • 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
    • B01L2300/069Absorbents; Gels to retain a fluid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2535/00Supports or coatings for cell culture characterised by topography
    • C12N2535/10Patterned coating

Definitions

  • the present invention relates generally to articles and methods for forming structures in microfluidic channels, and more specifically, to articles and methods for forming structures comprising gels in microfluidic channels.
  • Microfluidic devices are flow systems miniaturized to dimensions as small as a few micrometers ( ⁇ m), or even smaller in some instances. These devices can be used to manipulate and handle small fluid samples, to pattern biological materials such as proteins and cells, and to produce gel structures in the microchannels of the device. Reduction of the dimensions of gel structures has been a challenge. Several techniques have demonstrated that microfabrication makes it possible to reduce the dimensions of the structures of gel. For example, poly(dimethylsiloxane) (PDMS) stamps fabricated by soft lithography can mold layers of most types of gel, including biologically-derived ones that gel thermally, but typically they can generate only planar patterns — or layered stacks of them.
  • PDMS poly(dimethylsiloxane)
  • Certain photolithographic techniques can produce more complex structures than those based on molding, but may be limited to synthetic hydrogels that cure by UV light. Accordingly, although methods for fabricating gel structures exist, systems and methods that would allow for more flexible designs of gel structures, are compatible with biologically-derived gels, and/or allow easier integration of components within the gels would be beneficial.
  • a method comprises flowing a first fluid and a second fluid simultaneously in a microfluidic channel, and forming a gel structure comprising substantially all of the first fluid in the microfluidic channel.
  • the gel may have a length of at least 0.5 times, at least 0.75 times, or the same length of the microfluidic channel.
  • The may have a width of, for example, 50 microns, 100 microns, or 200 microns.
  • the method may include, in some embodiments, flowing a third fluid in the microfluidic channel simultaneously with the first and second fluids.
  • the second fluid may be flowed between the first and third fluids.
  • the third fluid is a gel precursor, and, optionally, the method may further comprise forming a gel comprising substantially all of the third fluid in the microfluidic channel.
  • the method may further comprise forming a gel comprising substantially all of the second fluid in the microfluidic channel.
  • the forming step may comprise applying heat to the microfluidic channel, for example, the gel may be formed at 37 degrees Celsius.
  • the forming step comprises crosslinking by the reaction of at least two reactants.
  • the method may further comprising forming a gradient of at least one component, e.g., a soluble factor, in the gel.
  • An article may be formed by the method described above. In another embodiment, an article is provided.
  • the article comprises a microfluidic channel and a first structure comprising a thermocurable gel that occupies a portion, but not all of a microfluidic channel.
  • the structure extends the length of the channel, wherein the thermocurable gel includes at least a first side and a second side that are not in contact with a surface of the channel.
  • a method is provided. The method comprises providing a microfluidic channel including a first cell disposed in a first gel portion and a second cell disposed in a second gel portion. The first and second gel portions are separated by a portion that does not contain cells. At least one gel portion comprises a thermocurable gel. The method also involves allowing interaction between the first and second cells.
  • Fig. IA shows the partitioning of a microfluidic channel, which can be used to form one or more gel structures, in one embodiment of the invention.
  • Fig. IB shows a photograph of an experimental set-up for the partitioning system shown in Fig. IA according to one embodiment of the invention.
  • Fig. 1C is a schematic diagram of the experimental set-up shown in Fig. IB according to one embodiment of the invention.
  • Fig. ID is a schematic diagram of an incubator that may be employed in some embodiments of the invention.
  • Figs. IE- II show various examples of gel cross-sections according to one embodiment of the invention.
  • Figs. 2A-2C show various designs of microchannels according to one embodiment of the invention.
  • Fig. 2D shows an example of a cross-sectional view of a microchannel according to one embodiment of the invention.
  • Fig. 3 is a photograph showing laminar flow in a microchannel when the liquids have the same viscosity according to one embodiment of the invention.
  • Figs. 4A-4C are photographs showing laminar flow in a microchannel when the liquids have different viscosities according to one embodiment of the invention.
  • Figs. 5A-5F show various examples of gel structures formed in microfluidic channels according to one embodiment of the invention.
  • Figs. 6A-6B show cells disposed within gel structures according to one embodiment of the invention.
  • Figs. 7A-7D show various examples of arrangements of subchannels within microchannels according to one embodiment of the invention.
  • Fig. 8A is a schematic diagram of a five-inlet channel according to one embodiment of the invention.
  • Fig. 8B shows one example of a gel structure that may be formed when employing a five-inlet channel according to one embodiment of the invention. The figure also shows cells embedded in the gel structure.
  • Fig. 8C shows migration of cells cultured within a gel structure according to one embodiment of the invention.
  • Figs. 9A-9D show gradients that may be produced in microchannels according to one embodiment of the invention.
  • Fig. 9E is a graph of the fluorescence intensity of the gradients shown in Figs. 9A-9D according to one embodiment of the invention.
  • Fig. 10 is a table of results collected from a study directed at shearing cells, according to one embodiment of the invention.
  • Methods described herein may include the use of spatially-defined flows of fluid within microchannels to form portions of gel (e.g., gel structures) inside the microchannels.
  • the gel structures may be formed by flowing, e.g., laminarly, one or more streams of fluid in a microfluidic channel, at least one of the streams including a gel precursor.
  • the stream(s) of gel precursor can be polymerized (e.g., gelled) to form one or more gel structures by various methods such as by application of heat.
  • the dimensions of the gel structures may be varied, for example, by applying different flow rates to the fluid streams, choosing different viscosities of the fluids, and/or by varying the dimensions of the microchannel.
  • different configurations of gel structures in microfluidic channels can be formed.
  • a stream of gel precursor is flanked by two streams of spacing solution (e.g., non-gel precursors), which may be flowed into a microchannel simultaneously.
  • spacing solution e.g., non-gel precursors
  • a self-standing gel structure may be formed between the two streams of spacing solution inside the microchannel.
  • the spacing solutions may be rinsed out and replaced by other solutions.
  • Gel structures having different components encapsulated therein may also be formed.
  • Articles and methods described herein may involve, in some embodiments, i) culturing cells within or on surfaces of the gel structures, ii) patterning different types of cells on or in adjacent gel structures, and/or iii) applying gradients of soluble factors across the cell-containing gel structures.
  • Such structures may be used for studying intercellular communication between cells cultured within biologically-derived, 3-D matrices of microscopic size.
  • a method in one aspect of the invention, includes flowing a first fluid and a second fluid in a microfluidic channel, at least one of the first and second fluids being a gel precursor.
  • a third, fourth, or fifth, etc. fluid may also be flowed in the channel.
  • the gel precursor may be polymerized to form a gel structure that occupies a portion, but not all of the volume of the channel.
  • device 10 includes a microfluidic channel 12 containing fluid streams 16, 18, and 20 which form portions, or subchannels, 26, 28, and 30, respectively. Fluid streams 16, 18, and 20 may be flowed simultaneously in microfluidic channel 12 so as to form multiple, laminar streams of fluid.
  • At least one fluid may flow parallel to the walls of the microchannel, such that there is little or no turbulence between the adjacent streams of fluid. In some such embodiments, mixing between the streams occurs only by diffusion.
  • at least one of fluid streams 16, 18, and 20 contains a gel precursor. That is, all or a portion of the gel precursor-containing fluid stream can be polymerized to form a gel structure.
  • the gel structure has a substantially similar configuration and dimensions of the gel precursor used to form the structure.
  • the gel structure may occupy a portion, but not all, of the volume of the microfluidic channel.
  • fluid streams 16 and 20 are non-gel precursors, also referred to herein as spacing solutions.
  • subchannel 28 may contain a gel structure while the spacing solutions remain unpolymerized. These non-polymerized portions can be used to deliver fluids and/or components to one or more sides of the gel structure.
  • fluid stream 18 can be formed into a gel such that at least a first side and at least a second side of the gel is not in contact with a surface of the channel (e.g., the sides of fluid stream 18 that are in contact with fluid stream 16 and fluid stream 20 are not in contact with a surface of the channel, and therefore, the sides of the gel that forms from fluid stream 18 are also not in contact with a surface of the channel).
  • fluid stream 16 and/or fluid stream 20 may include a gel precursor so as to form gels in subchannels 26 and/or 30 of microfluidic channel 12.
  • the gels may be formed of the same or a different polymeric composition.
  • the difference in the polymeric compositions may be attributed to the addition of cells or other components, the type of material used to form the gel, the degree of crosslinking of the gel, or the like.
  • the term "gel” is given its ordinary meaning in the art and refers to a material comprising a polymer network that is able to trap and contain fluids.
  • the gel may comprise polymer chains that are crosslinked, either directly or via a crosslinking agent.
