CN117417829A - Microfluidic organ chip system for realizing tissue dynamic interface simulation - Google Patents

Microfluidic organ chip system for realizing tissue dynamic interface simulation Download PDF

Info

Publication number
CN117417829A
CN117417829A CN202311374475.2A CN202311374475A CN117417829A CN 117417829 A CN117417829 A CN 117417829A CN 202311374475 A CN202311374475 A CN 202311374475A CN 117417829 A CN117417829 A CN 117417829A
Authority
CN
China
Prior art keywords
groove
flow channel
cell culture
channel
fluid
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202311374475.2A
Other languages
Chinese (zh)
Inventor
刘肖
苏皓然
樊瑜波
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beihang University
Original Assignee
Beihang University
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 Beihang University filed Critical Beihang University
Priority to CN202311374475.2A priority Critical patent/CN117417829A/en
Publication of CN117417829A publication Critical patent/CN117417829A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • 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
    • C12M29/00Means for introduction, extraction or recirculation of materials, e.g. pumps
    • C12M29/10Perfusion
    • 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
    • C12M35/00Means for application of stress for stimulating the growth of microorganisms or the generation of fermentation or metabolic products; Means for electroporation or cell fusion
    • C12M35/04Mechanical means, e.g. sonic waves, stretching forces, pressure or shear stimuli
    • 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
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/44Means for regulation, monitoring, measurement or control, e.g. flow regulation of volume or liquid level
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/08Regulating or influencing the flow resistance
    • B01L2400/082Active control of flow resistance, e.g. flow controllers

Landscapes

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

Abstract

The application relates to a microfluidic organ chip system for realizing tissue dynamic interface simulation, which belongs to the technical fields of biomedical engineering and microfluidics, and comprises a microfluidic chip and a perfusion device for carrying out fluid perfusion on the microfluidic chip; the micro-fluidic chip is provided with a cell culture flow channel, an adjusting flow channel and a resistance flow channel which are communicated, and the perfusion device is configured to control the fluid flow in the adjusting flow channel and the cell culture flow channel in the perfusion process so as to enable the cell culture flow channel to achieve a biomechanical environment in a target state. According to the technical scheme, the simulation of the tissue dynamic interface can be realized, and an effective platform tool is provided for researching tissue mechanics biology.