  • the degree of crosslinking may be varied, in some cases, to tailor the extent to which the gel absorbs or retains fluids.
  • the gel precursor comprises a material that forms a gel upon reaction of at least two components.
  • the gel precursor comprises a material that forms a gel upon application of light to the material. Different types of gels that can be used in accordance with the present invention are described in more detail below.
  • a gel precursor comprises a material that forms a thermocurable gel upon the application of heat.
  • all or a portion of the gel precursor is exposed to an environment having a first temperature prior to forming a gel and a second temperature during formation of the gel.
  • device 10 containing the gel precursor may be kept at a first temperature prior to and/or during flowing of the gel precursor in the channel.
  • the gel precursor may be exposed to an environment having a second temperature. The second temperature may be greater than the first temperature.
  • Figs. IB-ID describe one method for forming a device including a microfluidic channel and a gel structure according to one embodiment of the invention.
  • a device 10 including a microchannel may be placed on a thermally-conductive disc 32, which may facilitate conduction of heat away from or towards the microchannel.
  • a thermally-conductive disc 32 may facilitate conduction of heat away from or towards the microchannel.
  • One or more outlet(s) 33 of the microchannel may be connected to a syringe pump via tubing 34.
  • the microchannel may be formed from a polymer 35 positioned on a surface 36 (e.g., a glass surface).
  • surface 36 may be positioned on top of thermally-conductive disc 32 and a material 37, e.g., tissue paper, may be positioned between the surface and the thermally- conductive disc.
  • the thermally-conductive disc may keep the device at a temperature below the gelling temperature of the gel.
  • Material 37 may aid in the visualization and/or identification of certain components in the system, as discussed below.
  • the conductive disc may be an aluminum plate that is cooled, for example, by ice 38.
  • Fluid streams may be introduced into the microfluidic channel(s) by any suitable method.
  • the fluid streams are flowed into the microfluidic channel by applying a reduced pressure (e.g., vacuum) at outlet 33.
  • a droplet of gel precursor and/or of spacing solution is deposited at each of inlets 41-45.
  • the solutions may be pulled into the channel by applying a vacuum to the outlet of the channel using a syringe 39 or other suitable device; however, in other embodiments, a positive pressure may be applied to one or more inlets of the device, as described in more detail below.
  • the tubing at the outlet may be disconnected from the syringe to immediately stop the flow. This may be done, for example, by cutting the tubing with scissors.
  • the gel precursor may be allowed to gel after the flow has stopped, or the act of polymerization itself may cause the flow to stop.
  • polymerization of the gel may be performed by placing the conductive disc in an incubator or sterile bath 53 having a temperature suitable for crosslinking the gel precursor.
  • other methods of polymerization may be used, such as exposure of the device to light to initiate polymerization of a photopolymerizable gel precursor.
  • the inlets for delivering a gel precursor may become plugged and may no longer pass fluid.
  • the inlets used for delivering spacing solutions may remain open and could be used afterwards to access the parts of the channel not filled with gel.
  • the inlets for delivering a gel precursor are not plugged; for example, the gel may be crosslinked only at a main portion of a channel and not at the inlets. Such local crosslinking may be performed for thermocurable gels, for example, by applying heat locally to certain portions of the channel.
  • inlets that are plugged with a gel precursor can become unplugged, e.g., by removing all or portions of the gel.
  • Removal can be performed, for example, by melting a gel portion (e.g., by placing the gel into a cooler environment, e.g., 4 degrees Celsius to liquefy the gel), dissolving a gel portion (e.g., using an enzyme), and/or physically removing a gel portion (e.g., by cutting).
  • a solution of enzyme such as a collagenase (e.g., dispase) can be introduced into a fluid stream adjacent a gel and the enzyme can diffuse into the gel.
  • the enzyme solution can be introduced into an inlet of a channel containing a gel structure and the enzyme can be allowed to diffuse into the gel. It should be understood that all or portions of a gel structure can be removed in a microfluidic channel (e.g., a main channel) by using such or other methods.
  • such and other methods can remove all or portions of a gel structure while maintaining integrity of any components embedded in the gel structure. For instance, where cells are embedded in a gel structure, removal of a gel can be performed without substantially changing the morphology of the cell, without causing the cell to die, and/or without causing denaturation of any components.
  • substantially all of a fluid (e.g., a gel precursor) in a channel is used to form the gel.
  • a gel precursor in fluid stream 18 may form a gel structure in the form of subchannel 28 upon polymerization of the precursor, and substantially all of the fluid of fluid stream 18 present in the channel may be used to form this gel structure.
  • substantially all of the two or more fluids present in the channel form a gel in the channel.
  • different portions of the gel across the width of the channel can be formed simultaneously and are not limited by diffusion.
  • a gel structure is formed at the interface between first and second fluids.
  • a first fluid stream may include a first gel precursor and a second fluid stream may include a second gel precursor, the first and second gel precursors forming a gel structure upon interaction of the precursor components.
  • first and second fluids present in the microchannel are used to form the interfacial gel structure.
  • the gel precursor that does not interact with a necessary component to form the gel flows out of the channel.
  • different portions of the gel across the width of the channel are not formed simultaneously since the gel first forms at the interface of the precursor fluids, and then the gel can become wider as diffusion of the precursor takes place.
  • the number of inlets and/or outlets of the channel and/or the distribution of solutions at the inlets may be additional factors that influence the final arrangement of gel structures inside the channel.
  • the distribution of gel precursor solution(s) and spacing solution(s) can be arranged such that first and second gel structures are positioned immediately adjacent to one another or adjacent to one another via a non-gel portion.
  • fluid streams 16 and 18 include gel precursors, and upon crosslinking of the gels, the gels in subchannels 26 and 28 are formed immediately adjacent to one another.
  • Subchannel 30 may include a fluid stream in one embodiment; in another embodiment, fluid stream 20 may be used to form a solid or other non-gel structure. In some cases, fluid stream 20 is replaced with a gas (e.g., air).
  • Microfluidic channels including a first and a second gel may, therefore, be positioned directly adjacent to one another, or may be separated from one another by non-gel component(s). As described in more detail below, a series of gel structures can be formed within a single microfluidic channel by methods described herein.
  • a gel structure formed in a microfluidic channel may extend the length of the channel. In other embodiments, however, a gel structure may be formed at discrete points along the channel.
  • a channel may include discrete conductive elements (e.g., electrodes, films, meshes, and the like) that can cause local polymerization of a thermocurable gel precursor.
  • the conductive elements may be positioned at a surface of the channel or may extend into the channel, and may be separated from one another by non-conductive elements.
  • FIG. 1E-1H show exemplary cross sections of a microfluidic channel containing a gel structure.
  • gel structure 40 may extend from a top surface 46 to a bottom surface 48 of microfluidic channel 12.
  • Portions 50 and/or 54 of microfluidic channel 12 may include a fluid, a second gel structure, or a solid, in some embodiments.
  • microfluidic channel 12 may include a gel structure 40 that is not in physical contact with top portion 46, bottom portion 48, side portion 60, or side portion 62 of the microfluidic channel at least one point along the length of the channel.
  • Such an embodiment may be formed, for example, by flowing top and bottom streams, as well as side streams, so as to encapsulate gel structure 40.
  • Fig. IG shows another example of a gel structure 40 that occupies a portion, but not all of a microfluidic channel. As illustrated in Fig. IH, gel structure 40 may be formed such that it is in contact with a side portion 62 of a microfluidic channel. A gel structure 40 may be in contact with a single bottom (or top) portion, as shown in Fig. II. It should be understood that Figs. IE- II re exemplary and that other configuration of gels can be formed in microfluidic channels using methods described herein.
  • all or a portion of the microfluidic channel can be removed, e.g., leaving behind a gel structure positioned on a surface.
  • the gel structure can then be manipulated as desired. For instance, in one embodiment, a second microfluidic channel is positioned on top of or adjacent to the at least one gel structure. In another embodiment, the gel structure is encapsulated in a second gel.
  • a gel structure in a microfluidic channel may have any suitable width.
  • the width of the gel structure may be, for example, at least 25 microns, at least 50 microns, at least 100 microns, at least 200 microns, at least 300 microns, at least 500 microns, or at least lmm.
  • the gel may have any suitable length within a microfluidic channel. In some cases, the gel may be the same length than that of the microfluidic channel. In other embodiments, the gel may have a different length than that of the microfluidic channel. For example, the gel may have a length of at least 0.25 times, at least 0.50 times, or at least 0.75 times the length of the microfluidic channel.
  • the width of a microfluidic channel may be chosen, in part, by the number of inlets in fluid communication with the channel.
  • Fig. 2D shows an example of a cross-sectional view of a microchannel 10.
  • the dimensions of the microfluidic channels and/or the operating conditions are chosen such that the Reynolds number is less than 2000, less than 1000, less than 100, or less than 1.
  • Laminar flow may be defined when the Reynolds numbers is less than about 2000.
  • the Reynolds number is the ratio of internal forces to viscous forces and consequently it quantifies the relative importance of these two types of forces for given flow conditions.
  • the Reynolds numbers for an aqueous solution flowing at 15 ⁇ L/min in these microchannels may be less than 1 and the resulting flows may be laminar. Matching the viscosity of the precursor gel and the spacing solutions may minimize the dependence of the width of the streams flowing in microchannels upon the differences in viscosity between the fluids forming each stream, as discussed herein.
  • one or more fluids may be introduced and/or flowed into a channel by applying a reduced pressure (e.g., vacuum) at one or more outlets of the device.
  • the inlet channels may comprise one or more wells, which can act as reservoirs for one or more fluids.
  • Fluid(s) such as a gel precursor or a spacer solution, may be introduced into each well and upon application of a reduced pressure at the outlet, the fluids can flow into the channel.
  • the reduced pressure may be applied using a syringe pump, a syringe, a microfluidic pump, or any other suitable device.
  • one or more fluids may be introduced and/or flowed in a channel by applying positive pressure at one or more inlets.
  • the positive pressure may be applied using a syringe pump, a syringe, or a microfluidic pump, for example.
  • the fluids may continue to flow while the gel is being cured.
  • the gel forming step may be initiated and/or completed while at least one of the fluids (e.g., the gel precursor or a non-gel precursor) is flowing. In other cases, the gel forming step is initiated and/or completed while at least one of the fluids is not flowing.
  • the width and/or configuration of the gel structure(s) formed in a microfluidic channel may be varied by a variety of methods including viscosity and flow rate.
  • the width of a gel structure is varied by applying different flow rates to fluid streams 16, 18, and 20 of Fig. IA.
  • the fluid streams may have substantially the same width across microfluidic channel 12. If a high flow rate is applied to fluid stream 18 compared to fluid streams 16 and 20, fluid stream 18 may have a larger width than fluid streams 16 and 20.
  • a gel structure may be formed in subchannel 28 so as to have a larger width than subchannels 26 and 30.
  • a narrower gel structure in the form of subchannel 28 may be formed compared to subchannels 26 and 30.
  • Each of fluid streams 16, 18, and 20 may also be flowed at different flow rates to form gel structures having various dimensions.
  • a gel structure having any suitable width (and/or height) may be formed in a microfluidic channel.
  • the width of a microchannel may be chosen, at least in part, based on the differences in viscosity between the fluids forming each stream. In certain embodiments, where the viscosity of the gel precursor is greater than the viscosity of the spacer solution, the width of the gel precursor subchannels may increase with increasing flow rate. In certain embodiments where the viscosity of the precursor gel is less than the viscosity of the spacer solution, the width of the gel precursor subchannels may decrease with increasing flow rate. As a specific example, as shown in the illustrative embodiment of Fig. 3, a four inlet system was designed wherein two inlets were filled with spacer solutions (251 and 253), and the other two inlets were filled with spacer solutions plus dye (250 and 252).
  • Fig. 4A shows a three inlet system wherein the center inlet 302 includes a more viscous material than the spacer solutions 301 and 303. Particularly in this case, the viscosity of the center fluid is approximately three times that of the spacer fluid.
  • the flow rate Fl of the fluid in Fig. 4A is greater than the flow rate F2 of the fluid in Fig. 4B, which is greater than the flow rate F3 of the fluid in Fig. 4C.
  • Fig. 5 shows various examples of gel structures formed in microfluidic channels. Schematic diagrams of the distribution of flowing solutions (e.g., precursor gel and spacing solution) to form one (Fig. 5A) or two (Fig. 5B) gel structures in microchannels are also shown.
  • spacing solutions 65 and 67 may be flowed on either side of gel precursor 66.
  • the example shown in Fig. 5B shows spacing solutions 68, 70 and 72 being flowed in conjunction with precursor gel solutions 69 and 71.
  • Fig. 5E and Fig. 5F show the portions of gel 66, 69, and 71 dividing microchannel 12 after curing the gel portions into subchannels that were accessible through the corresponding inlets.
  • the gels may be cured after or during flow of the gel precursor solutions .
  • a suspension of fluorescent 50-nm Nile Red polystyrene beads 73 and 50-nm Yellow Green polystyrene beads 74 were added to the spacing solutions.
  • the beads remained in their respective subchannel compartments.
  • the beads can remain in their respective subchannel compartments for various amounts of time (e.g., more than 30 minutes, 1 hour, 1 day, 1 week, etc.) as the suspensions are flowing or after stopping the flow of the fluids.
  • the present invention provides methods for i) culturing cells either within the gel structures or on one or more surfaces of the gel structures, ii) patterning different types of cells in adjacent gel structures, and iii) applying gradients of soluble factors across the gel structures containing cells.
  • culturing cells either within the gel structures or on one or more surfaces of the gel structures, ii) patterning different types of cells in adjacent gel structures, and iii) applying gradients of soluble factors across the gel structures containing cells.
  • other components such as nanoparticles, beads, soluble components, or other entities may be used in place of or in conjunction with cells in certain embodiments.
  • Much of cell biology has developed based on studies of cells grown in glass or polystyrene Petri dishes. The limitations of these systems, and their differences from 3D, in-vivo biology, are well accepted but the need to produce useful systems that provide a practical, and more biologically-relevant environment for cell and tissue culture is still required.
  • In-vivo cellular behavior is complex: cells integrate and respond to numerous signals that originate both from their local microenvironment, and from distant sources. These signals come from direct contact with other local cells (such as gap junctions in myocytes), from soluble factors secreted by neighboring or distant cells (such as growth factors and hormones), and from the surrounding extracellular matrix (ECM), which contributes both chemical and mechanical signals. Intercellular communication is necessary for coordinating collective responses among multiple cells.
  • One example is the type of inflammatory response in the immune system in which macrophages, recruited to a site of injury, secrete TNF (tumor necrosis factor) to activate leukocytes.
  • TNF tumor necrosis factor
  • a second example occurs during wound healing, where fibroblasts and epidermal cells secrete IGF (insulin-like growth factor) to induce the formation of granulation tissue and the reepithelialization of the wound.
  • the gel structures described herein and methods associated therewith can overcome the above-mentioned challenges and may allow some or all of the following features: i) the cultivation of mammalian cells in 3D environments; ii) exposing these cells to chemical signals, especially gradients; iii) observing them (using optical microscopy) in real time; iv) separating them from other cells; v) the use of a matrix that is relevant to in-vivo cell biology.
  • a microfluidic channel including a gel disposed therein includes at least one cell.
  • the cell may be disposed in or on a gel portion of the channel, or in or on a non-gel portion of the channel.
  • microfluidic channel 100 includes portions, or subchannels 110, 112, and 114.
  • Subchannel 112 may include a gel 113 which may optionally include one or more cells disposed therein.
  • Subchannel 114 may include, for example, a fluid, optionally having one or more cells disposed therein.
  • the cells in subchannel 114 are attached to at least one surface of microfluidic channel 100.
  • the surface portion of the channel having cell(s) attached thereto in one embodiment, is not in physical contact with gel 113.
  • the cells attached to a surface of the channel such as those shown in Fig. 6A, may have a different morphology than the cells that are disposed in the gel, as shown in Fig. 6B.
  • interactions between cells that have different morphologies can be determined.
  • NIH/3T3 fibroblasts were cultured on the surface of a subchannel 114 adjacent to gel 113.
  • Fig. 6B shows a phase-contrast image of NIH/3T3 fibroblasts after two days in culture within gel in a microchannel.
  • cells can be allowed to interact with one another, with a chemical substance, and/or with a physical force, e.g., under flowing conditions or non-flowing conditions.
  • a change in characteristic of at least one cell as a result of the interaction(s) may be determined.
  • gel 113 including at least one cell disposed therein may be adjacent a fluid flowing in subchannel 110.
  • the flowing stream of fluid may have a suitable flow rate so as to apply a shearing stress to the at least one cell disposed in gel 112.
  • a characteristic of a cell as a result of the applied shearing stress may be determined.
  • a change in morphology of a cell may be determined using articles and methods described herein.
  • the effect of exposing a cell to a chemical substance can be determined.
  • a chemical substance e.g., a toxin
  • such effects can be determined controllably while the cells are positioned within biologically-derived, 3-D matrices of microscopic size that can mimic in vivo systems.
  • an article comprises a microfluidic channel including at least one first cell and at least one second cell, the cells associated with a surface of the channel and/or a gel structure within the channel.
  • the microfluidic channel may comprise a first gel including at least a first cell disposed therein and a second gel including at least a second cell disposed therein.
  • the first and second cells may be the same cells in some embodiments, or may be different cells in other embodiments.
  • the first and second cells have at least one different characteristic (e.g., different cell surface receptors, different exposure to a substance, etc.).
  • a microfluidic channel includes at least two gel structures, e.g., a first and a second gel structure