Description

Microfluidic organ chip system for realizing tissue dynamic interface simulation
Technical Field
The application belongs to the technical fields of biomedical engineering and microfluidics, and particularly relates to a microfluidic chip system for realizing tissue dynamic interface simulation.
Background
Dynamic curved interface features are common in many tissues of the human body, and are manifested as blood endothelial interfaces between tissues or between tissues and body cavities, such as in pulsatile vessels and the heart. The fluid-epithelial interface of intestinal motility, bladder and urethra movements, the fluid-air interface of the lungs during respiratory movements and the fluid-air interface of the cornea during blinking, and the dynamic interface of skeletal muscle stretching. More importantly, these tissue dynamic interfaces often also accompany multi-modal and complex biomechanical environments, mainly involving matrix stiffness, shear stress and strain. Due to its own complexity, with the development of tissue engineering, research of tissue organ systems has profound applications in biomechanics and mechanochemical biology, and the mechanism exploration of various vascular-related diseases, while also providing a basis for potential drug screening.
In recent years, by combining a microelectronic technology and a micromachining technology, the microfluidic organ chip becomes a physiological and pathological bionic platform which is widely applied and truly simulates tissues and organs in vitro, and the technology greatly overcomes the defect of poor prediction effectiveness of traditional animal model evaluation medicine curative effect, and does not relate to the problems of human body influence and ethics. Among them, polydimethylsiloxane (PDMS) has become one of the most useful materials in microfluidic chips due to its advantages of good light transmission, easy processing and molding, high biocompatibility, excellent air permeability, etc
In the related art, organ chips including a blood vessel chip, a lung chip, an intestine chip, a heart chip, and a bone chip have been developed. These organ-chips all contain this curved feature of tissue organs and are used in drug screening. However, the internal mechanical mode is either single or lacks dynamic change, and has a great gap from the complex biomechanical environment of the human body; in other words, the most essential and basic parts of the structure and function of the human body of the existing in-vitro organ chip platform have a plurality of defects on the reconstructed tissue level, in particular, the tissue or organ is a dynamic change bending interface containing complex biomechanics, and an effective physiological and pathological bionic platform is also lacking.
Therefore, how to provide a tool for effectively performing complex biomechanical study of tissue dynamic interfaces is a technical problem to be solved.
The foregoing is provided merely for the purpose of facilitating understanding of the technical solutions of the present invention and is not intended to represent an admission that the foregoing is prior art.
Disclosure of Invention
In order to overcome the problems existing in the related art at least to a certain extent, the application provides a microfluidic organ chip system for realizing tissue dynamic interface simulation, which is based on specific system constitution and configuration to realize tissue dynamic interface simulation so as to solve the technical problem that the prior art lacks effective tools for complex biomechanical research of tissue dynamic interfaces.
In order to achieve the above purpose, the present application adopts the following technical scheme:
the application provides a microfluidic organ chip system for realizing tissue dynamic interface simulation, which comprises a microfluidic chip and a perfusion device for carrying out fluid perfusion on the microfluidic chip;
the perfusion device is configured to control fluid flow in the regulating flow channel and the cell culture flow channel in a perfusion process so as to enable the cell culture flow channel to achieve a biomechanical environment in a target state.
Optionally, the microfluidic chip sequentially comprises a runner plate, a PDMS film layer and a substrate plate from top to bottom, wherein the runner plate, the PDMS film layer and the substrate plate are matched together in a bonding mode;
a first groove, a second groove and a third groove are formed on the lower surface of the runner plate, the space formed by the first groove and the PDMS film layer forms the cell culture runner, the space formed by the second groove and the PDMS film layer forms the regulating runner, and the space formed by the third groove and the PDMS film layer forms the resistance runner;
one end of the first groove is communicated with one end of the second groove and one end of the third groove, the other end of the first groove is communicated with an inlet hole penetrating through the runner plate, the other end of the second groove is communicated with an adjusting hole penetrating through the runner plate, and the other end of the third groove is communicated with an outlet hole penetrating through the runner plate;
the depth sizes of the first groove, the second groove and the third groove are all first preset values, the width sizes of the first groove and the second groove are second preset values, the width size of the third groove is a third preset value, and the first preset value is similar to the three preset values in size and is far smaller than the second preset value.
Optionally, the first preset value is 0.06mm, the second preset value is 0.6mm, and the third preset value is 0.08mm.
Optionally, the lengths of the first groove, the second groove and the third groove are respectively 10mm, 5mm and 4mm, and the aperture sizes of the inlet hole, the adjusting hole and the outlet hole are all 3mm.
Optionally, the substrate board is further provided with an opening, and a projection area of the opening on the lower surface of the runner board at least covers an area where the first groove is located.
Optionally, the perfusion device comprises a liquid reservoir, a first two-position three-way valve, a first programmable micro-syringe pump, a second two-position three-way valve, a second programmable micro-syringe pump, a three-way joint and a micro peristaltic pump;
one end of a first channel of the first two-position three-way valve is connected with the inlet hole through a pipeline, one end of a second channel of the first two-position three-way valve is connected with the liquid storage device through a pipeline, and the other end of the first channel of the first two-position three-way valve and the other end of the second channel are connected with the output end of the first programmable micro-injection pump through a Y-shaped pipeline;
one end of a first channel of the second two-position three-way valve is connected with the regulating hole through a pipeline, one end of a second channel of the second two-position three-way valve is connected with the liquid storage device through a pipeline, and the other end of the first channel of the second two-position three-way valve and the other end of the second channel are connected with the output end of the second programmable microinjection pump through a Y-shaped pipeline;
the first end of the three-way joint is connected with the outlet hole, the second end of the three-way joint is communicated with the outside atmosphere, the third end of the three-way joint is connected with the inlet end of the micro peristaltic pump through a pipeline, and the outlet end of the micro peristaltic pump is connected with the liquid reservoir through a pipeline;
the perfusion apparatus further includes a controller for controlling driving the first two-position three-way valve, the first programmable microinjection pump, the second two-position three-way valve, the second programmable microinjection pump, and the micro peristaltic pump to form a fluid perfusion cycle of the system.
Optionally, the biomechanical environment for achieving the target state in the cell culture flow channel comprises simulating the strain and the shear stress of the PDMS film layer in the cell culture flow channel based on the following expression:
wherein Q is A Indicating the flow of fluid at the inlet orifice,represents the average shear stress generated by the fluid in the cell culture flow channel, epsilon represents the strain of the PDMS membrane layer, mu represents the viscosity of the fluid, and h 0 Representing the depth of the first trench, a represents one half of the width of the first trench,
Q B the fluid flow rate at the regulating hole is shown, P is the pressure born by the PDMS membrane layer in the cell culture flow channel, R C Represents the resistance of the fluid in the resistance flow channel, R r Indicating the resistance to fluid in the cell culture flow channel.
Optionally, the PDMS membrane layer in the cell culture flow channel is used for culturing target cells, and the fluid poured into the system is a culture medium for corresponding target cells;
the target cell includes endothelial cell and epithelial cell.
Optionally, the materials of the runner plate and the substrate plate are PDMS.
The application adopts the technical scheme, possesses following beneficial effect at least:
the microfluidic organ chip system for realizing the tissue dynamic interface simulation comprises a microfluidic chip and a perfusion device for carrying out fluid perfusion on the microfluidic chip; the micro-fluidic chip is provided with a cell culture flow channel, an adjusting flow channel and a resistance flow channel which are communicated, and the perfusion device is configured to control the fluid flow in the adjusting flow channel and the cell culture flow channel in the perfusion process so as to enable the cell culture flow channel to achieve a biomechanical environment in a target state. According to the technical scheme, the micro-fluidic chip with the cell culture flow channel, the regulating flow channel and the resistance flow channel which are communicated is adopted, the flow rates of the fluid in the regulating flow channel and the cell culture flow channel are controlled through the perfusion device, and the biomechanical environment of a target state can be realized in the cell culture flow channel by using the method, so that the simulation of a tissue dynamic interface can be realized, and an effective platform tool is provided for researching tissue mechanics biology.
Additional advantages, objects, and features of the invention will be set forth in part in the description which follows and in part will become apparent to those having ordinary skill in the art upon examination of the following or may be learned from practice of the invention.
Drawings
The accompanying drawings are included to provide a further understanding of the technical aspects or prior art of the present application and constitute a part of this specification. The drawings, which are used to illustrate the technical solution of the present application, together with the embodiments of the present application, but do not limit the technical solution of the present application.
FIG. 1 is a schematic diagram of a system block diagram of a microfluidic organ-chip system according to one embodiment of the present application;
FIG. 2 is a schematic exploded view illustrating the structure of a microfluidic chip according to one embodiment of the present application;
FIG. 3 is a schematic view of a part of a flow channel plate of a microfluidic chip according to an embodiment of the present application;
FIG. 4 is a schematic illustration of a first embodiment of the present application for establishing a coordinate system for a model of a flow channel structure;
FIG. 5 is a schematic illustration II of the establishment of a coordinate system for a model of a flow channel structure in the present application;
FIG. 6 is a schematic illustration of the connection of a perfusion device to a microfluidic chip according to one embodiment of the present application;
fig. 7 is a schematic illustration of a drug release assay using a microfluidic organ-chip system in one embodiment of the present application.
In the drawing the view of the figure,
100-microfluidic chip;
110-runner plate; 111-a first trench; 112-a second trench; 113-a third trench; 114-an inlet aperture; 115-an adjustment aperture; 116-an outlet aperture;
120-PDMS film layer; 130-a substrate board; 131-opening;
200-priming device;
210-a reservoir; 220-a first two-position three-way valve; 230-a first programmable microinjection pump; 240-a second two-position three-way valve; 250-a second programmable microinjection pump; 260-tee joint; 270-a micro peristaltic pump; 280-controller.
Detailed Description
For the purpose of making the objects, technical solutions and advantages of the present application more apparent, the technical solutions of the present application will be described in detail below. It will be apparent that the described embodiments are only some, but not all, of the embodiments of the present application. All other embodiments, based on the examples herein, which are within the scope of the protection sought by those of ordinary skill in the art without undue effort, are intended to be encompassed by the present application.