  • the first and second gel structures may be positioned directly adjacent to each other.
  • the first and second gel structures are not positioned directly adjacent to each other; e.g., at least one liquid and/or an intervening gel structure may be positioned between the first and second gel structures.
  • Additional channel portions or subchannels within the microchannel may be or comprise a liquid (e.g., in the form of a fluid stream) or a gel, which may or may not comprise cells.
  • the additional subchannels with the microchannel may be formed of the same substance as the first and second gel structures, or may be formed of different substances.
  • Fig. 7A In instances where three subchannels are formed within a microchannel, non- limiting examples of arrangement of the substances within the subchannels are shown in Fig. 7A. Similarly, when four or five subchannels are formed within a microchannel, non-limiting examples of arrangement of the substances within the subchannels are given in Fig. 7B or 7C, respectively.
  • Fig. 7D depicts non-limiting arrangements for five subchannels formed within a microchannel and when three cell types are present.
  • a method involving such and other structures may include allowing interaction between the first and second cells, and determining a change in characteristic of at least one cell as a result of the interaction.
  • microfluidic channel 160 may include subchannels 162, 164, 166, 168, and 170, which may be formed by the laminar flow of fluids in each of the channels.
  • subchannel 162 includes a fluid and subchannel 164 includes a first gel (or first gel precursor) including a first cell or set of first cells 163 disposed therein.
  • Subchannel 166 includes a second gel (or second gel precursor) that does not include a cell disposed therein.
  • Subchannel 168 includes a third gel (or third gel precursor) including a second cell or set of second cells 165 disposed therein, and subchannel 170 includes a fourth gel (or fourth gel precursor) that does not comprise a cell disposed therein.
  • the gel precursors can be crosslinked to form gels in the respective subchannels (Fig. 8B).
  • the first and second cells can be allowed to interact with one another by allowing the cells to migrate from one subchannel to another.
  • the cells may migrate between different gel portions, from gel portions to non-gel portions, or from non-gel portions to gel portions.
  • Such experiments can be done under different conditions, such as under flowing conditions, non-flowing conditions, without nutrients (e.g., conditions that starve the cells), or with various buffers, solvents, or other components (e.g., drugs or toxins).
  • BAC cells macrophage-like BAC1.2F5 cells
  • monocyte-derived LADMAC cells BAC cells
  • BAC cells require the presence of CSF-I, a growth factor secreted by LADMAC cells that stimulates the survival and proliferation of macrophages.
  • a five-inlet microchannel was fabricated, such as the one shown in Fig. 8.
  • subchannels 162, 164, 166, 168 and 170 were filled with five differing solutions, for example, a spacer fluid in subchannel 162 and gel-precursor fluids in the four adjacent subchannels.
  • the gel portions contained: i) BAC cells embedded in a gel within subchannel 164, ii) a gel portion without cells in subchannel 166 iii) a gel portion containing LADMAC cells in subchannel 168, and another gel portion without cells in subchannel 170.
  • the spacing solution in the subchannel may be replaced with culturing medium (for example, alpha MEM supplemented with 10% newborn calf serum and 36 ng/ml of recombinant human CSF-I).
  • the microchannel may then be incubated. In this example, after 12 hours of incubation, the cultering medium lacking recombinant CSF-I (starving medium) may be injected into subchannel 162.
  • the only source of CSF-I available to the BAC cells is the nearby LADMAC cells.
  • the starving medium in the channels may be renewed (for example, daily), and in the presence of LADMAC cells, BAC cells were viable for a week in the channel.
  • a similar microchannel was prepared as mentioned above: i) BAC cells embedded in subchannel 164 ii) a gel portion without cells in subchannel 166 iii) a gel portion without cells in subchannel 168 and iv) another gel portion without cells in subchannel 170.
  • the BAC cells died within two days after the addition of starving media. These example may confirm that the presence of LADMAC cells was necessary for the proliferation of BAC cells in starving medium.
  • a gradient of components can be formed in at least one gel structure positioned in a microfluidic channel.
  • microfluidic channel 180 may include subchannels 182, 184, and 186 that may be formed, for example, by laminar flow.
  • subchannel 184 includes a gel structure and subchannels 182 and 186 includes one or more fluids.
  • the fluid in subchannel 182 may include a first component and the fluid in subchannel 186 may include a second component different from the first component.
  • a gradient may be formed across the width of the gel structure.
  • subchannel 182 including the first component may have a higher concentration of the first component near portion 190 of the gel structure compared to portion 192 of the gel structure.
  • subchannel 186 including the second component may include a higher amount of the second component at portion 192 of the gel structure compared to portion 190 of the gel structure.
  • the gel structure can, therefore, comprise two different concentration gradients at different portions of the structure.
  • the gradient formed in the gel structure may depend on factors such as, for example, the cross linking density of the gel, and the physical characteristics of the first and second components such as the size and molecular weight of the first and second components. For instance, as shown in Fig.
  • a high molecular weight component in subchannel 182 may diffuse slower through the gel structure compared to a low molecular weight component, as shown in Fig. 9D, all other factors being equal.
  • the flow rates of the fluids may be the same, for example, 5 ⁇ L/min. In other cases, the flow rate of the fluids in adjacent channels may be different.
  • FIG. 9B, 9C, and 9D show fluorescent images of the microchannel after injecting the solutions into the subchannels for 300 minutes.
  • Fig. 9E shows the fluorescence intensity profiles reached by diffusion of TRITC-dextran with different molecular weights (after 300 minutes) plotted against the width of the channel.
  • the calculated values of the diffusion constant for 40-kDa, 70- kDa, and 155-kDa TRITC-dextran through the gel structures (Matrigel) were determined to be 0.45, 0.34, and 0.18 ⁇ m 2 /s, respectively. Equations for calculating the diffusion constants are given below.
  • gel structures can be act as barriers to molecules having a size of greater than or equal to 30 kD, 50 kD, 75 kD, 100 kD, 150 kD, 200 kD, or 300 kD, which sizes can be modified by the particular type of gel, the crosslinking density, etc.
  • gel structures may include polymer gels which are typically characterized by long chain polymer molecules that are crosslinked to form a network. This network can trap and hold fluid, which can give gels properties somewhere between those of solids and liquids.
  • gel structures may include polymer gels which are typically characterized by long chain polymer molecules that are crosslinked to form a network. This network can trap and hold fluid, which can give gels properties somewhere between those of solids and liquids.
  • various properties of a particular gel can be tailored. For example, a highly crosslinked gel generally is structurally strong and a lightly crosslinked gel may be weaker structurally.
  • a gel described herein may be a gel, including a crosslinkable gel.
  • gel refers to water-soluble polymer chains that are crossl inked in the presence of water to form a network.
  • hydrogels may be suitable for use in certain embodiments of the invention. It should be noted that the list is not exhaustive, and those of ordinary skill in the art may readily select or form other suitable gel materials using available information regarding, for example, curing temperature of various materials, and/or using routine experimentation and simple screening tests.
  • polymers capable of forming hydrogels include, but are not limited to, silicon-containing polymers, polyacrylamides, crosslinked polymers (e.g., polyethylene oxide, poly AMPS and polyvinylpyrrolidone), polyvinyl alcohol, acrylate polymers (e.g., sodium polyacrylate), and copolymers with an abundance of hydrophilic groups.
  • the gel may be a sol-gel.
  • a "sol-gel” refers to a colloidal suspension capable of being gelled to form a solid.
  • the sol-gel may be formed from a mixture of solid particles (e.g., inorganic salts) suspended in a liquid, wherein a series of reactions including hydrolysis and polymerization reactions may be performed to form a colloidal suspension.
  • the gel may be an organogel, wherein the polymer may be swollen by addition of an organic solvent.
  • the gel is a thermocurable gel (i.e., a gel that can be cured by application of heat).
  • the gel is cured by raising the temperature from a lower temperature to a higher temperature, for example, 4 0 C to 37 0 C.
  • the temperature may be raised from a lower temperature to at least 15 0 C, 20 0 C, 22 0 C, 25 0 C, 30 0 C, 35 0 C, 40 0 C, or to any other suitable temperature that may cause the gel to cure partially or completely.
  • the temperature is chosen in part such that the cells contained within the gel remain viable.
  • the gel may be returned to a gel precursor by altering the temperature. For instance, in some cases the temperature may be decreased, e.g., to cause at least a portion of the gel to return to a more fluid state. In one particular example, the temperature may be decreased from 37 0 C to 4 0 C. By redissolving the gel, the cells or other entities that are in the gel may be recovered.
  • the gel may be returned to a gel precursor liquid and the cell(s) may be collected and/or analyzed (e.g., counted, subjected to certain techniques such as Western Blotting, further expanded in cell culture).
  • the gel is a natural gel; that is, a biologically-derived gel.
  • a natural gel may include, for example, collagen I, collagen IV, fibrin, laminin, and combinations thereof.
  • Matrigel MG
  • MG Matrigel
  • Benefits that may be attributed to MG and other gels useful in embodiments described herein include, but are not limited to, i) it is biologically-derived as it is a soluble extract of the basement membrane of murine tumoral epithelia, mainly composed of laminin and collagen IV; ii) it is commercially available, and has been used extensively both in vivo and in vitro in the culture of many types of cells; iii) it is liquid at 4 0 C, but gels within 15 minutes at 22 0 C - 37 0 C, and iv) it is sufficiently stiff after gelling to form self-standing structures of gel.
  • Matrigel may be employed in its purified form, i.e. Growth Factor Reduced Matrigel.
  • Non-limiting examples of crosslinkable polymers include: polyvinyl alcohol, polyvinylbutryl, polyvinylpyridyl, polyvinyl pyrrolidone, polyvinyl acetate, acrylonitrile butadiene styrene (ABS), ethylene-propylene rubbers (EPDM), EPR, chlorinated polyethylene (CPE), ethelynebisacrylamide (EBA), acrylates (e.g., alkyl acrylates, glycol acrylates, polyglycol acrylates, ethylene ethyl acrylate (EEA)), hydrogenated nitrile butadiene rubber (HNBR), natural rubber, nitrile butadiene rubber (NBR), certain fluoropolymers, silicone rubber, polyisoprene, ethylene vinyl acetate (EVA), chlorosulfonyl rubber, flourinated poly(arylene ether) (FPAE
  • the polymeric structure may comprise polyvinyl pyridine or quaternized polyvinyl N- methyl-pyridine.
  • polyvinyl pyridine or quaternized polyvinyl N- methyl-pyridine.
  • gels described herein include one or more types of particles such as cells (e.g., mammalian or bacterial), beads, nanoparticles, nanotubes, colloids, etc.
  • the particles may have any suitable particle size.
  • particle size refers to the largest characteristic dimension (i.e. of a line passing through the geometric center of the particle e.g., diameter) that can be measured along any orientation of a particle (e.g., a polymer particle).
  • Particle size as used herein may be measured or estimated, for example, using a sieve analysis, wherein particles are passed through openings of a standard size in a screen. The particle-size distribution may be reported as the weight percentage of particles retained on each of a series of standard sieves of decreasing size, and the percentage of particles passed of the finest size. That is, the average particle size may correspond to the 50% point in the weight distribution of particles.
  • methods described herein involve binding between at least two entities.
  • binding refers to the interaction between a corresponding pair of components that exhibit mutual affinity or binding capacity, typically specific or non-specific binding or interaction, including biochemical, physiological, and/or pharmaceutical interactions.
  • Biological binding defines a type of interaction that occurs between pairs of components including proteins, (e.g., adhesion proteins), nucleic acids, glycoproteins, carbohydrates, hormones and the like.
  • cell e.g., cell surface receptor
  • adhesion protein/integrin e.g., cell surface receptor
  • antibody/antigen e.g., antibody/hapten
  • enzyme/substrate enzyme/inhibitor
  • enzyme/cofactor binding protein/substrate
  • carrier protein/substrate e.g., lectin/carbohydrate
  • receptor/hormone e.g., receptor/effector
  • complementary strands of nucleic acid e.g., cell surface receptor
  • protein/nucleic acid repressor/inducer e.g., ligand/cell surface receptor
  • virus/ligand e.g., virus/ligand, etc.
  • binding of a sample component includes interaction between two or more different corresponding pairs of components that exhibit mutual affinity or binding capacity, such as two or more of the corresponding pairs listed above.
  • the interaction of at least two corresponding pairs of binding partners e.g., binding of both an adhesion molecule and an antibody with the sample component, can cause effective recognition and capture of the sample component from a solution, resulting in immobilization of the component at a surface.
  • binding of a sample component may include interaction between an integrin (a receptor in the plasma membrane of the cell) and an adhesion molecule (e.g., ICAM-I and/or LF A-3), as well as interaction between an antibody of the cell (e.g., anti-CD3, anti-CD4, anti-CD8, and/or anti-CD5) and its corresponding antigenic binding partner (e.g., CD3, CD4, CD8, and/or CD5, respectively).
  • an integrin a receptor in the plasma membrane of the cell
  • an adhesion molecule e.g., ICAM-I and/or LF A-3
  • an antibody of the cell e.g., anti-CD3, anti-CD4, anti-CD8, and/or anti-CD5
  • its corresponding antigenic binding partner e.g., CD3, CD4, CD8, and/or CD5, respectively.
  • Signaling entity means an entity that is capable of indicating its existence in a particular sample or at a particular location.
  • Signaling entities of the invention can be those that are identifiable by the unaided human eye, those that may be invisible in isolation but may be detectable by the unaided human eye if in sufficient quantity (e.g., colloid particles), entities that absorb or emit electromagnetic radiation at a level or within a wavelength range such that they can be readily detected visibly (unaided or with a microscope including an electron microscope or the like), optically, or spectroscopically, entities that can be detected electronically or electrochemically, such as redox-active molecules exhibiting a characteristic oxidation/reduction pattern upon exposure to appropriate activation energy (“electronic signaling entities”), or the like.
  • Examples include dyes, pigments, electroactive molecules such as redox-active molecules, fluorescent moieties (including, by definition, phosphorescent moieties), up- regulating phosphors, chemiluminescent entities, electrochemiluminescent entities, or enzyme-linked signaling moieties including horseradish peroxidase and alkaline phosphatase.
  • "Precursors of signaling entities” are entities that, by themselves, may not have signaling capability but, upon chemical, electrochemical, electrical, magnetic, or physical interaction with another species, become signaling entities.
  • An example includes a chromophore having the ability to emit radiation within a particular, detectable wavelength only upon chemical interaction with another molecule.
  • Precursors of signaling entities are distinguishable from, but are included within the definition of, "signaling entities" as used herein.
  • a material 37 for example, tissue paper
  • tissue paper may be positioned between conductive disc 32 and surface 36 to aid in the visualization and/or identification of the signaling entities.
  • the spacing solution in these experiments employed may be a solution of PEG.
  • Benefits that may be observed when using PEG include, PEG is nontoxic and biocompatible and for certain examples, the viability of certain cells cultured in the media may not be affected.
  • microfluidic refers to a device, apparatus or system including at least one fluid channel having a cross-sectional dimension of less than 1 mm, and a ratio of length to largest cross-sectional dimension of at least 3:1.
  • a "microfluidic channel,” as used herein, is a channel meeting these criteria.
  • the "cross-sectional dimension" of the channel is measured perpendicular to the direction of fluid flow.
  • Most fluid channels in components of the invention have maximum cross-sectional dimensions less than 2 mm, and in some cases, less than 1 mm.
  • all fluid channels containing embodiments of the invention are microfluidic or have a largest cross sectional dimension of no more than 2 mm or 1 mm.
  • the fluid channels may be formed in part by a single component (e.g., an etched substrate or molded unit).
  • larger channels, tubes, chambers, reservoirs, etc. can be used to store fluids in bulk and to deliver fluids to components of the invention.
  • the maximum cross-sectional dimension of the channel(s) containing embodiments of the invention are less than 500 microns, less than 200 microns, less than 100 microns, less than 50 microns, or less than 25 microns.
  • a "channel,” as used herein, means a feature on or in an article (substrate) that at least partially directs the flow of a fluid.
  • the channel can have any cross-sectional shape (circular, oval, triangular, irregular, square or rectangular, or the like) and can be covered or uncovered.
  • At least one portion of the channel can have a cross-section that is completely enclosed, or the entire channel may be completely enclosed along its entire length with the exception of its inlet(s) and outlet(s).
  • a channel may also have an aspect ratio (length to average cross sectional dimension) of at least 2:1, more typically at least 3:1, 5:1, or 10:1 or more.
  • An open channel generally will include characteristics that facilitate control over fluid transport, e.g., structural characteristics (an elongated indentation) and/or physical or chemical characteristics (hydrophobicity vs. hydrophilicity) or other characteristics that can exert a force (e.g., a containing force) on a fluid.
  • the fluid within the channel may partially or completely fill the channel.
  • the fluid may be held within the channel, for example, using surface tension (i.e., a concave or convex meniscus).
  • a channel may have form any suitable shape and may be, for example, linear, curved, serpentine, or may form a circle, square, or an irregular shape.
  • the channel may be of any size, for example, having a largest dimension perpendicular to fluid flow of less than about 5 mm or 2 mm, or less than about 1 mm, or less than about 500 microns, less than about 200 microns, less than about 100 microns, less than about 60 microns, less than about 50 microns, less than about 40 microns, less than about 30 microns, less than about 25 microns, less than about 10 microns, less than about 3 microns, less than about 1 micron, less than about 300 nm, less than about 100 nm, less than about 30 nm, or less than about 10 nm.
  • the dimensions of the channel may be chosen such that fluid is able to freely flow through the article or substrate.
  • the dimensions of the channel may also be chosen, for example, to allow a certain volumetric or linear flowrate of fluid in the channel.
  • the number of channels and the shape of the channels can be varied by any method known to those of ordinary skill in the art. In some cases, more than one channel or capillary may be used. For example, two or more channels may be used, where they are positioned inside each other, positioned adjacent to each other, positioned to intersect with each other, etc.
  • the geometry of the microfluidic channel may provide for the laminar flow of fluids through the channel, even at relatively high flow rates.
  • turbulent flow may be employed for example, by using even faster flow rates, wider channels, or devices such as microfluidic mixers. Such mixing may provide for a greater amount of contact between potential binding partners.
  • a channel may also include a number of different inlets and/or outlets.