As described in the background art, in the related art, organ chips including a blood vessel chip, a lung chip, an intestine chip, a heart chip, and a bone chip have been developed. These organ-chips all contain this curved feature of tissue organs and are used in drug screening. However, the internal mechanical mode is either single or lacks dynamic change, and has a great gap from the complex biomechanical environment of the human body; in other words, the most essential and basic parts of the structure and function of the human body of the existing in-vitro organ chip platform have a plurality of defects on the reconstructed tissue level, in particular, the tissue or organ is a dynamic change bending interface containing complex biomechanics, and an effective physiological and pathological bionic platform is also lacking.
In view of the above, the application provides a microfluidic organ chip system for realizing tissue dynamic interface simulation, which realizes tissue dynamic interface simulation based on specific system constitution and configuration, so as to solve the technical problem that in the prior art, there is no effective tool for complex biomechanics research of tissue dynamic interfaces.
As shown in fig. 1, in one embodiment, the microfluidic organ-chip system for implementing tissue dynamic interface simulation provided in the present application includes a microfluidic chip 100 and a perfusion device 200 for performing fluid perfusion on the microfluidic chip 100;
wherein the microfluidic chip 100 is provided with a cell culture flow channel, an adjusting flow channel and a resistance flow channel which are communicated, and the perfusion device 200 is configured to control the fluid flow rates in the adjusting flow channel and the cell culture flow channel during the perfusion process so as to enable the biomechanical environment of the target state in the cell culture flow channel.
Specifically, as shown in fig. 2, in this embodiment, the microfluidic chip 100 sequentially includes a runner plate 110, a PDMS film layer 120 and a substrate plate 130 from top to bottom, where the runner plate 110, the PDMS film layer 120 and the substrate plate 130 are matched together by a bonding manner;
as shown in fig. 2 and 3, the lower surface of the flow channel plate 110 is formed with a first groove 111, a second groove 112 and a third groove 113, the space formed by the first groove 111 and the PDMS film layer 120 forms a cell culture flow channel, the space formed by the second groove 112 and the PDMS film layer forms a regulating flow channel, the space formed by the third groove 113 and the PDMS film layer 120 forms a resistance flow channel, it should be noted that, in the application of the technical scheme of the present application, the PDMS film layer in the cell culture flow channel is used for culturing target cells, and the fluid poured in the system is the culture medium for the corresponding target cells, for example, the types of the target cells include endothelial cells, epithelial cells and the like;
as shown in fig. 2 and 3, one end of the first groove 111 communicates with one end of the second groove 112 and one end of the third groove 113, the other end of the first groove 111 communicates with an inlet hole 114 penetrating the flow channel plate, the other end of the second groove 112 communicates with an adjustment hole 115 penetrating the flow channel plate, and the other end of the third groove 113 communicates with an outlet hole 116 penetrating the flow channel plate;
the depth dimensions of the first groove 111, the second groove 112 and the third groove 113 are all a first preset value, the width dimensions of the first groove 111 and the second groove 112 are all a second preset value, the width dimension of the third groove 113 is a third preset value, the first preset value is close to the three preset values and is far smaller than the second preset value (i.e. the width of the first groove required for forming the cell culture flow channel is far greater than the depth thereof to meet the requirements of the control expression in this embodiment), for example, the first preset value is 0.06mm, the second preset value is 0.6mm (i.e. the width of the first groove is greater than or equal to 10 times the depth), the third preset value is 0.08mm, and in this embodiment, the lengths of the first groove 111, the second groove 112 and the third groove 113 can be respectively 10mm, 5mm, 4mm, and the aperture dimensions of the inlet hole 114, the regulating hole 115 and the outlet hole 116 are 3mm.
It should be noted that, each groove (cavity) structure of the microfluidic chip is manufactured by adopting a standardized micromachining method; in this embodiment, the materials of the runner plate 110 and the substrate plate 120 are PDMS, and the overall length and width of the runner plate, the PDMS film layer, and the substrate plate are equal.
In order to facilitate understanding of the technical solution of the present application, the control implementation principle of the present application is described below.
As can be easily understood by those skilled in the art, with the microfluidic chip of the above-mentioned structure, the PDMS membrane layer at the cell culture flow channel is not deformed under the condition of no pressure load, but fluid enters the resistance flow channel through the cell culture flow channel, the PDMS membrane layer is deformed under the pressure driving, and the cells cultured at the membrane layer are simultaneously subjected to the wall shear force and strain;
in the first aspect, the relation between the pressure in the flow channel and the membrane strain needs to be established;
based on an analytical model of the plate (PDMS membrane) structure in the relevant publications (VLASSAK J J, NIX W D.A new bulge test technique for the determination of Young's modulus and Poisson's ratio of thin films [ J ]. J Mater Res,1992,7 (12): 3242-9.) when it is subjected to pressure, a coordinate system (as shown in FIGS. 4 and 5, where the origin of coordinates is the groove center line) is established for the flow channel structure model in the chip in the present application, and an expression of the relation between the pressure function and membrane deflection in the cell culture flow channel [:
in expression (1), W represents the deflection of the PDMS membrane layer, P represents the pressure applied to the PDMS membrane layer, x represents the x-direction coordinate of the position point in the coordinate system shown in FIG. 4, T represents the thickness of the membrane layer, a represents half the width of the cell culture channel (i.e., half the width of the first groove), and sigma 0 Representing the film residual stress.
Further, when x=0, the maximum deflection value W of the PDMS film layer is obtained from expression (1) 0
Further, from the expressions (1) and (2), a functional expression (3) of the membrane deflection change with the maximum deflection as a parameter is obtained:
in order to obtain the relationship between the maximum deflection and the strain of the film layer, a functional expression of the film deformation arc length L and the strain epsilon can be obtained based on a basic formula of the known relationship between the material deformation and the original length:
based on the structural model shown in fig. 5, the arc length can also be expressed by the following expression (5) based on a well-known arc length integral formula:
in expression (5), W' represents the reciprocal of W;
and then based on the expression (4) (5), it is possible to obtain:
based on the relevant references (VLASSAK J J, NIX W D.A new bulge test technique for the determination of Young's modulus and Poisson's ratio of thin films [ J ]. J Mater Res,1992,7 (12): 3242-9), it is known that the function between the maximum deflection of the membrane and the pressure is expressed as the following expression (7):
in expression (7), E represents the young's modulus of the film layer, and ν represents poisson's ratio.
As shown in the above expression, it is obvious that to achieve control, it is necessary to ensure that the material mechanical properties of the PDMS film layer (thin film) are a determinable value; for example, in one embodiment, the prepolymer ratio in the preparation of the PDMS film layer is 10:1, baking and heating time is 30min, thickness is 0.02mm, and mechanical property test is carried out (pressure source adopts components manufactured by Elveflow company)Finally, the Young modulus E=0.86 MPa and the residual stress sigma of the PDMS film are determined 0 =0.24MPa。
Based on the above expression, a function of the relation between the pressure in the flow channel and the membrane strain can be obtained based on the expressions (6) and (7):
in a second aspect, to achieve computational control of fluid flow shear stress, considering the deformation of the PDMS membrane layer, the flow velocity function of the fluid along the flow channel (cell culture flow channel) can be obtained from the N-S equation:
in expression (9), v y The flow velocity in the y direction of the position point in the flow passage is represented by y, the y direction coordinate of the position point in the coordinate system shown in fig. 4 is represented by z, the z direction coordinate of the position point in the coordinate system shown in fig. 4 is represented by μ, the viscosity of the fluid is represented by h, and the flow passage height h is represented by 0 And the sum of the deflection W (shown in connection with fig. 5, h=h 0 +W);
Based on expression (9), the y-direction flow rate (Q) in the cell culture flow channel can be calculated A ) I.e. the y-direction flow rate integral is:
further, by the expression (10), the expression (9) is expressed deformably,
further, the formula is defined by the shear stress:can obtain the shear force function at the maximum deformationThe number is as follows:
further, the average shear stress function of the PDMS film layer near the surface of the runner is as follows:
in the expression (13) of the present invention,based on the expressions (6) (13), Q can be obtained A The relational expression of tau and epsilon, namely the flow function of shear stress and strain generated by cell culture flow channel fluid in a microfluidic chip:
further, in the chip structure shown in fig. 2 and 3, the pressure of the fluid in the chip passing through the cell culture channel, the channel resistance and the outlet resistance together determine the thickness of the PDMS membrane layer, and the functional relationship is as follows:
Q A R C +(Q A +Q B )R r =P (15)
in expression (15), Q B Indicating the flow rate of the outlet hole, rc indicating the resistance of the fluid in the cell culture flow channel, rr indicating the resistance of the fluid in the resistance flow channel;
and Rc, rr can be determined based on the single-channel resistance formula in the relevant literature (BRUUS H. Thermountal microfluidics [ M ]. Oxford Univ., 2007), the following expression (16):
in expression (16), l r Represents the resistance flow path length (third groove length), l c Represents the length of the cell culture channel (first groove length), a r Represents half of the width dimension of the resistance flow channel (namely half of the width of the third groove), n represents a natural number, odd represents an odd number;
further, by performing the deformation according to expression (15), the following expression can be obtained:
thus, based on the expressions (14) (17), the cell culture channel flow rate (equivalent to the inlet port flow rate) Q can be adjusted A And regulating flow rate (equivalent to outlet hole flow rate) Q B The shear stress τ and strain ε of the fluid in the cell culture channel that is desired to be set, i.e., in this embodiment of the present application, the biomechanical environment of the target state in the cell culture channel is achieved, as will be readily appreciated, involving the relevant calculations using the existing expressions (8) (16).
In the following description, as shown in fig. 1 and 6, the perfusion apparatus 200 includes a reservoir 210, a first two-position three-way valve 220 (specifically, two-position three-way valves are all electromagnetic valve valves of two-position three-way valves), a first programmable micro-injection pump 230, a second two-position three-way valve 240, a second programmable micro-injection pump 250, a three-way joint 260, and a micro peristaltic pump 270, and it should be noted that, to better control the flow, the micro-injection pumps all have positive pressure (or pushing) and negative pressure (or pulling) functions;
as shown in fig. 6, one end of the first channel of the first two-position three-way valve 220 is connected to the inlet hole 114 through a pipeline, one end of the second channel of the first two-position three-way valve 220 is connected to the liquid reservoir 210 through a pipeline, and the other end of the first channel of the first two-position three-way valve 220 and the other end of the second channel are commonly connected to the output end of the first programmable micro-injection pump 230 through a Y-shaped pipeline;
one end of the first channel of the second two-position three-way valve 240 is connected with the regulating hole 115 through a pipeline, one end of the second channel of the second two-position three-way valve 240 is connected with the liquid reservoir 210 through a pipeline, and the other end of the first channel of the second two-position three-way valve 240 and the other end of the second channel are commonly connected with the output end of the second programmable micro-injection pump 250 through a Y-shaped pipeline;
a first end of the three-way connection 260 is connected to the outlet hole 116, a second end of the three-way connection 260 is in communication with the outside atmosphere (i.e., in use, the end is exposed to the incubator air), a third end of the three-way connection 260 is connected to an inlet end of the micro peristaltic pump 270 (right side end of the micro peristaltic pump 270 in the illustration of fig. 6) via a pipeline, and an outlet end of the micro peristaltic pump 270 is connected to the reservoir 210 via a pipeline;