  • a channel may have 1, 2, 3, 4, 5, 6, etc. inlets and/or outlets.
  • Microfluidic systems described herein may optionally include one or more platforms for performing chemical reactions, combining and separating fluids, diluting samples, and generating gradients, such as those described in US Patent No. 6,645,432, hereby incorporated by reference herein.
  • a microfluidic device described herein can be fabricated of a polymer, for example an elastomeric material such as poly(dimethylsiloxane) (PDMS) using rapid prototyping and soft lithography.
  • a high resolution laser printer may be used to generate a mask from a CAD file that represents the channels that make up the fluidic network.
  • the mask may be a transparency that may be contacted with a photoresist, for example, SU-8 photoresist (MicroChem), to produce a negative master of the photoresist on a silicon wafer.
  • a positive replica of PDMS may be made by molding the PDMS against the master, a technique known to those skilled in the art.
  • a flat substrate for example, a glass slide, silicon wafer, or polystyrene surface may be placed against the PDMS surface and may be held in place by van der Waals forces, or may be fixed to the PDMS using an adhesive.
  • holes e.g., 1 millimeter in diameter
  • tubing for example of polyethylene, may be sealed in communication with the holes to form a fluidic connection.
  • the connection may be sealed with a sealant or adhesive such as epoxy glue. Examples of methods of manufacturing a microfluidic device are provided in U.S. Patent No. 6,645,432, incorporated by reference in its entirety herein.
  • the microchannels may be fabricated by soft lithography using standard protocols to generate replicas of a mold.
  • the mold may be chosen from any common lithography material which includes, but is not limited to, poly(dimethylsiloxane) (PDMS).
  • PDMS poly(dimethylsiloxane)
  • the replicas may be sealed to glass slides irreversibly.
  • the replicas-on-glass microchannels may be sterilized by rinsing with a selected solution which may include, but is not limited to, 70% ethanol and/or sterile phosphate-buffered saline (PBS).
  • PBS sterile phosphate-buffered saline
  • NIH/3T3 fibroblasts were obtained from American Type Culture
  • Monocyte-derived LADMAC cells which constitutive Iy secrete colony stimulating factor- 1 (CSF-I), were purchased from ATCC (CRL-2420). The frozen aliquot was thawed in a 37 0 C water bath, transferred to a 25-mm 2 tissue-culture flask containing 13 ml of culture medium 10 (Eagle's Minimum Essential Medium (MEM) with 1.5 g/1 sodium bicarbonate and 10% fetal bovine serum), and cultured at 5% CO 2 . These weakly-adherent cells were passaged every 3 days by resuspending about 20% of the volume within the flask in fresh culturing medium in a new 25-mm 2 flask.
  • MEM Minimum Essential Medium
  • BAC1.2F5 macrophages (donated by Dr. E. R. Stanley, Albert Einstein College of Medicine, New York) were cultured at 5% CO 2 in alpha MEM (Eagle's Minimum Essential Medium, Alpha modification; Invitrogen) supplemented with 10% newborn calf serum (Invitrogen) and 36 ng/ml recombinant human CSF-I (also a gift of R. Stanley). The culture medium was replaced every 2 days to renew. At 70% confluence, BAC cells were gently scraped from the tissue-culture treated flask (Falcon) with a cell scraper (Falcon) and replated at -30% confluence. BAC cells were not cultured for more than 20 passages.
  • BAC and LADMAC cells were co-cultured in alpha MEM (Eagle's Minimum Essential Medium, Alpha modification; Invitrogen) supplemented with 10% newborn calf serum (Invitrogen) and 36 ng/ml recombinant human CSF-I or alpha MEM (Eagle's Minimum Essential Medium, Alpha modification; Invitrogen) supplemented with 10% newborn calf serum (Invitrogen), a starving medium.
  • alpha MEM Eagle's Minimum Essential Medium, Alpha modification; Invitrogen
  • alpha MEM Eagle's Minimum Essential Medium, Alpha modification; Invitrogen
  • BAC cells prior to co-culture, prior to co-culture, BAC cells were grown in L-conditioned medium (alpha MEM with 10% newborn calf serum and 10% medium in which LADMAC cells had been cultured for two days) and LADMAC cells in starving medium for a week.
  • the proliferation and survival of BAC and LADMAC cells in the controls were not affected by the changes of medium.
  • PDMS-on-glass microchannels Fabrication of PDMS-on-glass microchannels. Designs for the channels were generated in a Computer-Aided Design (CAD) program (CIe Win, WieWeb Software). The designs for the microchannels were printed on a 10,000 dpi-resolution transparency by a commercial printer (CAD/Art Services, Inc.) from the CAD files. Negative photoresist (SU-8 50; MicroChem Corp.) was spin-coated (2100 rpm, 30s) on a silicon wafer (3-inch test-grade wafers; Silicon Sense) and baked to drive off solvent (10 min at 65 0 C, and 30 min at 95 0 C).
  • CAD Computer-Aided Design
  • SU-8 50 MicroChem Corp.
  • the photolithography was performed on the photoresist with the transparency film as the photomask (30-s exposure time) to generate features about 75 ⁇ m high.
  • the post-exposure baking step (1 min at 65 0 C and 10 min at 95 0 C)
  • the microfeatures were developed on the silicon wafer in propylene glycol methyl ether acetate (PGMEA) to remove the unexposed areas of photoresist.
  • PMEA propylene glycol methyl ether acetate
  • the surface of the SU-8 master was exposed to a vapor of (tridecafluoro-l,2,2-tetrahydrooctyl)-l- trichlorosilane (United Chemical Technologies, www.unitedchem.com) in a vacuum desiccator for about 8 h to prevent the adhesion of the PDMS to the silicon wafer.
  • the poly(dimethylsiloxane) prepolymer (PDMS; Sylgard 184 from Dow Corning) was poured onto the silanized master, degassed it for about 30 min in a vacuum desiccator, and allowed it to cure overnight in an oven at 60 0 C to minimize the quantity of un-crosslinked oligomers. After peeling the PDMS replicas off the master, holes were punched through the slabs of PDMS for the inlets and outlets of the microchannels. New glass slides (VWR) were submerged in an aqueous 0.1% (w/v) Triton X-100 solution for 5 min, then rinsed twice with distilled water, rinsed once with 100% ethanol, and dried overnight in an oven at 120 0 C.
  • VWR New glass slides
  • VWR thermally-insulated container
  • the solutions of Matrigel were prepared to form the different structures of gel inside the microchannels.
  • MG Matrigel
  • a microchannel — filled with PBS — was placed on the chilled, aluminum disc 32 and rinsed thoroughly with PBS.
  • the outlet 33 of the channel was connected to a gas- tight syringe (Hamilton) in the syringe pump (Harvard Apparatus) with polyethylene tubing 34 (PE60; Becton Dickinson) that was prefilled with PBS to maintain a uniform pressure drop throughout the tubing.
  • PE60 polyethylene tubing 34
  • the channel was rinsed once again by depositing droplets of PBS on the inlets and delivering them into the channel by the action of the syringe pump; this additional rinsing step ensured that no bubbles were trapped in the channel.
  • the warm PBS bath consisted of a Petri dish (VWR) filled with enough PBS to cover the side walls of the microchannel after it was placed in the bath. After 10 minutes in the warm bath, the spacing solutions were rinsed out with PBS, and additional drops of PBS were deposited on every inlet and outlet to prevent the hydrogels from drying out, and kept the microchannels in the incubator for further experiments.
  • VWR Petri dish
  • a concentrated suspension of cells in chilled culturing medium were prepared, and mixed with liquid Matrigel at a ratio of 80:20 in volume (MGxell solution). 6- ⁇ l droplets of either the suspension of cells in liquid Matrigel, or of the spacing solution were deposited on each inlet, and delivered into the microchannel by applying vacuum at the outlet with a syringe pump. Characterization of the portions of gel.
  • PI propidium iodide
  • PI 3,8-Diamino-5-[3-(diethylmethylammonio)propyl]-6- phenylphenanthridinium diiodide
  • Sigma propidium iodide
  • PI solution 100 ⁇ g/ml in the appropriate culturing medium
  • PI 0.68 kDa
  • CellTracker Green (5-chloromethylfluorescein diacetate; Invitrogen) was used to fluorescently label LADMAC cells.
  • CellTracker Green is a cell-permeable dye that becomes fluorescent ( ⁇ abs about 480 nm, ⁇ ems about 525 nm) upon cleavage by cytosolic esterases.
  • the stock solution was diluted to 1OmM in dimethylsulfoxide (DMSO), and stored it at -20 0 C.
  • the cells were labeled by suspending them in a solution of CellTracker in culture medium (5 ⁇ M) for 15 minutes. The cells were rinsed twice with culture medium and the samples were imaged.
  • DMSO dimethylsulfoxide
  • Vybrant DiI is a lipophilic dye that becomes highly fluorescent upon incorporation into cellular membranes ( ⁇ abs about 549 nm, ⁇ ems about 565 nm).
  • the cells were pelleted by centrifugation (5 min at 1100 rpm) and resuspended in 1 ml of culturing medium. To this solution was added 5 ⁇ l of the stock solution of Vybrant DiI. After an incubation period of 15 min at 37 0 C, the cells were pelleted again and mixed with liquid Matrigel.
  • PI propidium iodide
  • culturing medium 100 ⁇ g/ml
  • PI propidium iodide
  • Suspensions of nanometer-sized beads were also injected into the subchannels (as described herein) to determine if the structures of hydrogel extended the entire height of the channel. In these experiments, flow rates between 10 and 20 ⁇ l/min yielded hydrogel structures that extended from top and maximized the viability of the embedded cells (> 90%).
  • the channels were imaged using a charge-coupled device camera (CCD camera) on an inverted, epifluorescence microscope and the images were processed with MetaMorph and Adobe Photoshop.
  • CCD camera charge-coupled device camera
  • MetaMorph MetaMorph
  • Adobe Photoshop The fluorescence of Figs. 4 and 8 was false-colored that the contrast was increased using Photoshop.
  • Equation 1 is the general equation for diffusive transport and equations 2 - 4 are the boundary conditions for Eq. 1 according to the experiments in Fig. 9. The solution of Eqs. 1 -4 inside of the gel structure is
  • equation 5 was calculated for different (hypothetical) values of the diffusion constant. The calculated curves were compared with the experimental data (normalized intensity profiles) and the values of the diffusion coefficient of 40-kDa, 70- kDa, and 155-kDa TRITC-dextran were determined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Dispersion Chemistry (AREA)
  • Sustainable Development (AREA)
  • Clinical Laboratory Science (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physical Or Chemical Processes And Apparatus (AREA)

Abstract

L'invention porte sur des articles et des procédés pour former des structures dans des canaux microfluidiques. Les procédés selon l'invention peuvent comprendre l'utilisation de courants de fluide définis de façon spatiale à l'intérieur de microcanaux pour former des parties de gel (par exemple des structures de gel) à l'intérieur des microcanaux. Les structures de gel peuvent être formées par écoulement, par exemple de façon laminaire, d'un ou plusieurs courants de fluide dans un canal microfluidique, au moins l'un des courants comprenant un précurseur de gel. Le ou les courants de précurseur de gel peuvent être polymérisés pour former une ou plusieurs structures de gel par divers procédés par exemple par application de chaleur. Avantageusement, on peut faire varier les dimensions des structures de gel, par exemple par application de différents débits aux courants de fluide, par le choix de différentes viscosités des fluides et/ou par variation des dimensions du microcanal. A l'aide de tels procédés, différentes configurations de structures de gel dans des canaux microfluidiques peuvent être formées. Des structures de gel ayant différents composants encapsulés dans celles-ci peuvent également être formées. Les articles et procédés selon l'invention peuvent mettre en jeu, dans certains modes de réalisation, les opérations consistant à i) cultiver des cellules à l'intérieur ou sur des surfaces des parties de gel, ii) former des motifs de différents types de cellules sur ou dans des structures de gel adjacentes et/ou iii) appliquer des gradients de facteurs solubles à travers des parties de gel contenant les cellules. De telles structures peuvent être utilisées pour étudier la communication intercellulaire entre des cellules cultivées à l'intérieur de matrices tridimensionnelles d'origine biologique de dimension microscopique.
PCT/US2008/012445 2007-11-05 2008-11-04 Formation de structures de gel à l'aide de canaux microfluidiques WO2009061392A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US192307P 2007-11-05 2007-11-05
US61/001,923 2007-11-05
US881007P 2007-12-21 2007-12-21
US61/008,810 2007-12-21

Publications (1)

Publication Number Publication Date
WO2009061392A1 true WO2009061392A1 (fr) 2009-05-14

Family

ID=40243817

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/012445 WO2009061392A1 (fr) 2007-11-05 2008-11-04 Formation de structures de gel à l'aide de canaux microfluidiques

Country Status (1)

Country Link
WO (1) WO2009061392A1 (fr)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011080539A1 (fr) * 2009-12-28 2011-07-07 Achira Labs Pvt. Ltd. Élément de diagnostic et dispositif de diagnostic comprenant un élément de diagnostic
WO2011080538A1 (fr) * 2009-12-28 2011-07-07 Achira Labs Pvt. Ltd. Procédé de fabrication et d'utilisation d'un élément de diagnostic
WO2011080537A1 (fr) * 2009-12-28 2011-07-07 Achira Labs Pvt. Ltd. Composition de gel de diagnostic, procédé de fabrication d'une composition de gel de diagnostic
WO2015005863A1 (fr) * 2013-07-10 2015-01-15 Gradientech Ab Nouvelle utilisation de dispositif fluidique
EP2772530A4 (fr) * 2011-10-28 2015-07-15 Univ Korea Res & Bus Found Essai de culture cellulaire
EP3020480A1 (fr) * 2014-11-14 2016-05-18 ibidi GmbH Conduite de fluide destiné à l'examen de cellules
FR3030049A1 (fr) * 2014-12-11 2016-06-17 Andrew David Griffiths Systeme d'analyse et de culture cellulaire, procede d'analyse et de culture cellulaire associe et methode de fabrication de la puce associee
WO2017091601A1 (fr) * 2015-11-23 2017-06-01 Berkeley Lights, Inc. Structures d'isolation microfluidiques produites in situ, kits et procédés d'utilisation de celles-ci
CN110402281A (zh) * 2016-06-03 2019-11-01 牛津大学创新有限公司 凝胶网络的3d打印
US10705082B2 (en) 2015-12-08 2020-07-07 Berkeley Lights, Inc. In situ-generated microfluidic assay structures, related kits, and methods of use thereof
WO2020227648A1 (fr) * 2019-05-09 2020-11-12 EMULATE, Inc. Compositions et procédés d'utilisation de couches de gel partielles dans un dispositif microfluidique
WO2022258668A1 (fr) * 2021-06-10 2022-12-15 Mimetas B.V. Méthode et appareil pour former une structure de gel microfluidique

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000070080A1 (fr) * 1999-05-17 2000-11-23 Caliper Technologies Corp. Focalisation de microparticules dans des systemes microfluidiques
WO2002076878A2 (fr) * 2001-02-09 2002-10-03 Wisconsin Alumni Research Foundation Procede et structure permettant le guidage d'ecoulements microfluidiques
US20030162284A1 (en) * 2001-11-01 2003-08-28 Dordick Jonathan S. Biocatalytic solgel microarrays
US20040265183A1 (en) * 2003-06-26 2004-12-30 Narayanan Sundararajan Fabricating structures in micro-fluidic channels based on hydrodynamic focusing
EP1504812A1 (fr) * 2002-05-07 2005-02-09 Japan Science and Technology Agency Procede et appareil pour le prelevement d'un micromateriau
WO2005059088A1 (fr) * 2003-12-19 2005-06-30 University Of Waterloo Cellule de culture et procede et appareil destines a la culture de cellules
WO2006052223A1 (fr) * 2004-11-11 2006-05-18 Agency For Science, Technology And Research Dispositif de culture de cellules
WO2006102516A2 (fr) * 2005-03-23 2006-09-28 California Institute Of Technology Dispositifs presentant une resistance differentielle a l'ecoulement et procedes d'utilisation de ces dispositifs

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000070080A1 (fr) * 1999-05-17 2000-11-23 Caliper Technologies Corp. Focalisation de microparticules dans des systemes microfluidiques
WO2002076878A2 (fr) * 2001-02-09 2002-10-03 Wisconsin Alumni Research Foundation Procede et structure permettant le guidage d'ecoulements microfluidiques
US20030162284A1 (en) * 2001-11-01 2003-08-28 Dordick Jonathan S. Biocatalytic solgel microarrays
EP1504812A1 (fr) * 2002-05-07 2005-02-09 Japan Science and Technology Agency Procede et appareil pour le prelevement d'un micromateriau
US20040265183A1 (en) * 2003-06-26 2004-12-30 Narayanan Sundararajan Fabricating structures in micro-fluidic channels based on hydrodynamic focusing
WO2005059088A1 (fr) * 2003-12-19 2005-06-30 University Of Waterloo Cellule de culture et procede et appareil destines a la culture de cellules
WO2006052223A1 (fr) * 2004-11-11 2006-05-18 Agency For Science, Technology And Research Dispositif de culture de cellules
WO2006102516A2 (fr) * 2005-03-23 2006-09-28 California Institute Of Technology Dispositifs presentant une resistance differentielle a l'ecoulement et procedes d'utilisation de ces dispositifs

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011080539A1 (fr) * 2009-12-28 2011-07-07 Achira Labs Pvt. Ltd. Élément de diagnostic et dispositif de diagnostic comprenant un élément de diagnostic
WO2011080538A1 (fr) * 2009-12-28 2011-07-07 Achira Labs Pvt. Ltd. Procédé de fabrication et d'utilisation d'un élément de diagnostic
WO2011080537A1 (fr) * 2009-12-28 2011-07-07 Achira Labs Pvt. Ltd. Composition de gel de diagnostic, procédé de fabrication d'une composition de gel de diagnostic
US20120258309A1 (en) * 2009-12-28 2012-10-11 Achira Labs Pvt. Ltd. Diagnostic gel composition, method for making a diagnostic gel composition
US20120270337A1 (en) * 2009-12-28 2012-10-25 Dhananjaya Dendukuri Method for making and using a diagnostic element
JP2013515955A (ja) * 2009-12-28 2013-05-09 アチラ ラボズ プライベート リミテッド 診断用素子、及び診断用素子を含む診断装置
JP2013515954A (ja) * 2009-12-28 2013-05-09 アチラ ラボズ プライベート リミテッド 診断用素子の製造方法及びその使用方法
US8647742B2 (en) 2009-12-28 2014-02-11 Achira Labs Pvt. Ltd. Diagnostic gel composition, method for making a diagnostic gel composition
KR101414701B1 (ko) 2009-12-28 2014-07-03 아키라 랩스 프라이빗 리미티드 진단 소자의 제조 방법 및 사용 방법
KR101433114B1 (ko) 2009-12-28 2014-08-22 아키라 랩스 프라이빗 리미티드 진단용 겔 조성물, 진단용 겔 조성물의 제조 방법
KR101444827B1 (ko) 2009-12-28 2014-09-26 아키라 랩스 프라이빗 리미티드 진단 소자, 및 진단 소자를 포함하는 진단 장치
US9523672B2 (en) 2011-10-28 2016-12-20 Korea University Research And Business Foundation Cell culture assay
EP2772530A4 (fr) * 2011-10-28 2015-07-15 Univ Korea Res & Bus Found Essai de culture cellulaire
CN105378100A (zh) * 2013-07-10 2016-03-02 格瑞丁泰克公司 流体装置的新用途
WO2015005863A1 (fr) * 2013-07-10 2015-01-15 Gradientech Ab Nouvelle utilisation de dispositif fluidique
CN105378100B (zh) * 2013-07-10 2019-05-28 格瑞丁泰克公司 流体装置的新用途
US10487349B2 (en) 2013-07-10 2019-11-26 Gradientech Ab Use of a fluidic device
US11427851B2 (en) 2013-07-10 2022-08-30 Gradientech Ab Use of a fluidic device
EP3020480A1 (fr) * 2014-11-14 2016-05-18 ibidi GmbH Conduite de fluide destiné à l'examen de cellules
FR3030049A1 (fr) * 2014-12-11 2016-06-17 Andrew David Griffiths Systeme d'analyse et de culture cellulaire, procede d'analyse et de culture cellulaire associe et methode de fabrication de la puce associee
AU2016361413B2 (en) * 2015-11-23 2022-04-14 Berkeley Lights, Inc. In situ-generated microfluidic isolation structures, kits and methods of use thereof
WO2017091601A1 (fr) * 2015-11-23 2017-06-01 Berkeley Lights, Inc. Structures d'isolation microfluidiques produites in situ, kits et procédés d'utilisation de celles-ci
JP2019502365A (ja) * 2015-11-23 2019-01-31 バークレー ライツ,インコーポレイテッド インサイチュー生成マイクロ流体分離構造、そのキット、及びその使用方法
US11666913B2 (en) 2015-11-23 2023-06-06 Berkeley Lights, Inc In situ-generated microfluidic isolation structures, kits and methods of use thereof
US10705082B2 (en) 2015-12-08 2020-07-07 Berkeley Lights, Inc. In situ-generated microfluidic assay structures, related kits, and methods of use thereof
US11454629B2 (en) 2015-12-08 2022-09-27 Berkeley Lights, Inc. In situ-generated microfluidic assay structures, related kits, and methods of use thereof
CN110402281A (zh) * 2016-06-03 2019-11-01 牛津大学创新有限公司 凝胶网络的3d打印
GB2597030A (en) * 2019-05-09 2022-01-12 Emulate Inc Compositions and methods of using partial gel layers in a microfluidic device
WO2020227648A1 (fr) * 2019-05-09 2020-11-12 EMULATE, Inc. Compositions et procédés d'utilisation de couches de gel partielles dans un dispositif microfluidique
EP3965934A4 (fr) * 2019-05-09 2023-07-12 Emulate, Inc. Compositions et procédés d'utilisation de couches de gel partielles dans un dispositif microfluidique
GB2597030B (en) * 2019-05-09 2023-11-15 Emulate Inc Compositions and methods of using partial gel layers in a microfluidic device
WO2022258668A1 (fr) * 2021-06-10 2022-12-15 Mimetas B.V. Méthode et appareil pour former une structure de gel microfluidique
NL2028424B1 (en) * 2021-06-10 2022-12-20 Mimetas B V Method and apparatus for forming a microfluidic gel structure

Similar Documents

Publication Publication Date Title
WO2009061392A1 (fr) Formation de structures de gel à l'aide de canaux microfluidiques
Wong et al. Partitioning microfluidic channels with hydrogel to construct tunable 3-D cellular microenvironments
CN110997900B (zh) 用于快速产生用于化合物筛选的类器官/球状体的微流体平台
Vickerman et al. Design, fabrication and implementation of a novel multi-parameter control microfluidic platform for three-dimensional cell culture and real-time imaging
Gao et al. A versatile valve-enabled microfluidic cell co-culture platform and demonstration of its applications to neurobiology and cancer biology
Hung et al. Rapid microfabrication of solvent-resistant biocompatible microfluidic devices
US20190060861A1 (en) High-speed on demand microfluidic droplet generation and manipulation
US11999931B2 (en) High-throughput system and method for the temporary permeabilization of cells
KR102424970B1 (ko) 겔 네트워크의 3d 프린팅
Golden et al. Fabrication of microfluidic hydrogels using molded gelatin as a sacrificial element
EP1569510B1 (fr) Dispositif microfluidique pour la separation de cellules et utilisations de ce dispositif
US8715436B2 (en) Microfabricated biopolymer scaffolds and method of making same
US9764505B2 (en) Devices and methods for producing planar polymeric materials using microfluidics
Tanaka et al. Microcasting with agarose gel via degassed polydimethylsiloxane molds for repellency-guided cell patterning
US20200292944A1 (en) Method of making a patterned hydrogel and kit to make it
TW201736596A (zh) 在微流裝置中t淋巴球之選擇及選殖
Yamada et al. Transient microfluidic compartmentalization using actionable microfilaments for biochemical assays, cell culture and organs-on-chip
Desai et al. A photopatternable silicone for biological applications
Wu et al. Development of microfluidic alginate microbead generator tunable by pulsed airflow injection for the microencapsulation of cells
CN113893387A (zh) 一种载细胞微凝胶组装的组织工程支架及其制备方法和应用
Sahin et al. Flow lithography for structured microparticles: fundamentals, methods and applications
Ahmed et al. Microengineered 3D collagen gels with independently tunable fiber anisotropy and directionality
US20130035257A1 (en) Microfluidic arrays and methods for their preparation and use
WO2011135339A2 (fr) Réacteur
Takehara et al. Implementation of tetra-poly (ethylene glycol) hydrogel with high mechanical strength into microfluidic device technology

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08846675

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08846675

Country of ref document: EP

Kind code of ref document: A1