the infusion device 200 further comprises a controller 280, the controller 280 being configured to control driving (shown in phantom outline by the controller 280 in fig. 6, as part of the actual control lines) the first two-position three-way valve 220, the first programmable microinjection pump 230, the second two-position three-way valve 240, the second programmable microinjection pump 250, and the micro peristaltic pump 270, thereby forming a fluid infusion cycle of the system.
Specifically, for example, the controller 280 is implemented based on a PC, and the control of the components such as the syringe pump is programmed by combining the PC with the Labview platform, so as to generate fluid waveforms in different modes in the cell culture flow channel, thereby obtaining shear stress and strain with different amplitudes, different frequencies and different phase differences, and realizing a multi-mode biomechanical environment.
In the verification stage of the technical scheme, the shear stress tau and the strain epsilon to be responded and simulated are obtained through giving different flow waveforms, measurement verification is further carried out through an OCT technology and an PIV technology, and the accuracy of the technical scheme is verified through actual measurement results.
According to the technical scheme, the bending interface characteristics of different tissues with dynamic changes can be conveniently simulated, the bending interface characteristics comprise mechanical environments of various modes of shear stress and strain, the whole system is low in material consumption, circulation perfusion can be observed in real time, and a high-efficiency and powerful experimental platform is provided for researching tissue mechanics biology.
For example, in one embodiment, the microfluidic organ chip system of the present application may be used to study the relationship between blood flow mechanical signals and umbilical vein endothelial cell mechanics biology;
the specific experimental research process is as follows: calculating flow shear stress and cyclic strain in carotid bifurcation (e.g. by simulation three-dimensional modeling, calculating shear stress and strain), in order to obtain accurate geometry of arterial lumen and arterial wall required for modeling, in practice a combination of multi-weighted MRI techniques may be used, including T2 weighting (T2W) and time of flight (TOF) sequences, furthermore doppler ultrasound and blood pressure monitors may be used to determine target boundary conditions for blood flow velocity and blood pressure, finally calculating the shear stress and strain to be simulated, converting to flow Q by equation (14) (17) A ,Q B
In addition, the primary cultured human umbilical vein endothelial cells are subjected to subculture by using a special culture medium ECM, and the 2 nd to the 6 th substitutes are used for experiments; in the experiment, before the connection of the perfusion device, endothelial cells are planted at the PDMS film layer coated by the fiber with the concentration of 125ug/ml (namely, the cells are planted at the PDMS film layer of the cell culture flow channel and can be operated through an inlet hole), so that the cells are attached to the wall and the fusion degree reaches 90%;
the perfusion device is connected, fluid shear stress and strain under different conditions are loaded, and cell mechanics biology is monitored (such as by adopting an immunofluorescence luminescence method), and in practice, by adopting the technical scheme of the application, the dynamic behavior of the calcium ion response in the cultured endothelial cells is successfully monitored in real time, and biological related research can be carried out on the endothelial cell mechanics response proteins YAP and Piezo 1.
In addition, as known by those skilled in the art, as one of the most widely used materials for microfluidic chips, PDMS has a hydrophobic property, and is easy to absorb hydrophobic small molecules, thereby affecting drug screening; while the literature reports that open microfluidic chips can be conveniently added with reagents, imbibed biological liquids or objects, and removed bubbles, it is possible to overcome the above-mentioned difficulties encountered with conventional microfluidic devices.
Based on the above embodiments, in an embodiment, as shown in fig. 2, in the chip structure of the present application, the substrate board 130 is further provided with an opening 131, and a projection area of the opening 131 on the lower surface of the runner board 110 at least needs to cover an area where the first trench 111 is located, for example, in this embodiment, the first trench 111 has a structural dimension with a width of 0.6mm, a depth of 0.06mm, a length of 10mm, and a structural dimension of 2mm and a length of 10mm;
in the embodiment, by adopting the structure arrangement (the open micro-fluidic chip), related medicine release researches can be carried out by applying the micro-fluidic chip system of the application;
specifically, for example, when performing the drug release experiments, cell generation and implantation are performed as described in the previous examples, and then as shown in fig. 7, a pre-fabricated drug-loaded hydrogel sheet (the sodium alginate/drug correspondence diagram in fig. 7 is prepared mainly based on the principle that the drug added into sodium alginate is crosslinked in calcium chloride solution, and release of drug molecules is controlled mainly by controlling the concentration of sodium alginate and the concentration of calcium chloride as a crosslinking substance, so as to detect the effectiveness of the drug molecules in acting cells) is adopted, and the hydrogel sheet is adhered in the opening of the substrate plate 130, and fluorocarbon oil (FC-40) is added into the opening to reach a sufficient height to cover the hydrogel sheet, so that the state of the cells in the drug release environment can be further observed.
By adopting the technical scheme provided in the embodiment of the application, the limitation of PDMS can be overcome, the open microfluidic control can be effectively combined, the drug release function is given to the chip system, namely the preferable technical scheme can also provide an efficient and effective experimental platform for researching drug screening.
The present invention is not limited to the above-mentioned embodiments, and any changes or substitutions that can be easily understood by those skilled in the art within the scope of the present invention are intended to be included in the scope of the present invention. Therefore, the protection scope of the present invention should be subject to the protection scope of the claims.

Claims (9)

1. A microfluidic organ chip system for implementing tissue dynamic interface simulation, comprising a microfluidic chip and a perfusion device for performing fluid perfusion for the microfluidic chip;
the perfusion device is configured to control fluid flow in the regulating flow channel and the cell culture flow channel in a perfusion process so as to enable the cell culture flow channel to achieve a biomechanical environment in a target state.
2. The microfluidic organ chip system according to claim 1, wherein the microfluidic chip comprises a runner plate, a PDMS film layer and a substrate plate in sequence from top to bottom, the runner plate, the PDMS film layer and the substrate plate being mated together by bonding;
a first groove, a second groove and a third groove are formed on the lower surface of the runner plate, the space formed by the first groove and the PDMS film layer forms the cell culture runner, the space formed by the second groove and the PDMS film layer forms the regulating runner, and the space formed by the third groove and the PDMS film layer forms the resistance runner;
one end of the first groove is communicated with one end of the second groove and one end of the third groove, the other end of the first groove is communicated with an inlet hole penetrating through the runner plate, the other end of the second groove is communicated with an adjusting hole penetrating through the runner plate, and the other end of the third groove is communicated with an outlet hole penetrating through the runner plate;
the depth sizes of the first groove, the second groove and the third groove are all first preset values, the width sizes of the first groove and the second groove are second preset values, the width size of the third groove is a third preset value, and the first preset value is similar to the three preset values in size and is far smaller than the second preset value.
3. The microfluidic organ-chip system according to claim 2, wherein the first preset value is 0.06mm, the second preset value is 0.6mm, and the third preset value is 0.08mm.
4. The microfluidic organ-chip system according to claim 3, wherein,
the lengths of the first groove, the second groove and the third groove are respectively 10mm, 5mm and 4mm, and the aperture sizes of the inlet hole, the adjusting hole and the outlet hole are 3mm.
5. The microfluidic organ-chip system according to claim 2, wherein the substrate plate is further provided with an opening, and a projection area of the opening on the lower surface of the flow channel plate is at least required to cover an area where the first groove is located.
6. The microfluidic organ-chip system of claim 2, wherein the perfusion device comprises a reservoir, a first two-position three-way valve, a first programmable microinjection pump, a second two-position three-way valve, a second programmable microinjection pump, a three-way connection, and a micro peristaltic pump;
one end of a first channel of the first two-position three-way valve is connected with the inlet hole through a pipeline, one end of a second channel of the first two-position three-way valve is connected with the liquid storage device through a pipeline, and the other end of the first channel of the first two-position three-way valve and the other end of the second channel are connected with the output end of the first programmable micro-injection pump through a Y-shaped pipeline;
one end of a first channel of the second two-position three-way valve is connected with the regulating hole through a pipeline, one end of a second channel of the second two-position three-way valve is connected with the liquid storage device through a pipeline, and the other end of the first channel of the second two-position three-way valve and the other end of the second channel are connected with the output end of the second programmable microinjection pump through a Y-shaped pipeline;
the first end of the three-way joint is connected with the outlet hole, the second end of the three-way joint is communicated with the outside atmosphere, the third end of the three-way joint is connected with the inlet end of the micro peristaltic pump through a pipeline, and the outlet end of the micro peristaltic pump is connected with the liquid reservoir through a pipeline;
the perfusion apparatus further includes a controller for controlling driving the first two-position three-way valve, the first programmable microinjection pump, the second two-position three-way valve, the second programmable microinjection pump, and the micro peristaltic pump to form a fluid perfusion cycle of the system.
7. The microfluidic organ-chip system according to claim 2, wherein the biomechanical environment to achieve the target state in the cell culture flow channel comprises achieving a simulation of PDMS membrane layer strain and shear stress in the cell culture flow channel based on the following expression:
wherein Q is A Indicating the flow of fluid at the inlet orifice,represents the average shear stress generated by the fluid in the cell culture flow channel, epsilon represents the strain of the PDMS membrane layer, mu represents the viscosity of the fluid, and h 0 Representing the depth of the first trench, a represents one half of the width of the first trench,
Q B represents the fluid flow rate at the regulating hole, and P represents the cell culture flow channelThe pressure applied by the inner PDMS film layer, R C Represents the resistance of the fluid in the resistance flow channel, R r Indicating the resistance to fluid in the cell culture flow channel.
8. The microfluidic organ chip system according to claim 7, wherein the PDMS membrane layer in the cell culture flow channel is used for target cell culture, and the fluid perfused in the system is a culture medium for corresponding target cells;
the target cell includes endothelial cell and epithelial cell.
9. The microfluidic organ-chip system according to any one of claims 2 to 8, wherein the material of the flow channel plate and the substrate plate is also PDMS.
CN202311374475.2A 2023-10-23 2023-10-23 Microfluidic organ chip system for realizing tissue dynamic interface simulation Pending CN117417829A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311374475.2A CN117417829A (en) 2023-10-23 2023-10-23 Microfluidic organ chip system for realizing tissue dynamic interface simulation

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311374475.2A CN117417829A (en) 2023-10-23 2023-10-23 Microfluidic organ chip system for realizing tissue dynamic interface simulation

Publications (1)

Publication Number Publication Date
CN117417829A true CN117417829A (en) 2024-01-19

Family

ID=89529608

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311374475.2A Pending CN117417829A (en) 2023-10-23 2023-10-23 Microfluidic organ chip system for realizing tissue dynamic interface simulation

Country Status (1)

Country Link
CN (1) CN117417829A (en)

Similar Documents

Publication Publication Date Title
CN112280678B (en) Detachable and reusable hydrophobic or super-hydrophobic microfluidic organ chip
US7063942B2 (en) System and method to simulate hemodynamics
CN103805511B (en) The arteries simulation micro fluidic device directly can observed under high power objective
AU2002230396A1 (en) System and method to simulate hemodynamics
WO2021190215A1 (en) Micro-fluidic chip-level extracorporeal circulation system for vascular endothelial cell mechanics and biology research
CN103981085B (en) A kind of from establishing concentration gradient drug screening organ chip and preparation method thereof
RU171690U1 (en) Microfluidic Chip for the Creation of Cellular Models of Mammalian Organs
CN107955781A (en) The liver of metabolic process-kidney system in aids drug body based on micro-fluidic chip
US20160130543A1 (en) Modular Microtube Network for Vascularized Organ-On-A-Chip Models
US20140127795A1 (en) System and method to simulate hemodynamics
CN212688016U (en) Biomembrane dynamic culture system under simulation in vivo flow environment
Shi et al. Mock circulatory test rigs for the in vitro testing of artificial cardiovascular organs
CN117417829A (en) Microfluidic organ chip system for realizing tissue dynamic interface simulation
CN103146573B (en) Artery blood vessel simulation microfluidic device and use thereof
CN117327580A (en) Metal organ chip for biomedical research
Borenstein Organs-on-chips: how microsystems technology can transform the drug development process
Abbatiello et al. Development of an In-House Customized Perfusion-Based Bioreactor for 3D Cell Culture
EP3933028A1 (en) Devices comprising organoid chambers and uses thereof to culture, maintain, monitor or test organoids
CN107955782A (en) Liver-blood-brain barrier system of analogue body intracellular metabolite process based on micro-fluidic chip
CN113528334A (en) Microfluidic experimental plate and cell culture method
Vasant et al. Review on organ on chip
Rathod et al. Review on Organ on Chip
Batyuk et al. Biomechanical aspects of in vitro fertilization
CN110669670B (en) Human body micro-ecosystem chip and use method thereof
US20210008555A1 (en) Vascular development monitoring systems and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination