CN113201481B - Skin microsphere and preparation method and application thereof - Google Patents

Skin microsphere and preparation method and application thereof Download PDF

Info

Publication number
CN113201481B
CN113201481B CN202110419985.1A CN202110419985A CN113201481B CN 113201481 B CN113201481 B CN 113201481B CN 202110419985 A CN202110419985 A CN 202110419985A CN 113201481 B CN113201481 B CN 113201481B
Authority
CN
China
Prior art keywords
skin
cells
microsphere
dermal
microspheres
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.)
Active
Application number
CN202110419985.1A
Other languages
Chinese (zh)
Other versions
CN113201481A (en
Inventor
吴耀炯
谢俊东
杨清漾
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.)
Shenzhen International Graduate School of Tsinghua University
Original Assignee
Shenzhen International Graduate School of Tsinghua 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 Shenzhen International Graduate School of Tsinghua University filed Critical Shenzhen International Graduate School of Tsinghua University
Priority to CN202110419985.1A priority Critical patent/CN113201481B/en
Publication of CN113201481A publication Critical patent/CN113201481A/en
Priority to PCT/CN2022/086934 priority patent/WO2022222848A1/en
Application granted granted Critical
Publication of CN113201481B publication Critical patent/CN113201481B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0697Artificial constructs associating cells of different lineages, e.g. tissue equivalents
    • C12N5/0698Skin equivalents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/04Screening or testing on artificial tissues
    • C12N2503/06Screening or testing on artificial skin
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2513/003D culture
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • General Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The application discloses a skin microsphere and a preparation method and application thereof. In a first aspect of the present application, there is provided a skin microsphere comprising: a dermal cell microsphere comprising dermal cells having a first signaling pathway activity reporter system; and the epidermis layer is coated on the surface of the dermis cell microsphere, the epidermis layer comprises epidermis cells, and the epidermis cells are provided with a second signal path activity reporting system. The skin microsphere provided by the embodiment of the application has at least the following beneficial effects: the skin microsphere adopts a composite double-layer structure formed by wrapping inner core and outer layer epidermis cells formed by dermis cells, so that the skin structure of a human body is better simulated. Meanwhile, signal channel activity reporting systems are respectively carried on the inner core and the outer shell, so that the signal channel activity reporting systems can timely respond to the action effect of the to-be-detected product, and the detection of the action of the to-be-detected product on the epidermis layer and/or the dermis layer is realized.

Description

Skin microsphere and preparation method and application thereof
Technical Field
The application relates to the technical field of tissue engineering, in particular to a skin microsphere and a preparation method and application thereof.
Background
The skin is the largest organ of human body, mainly comprises epidermis, dermis, subcutaneous tissue and skin attachment, has the functions of physical barrier, sensing the outside, maintaining the body temperature and moisture, etc. Skin health is closely related to the state of cellular and matrix components in the epidermis and dermis. If one of the components is dysfunctional, it will lead to skin diseases, for example overactivated fibroblasts will lead to scar formation. At present, skin medicines and skin care products in the market can often act on different components in skin to achieve the purposes of treating diseases and improving skin conditions. The discovery of the effective components is greatly dependent on an in vitro model and a screening technology, and the in vitro model has a plurality of advantages of low cost, low ethical risk and the like compared with the human body and animal test. Therefore, a reliable and advanced skin in vitro model is of great significance for skin drug screening.
In vitro models used prior to drug screening platforms are often single cell lines, such as immortalized epidermal cells HaCaT, whose effectiveness is judged by detecting the effect of a compound on the proliferation, apoptosis, etc. cell behavior of such cell lines. However, because the cellular components are single and are far from the normal skin structure and function, the components really effective for skin health are difficult to find, and the requirements of high-flux drug screening are not met. Currently, attempts on tissue engineering skin of various cells have been used for cytotoxicity detection of cosmetics and partial replacement of animal experiments. Such tissue engineering skin products using various cells are composed of keratinocytes at the upper layer, which are keratinized to be used for testing the barrier function of the epidermis, and a simulated skin structure at the lower layer. However, it is difficult to confirm the actual effect of the drug in the epidermis and dermis to achieve the corresponding screening.
Disclosure of Invention
The present application aims to solve at least one of the technical problems existing in the prior art. Therefore, the application provides a skin microsphere capable of realizing detection of the effect of a drug in an epidermis or dermis layer, and a preparation method and application thereof.
In a first aspect of the present application, there is provided a skin microsphere comprising:
a dermal cell microsphere comprising dermal cells having a first signaling pathway activity reporter system;
and the epidermis layer is coated on the surface of the dermis cell microsphere, the epidermis layer comprises epidermis cells, and the epidermis cells are provided with a second signal path activity reporting system.
The skin microsphere provided by the embodiment of the application has at least the following beneficial effects:
the skin microsphere adopts a composite double-layer structure formed by wrapping inner core and outer layer epidermis cells formed by dermis cells, so that the skin structure of a human body is better simulated. Meanwhile, signal channel activity reporting systems are respectively carried on the inner core and the outer shell, so that the signal channel activity reporting systems can timely respond to the action effect of the to-be-detected product, and the detection of the action of the to-be-detected product on the epidermis layer and/or the dermis layer is realized.
The signal path activity reporting system refers to a system capable of detecting the activity of a specific signal path and making a qualitative or quantitative response according to the detection result, and a non-limiting manner of implementation of the signal path activity reporting system includes a reporter gene capable of detecting the specific signal path. The first signaling pathway activity reporter system and the second signaling pathway activity reporter system are used only to distinguish that the signaling pathway activity reporter system is carried by dermal cells in the dermal cell microsphere or epidermal cells in the epidermal layer, and are not used to specifically limit the kind and number of the signaling pathway activity reporter systems.
In some embodiments of the application, the first signaling pathway activity reporting system comprises at least one of a Shh signaling pathway activity reporting system, a MAPK/ERK signaling pathway activity reporting system. Shh plays an important role in regulating the development of various tissues and organs involved in cell structure, proliferation and survival, and in particular, can promote proliferation of papilla cells leading to hair follicle regeneration and hair growth, and signal pathways having similar regulatory effects thereto also include MAPK/ERK and the like. Therefore, the interaction between the drug and the cell can be detected by the signal channel reporting system carried by the dermal cell, so as to judge whether the drug can activate the cell channel in the dermal cell after penetrating the epidermis barrier, and further judge the activity of the signal channel after being activated.
In some embodiments of the application, the dermal cells are transfected with an SRE reporter gene. The MAPK/ERK signaling pathway is a major participant in cell growth and differentiation regulation, and during activation, transcription factor substrates form complexes with Serum Response Factors (SRF) and bind with Serum Response Elements (SRE) to realize gene expression regulation. Thus, selection of SRE as a reporter gene in a signaling pathway activity reporter system is effective in detecting MAPK/ERK signaling pathway activity.
In some embodiments of the application, the second signaling pathway activity reporting system comprises at least one of a canonical Wnt signaling pathway activity reporting system, an Akt signaling pathway activity reporting system. The Wnt signaling pathway is a complex regulatory network that includes three branches of canonical Wnt signaling pathway. The canonical Wnt signaling pathway is primarily involved in regulating multipotent differentiation of stem cells, development and regeneration of organs, and in particular in promoting cell proliferation and regeneration. And the Akt signal pathway has similar regulation and control effects with the classical Wnt signal pathway. Therefore, the interaction between the drug and the cell can be detected by carrying the signal channel activity reporting systems on the epidermal cells, and the effect of the interaction on cell proliferation and regeneration can be judged.
In some embodiments of the application, the epidermal cells are transfected with a TCF/LEF1 reporter gene. TCF/LEF is a HMG (High Mobility Group) transcription factor, which is used as a target gene of Wnt signaling, and participates in feedback regulation of Wnt signaling pathway, and a stable beta-catenin complex combined with the transcription factor will cause activation of canonical Wnt signaling pathway. Therefore, TCF/LEF1 is selected as a reporter gene in a signal path activity reporting system, so that the activity of a classical Wnt signal path can be detected more sensitively.
Thus, by setting the specific signaling pathway activity reporting system described above, the epidermal cell reporting system includes key pathways that promote cell proliferation and regeneration, such as canonical Wnt signaling pathway, akt pathway, and the like. The dermal cell reporting system selects signal pathways promoting hair follicle papilla formation and hair follicle regeneration, such as Shh signal pathway, MAPK/ERK signal pathway, etc., so that it can be used to observe whether or not the test article can activate the cell signal pathway and how much to activate it to find the effective components of the drugs promoting skin and hair follicle regeneration.
In some embodiments of the application, the skin microspheres have a diameter of 0.02 to 10mm. The actual size of the skin microspheres can be determined according to research requirements, but the diameter of the microspheres is preferably 0.02-10 mm, preferably within 1 mm, in consideration of the requirements of freezing and resuscitating the product.
In some embodiments of the application, the epidermal cells are skin or mucosal derived keratinocytes, which may be primary cells, or keratinocytes after culture expansion, or immortalized keratinocytes such as HaCaT. The keratinocytes in the epidermis layer may further be a monolayer or a plurality of layers according to the application requirements of the skin microsphere, for example, when detecting the effect of the drug on the proliferation of keratinocytes, the skin microsphere may employ the epidermis layer containing a monolayer of keratinocytes; when it is desired to examine the barrier function of the epidermis and the transdermal ability of the drug, keratinocytes can be further differentiated and matured to form the stratum corneum. In addition, other cells or substances besides keratinocytes can be doped into the epidermis layer according to different application requirements, for example, a certain proportion of melanocytes can be doped to test the influence of drugs on the melanocytes in the epidermis layer of the skin; the effect of drugs on skin defense can also be studied by infiltration of a proportion of immune cells (e.g., langerhans cells).
In some embodiments of the application, the dermal cells are fibroblasts isolated from the dermis of the skin, which may be primary cells, or fibroblasts after culture expansion, or immortalized fibroblasts; in addition, the fibroblast may be an embryonic stem cell or an iPS cell, which is formed by induced differentiation. The dermal cell microspheres containing fibroblasts can be used for detection and screening of drugs or cosmetics for dermal cells, in addition to functional support of the epidermal layer coated thereon. According to different application requirements of the skin microsphere, besides the fibroblast, a certain proportion of endothelial cells can be further added into the dermal cell microsphere to detect the angiopoiesis medicine, or a certain proportion of immune cells can be added into the dermal cell microsphere to carry out immune related detection or research.
In some embodiments of the application, the dermal cell microspheres may contain, in addition to dermal cells, an extracellular matrix macromolecule, such as collagen, by way of non-limiting example, and may further contain other extracellular matrix molecules, such as glycosaminoglycans, which may be hyaluronic acid. Alternatively, these extracellular matrix molecules may be replaced in part or in whole by synthetic materials.
In a second aspect of the present application, there is provided a method for preparing the skin microsphere described above, the method comprising the steps of:
mixing dermal cells with hydrogel uniformly to obtain dermal cell mixed solution;
dropwise adding the dermal cell mixture into an oil phase, and incubating to form dermal cell microspheres;
mixing dermal cell microspheres with epidermal cells, and performing 3D culture to obtain the skin microspheres.
The preparation method of the skin microsphere provided by the embodiment of the application has at least the following beneficial effects:
by adopting the method, the microspheres of the dermal cells are formed in the oil phase firstly, and then the microspheres are mixed with the epidermal cells for 3D culture, compared with the existing product, the epidermal shell formed on the outer side can form tight connection with the pore wall, so that the microspheres can be more completely covered on the surface of the dermal cell microspheres, and when the dermal microspheres are adopted for detecting and screening medicines, the medicines cannot directly enter the internal dermal cell microspheres from the pore wall, so that the barrier effect of the epidermal shell and the transdermal capability of the medicines can be reflected more. In addition, the morphology of the finally obtained skin microsphere also makes the skin microsphere easier to sample and analyze, and is more convenient to store and transport.
In some embodiments of the application, the dermal cell microspheres are resuspended in a medium and cultured for 12-36 hours prior to mixing with the epidermal cells, during which time the dermal cell microspheres are slightly reduced in size as they are spread, thereby achieving a more stable structure that facilitates subsequent adherence of the epidermal cells to their surfaces.
In a third aspect of the application, there is provided the use of a skin microsphere as described above for detecting a drug and/or cosmetic. The skin microsphere carries corresponding signal channel activity reporting systems on the inner core and the outer shell respectively, and can timely respond to the action effect of the medicine and/or the cosmetic to be detected, so that the skin microsphere can be used as a better detection model to realize the detection of the action of the medicine and/or the cosmetic on the epidermis layer and the dermis layer.
In a fourth aspect of the present application, there is provided a method for detecting an article to be detected, the method comprising the steps of:
and (3) contacting the skin microsphere with a to-be-detected product, and detecting the change of the skin microsphere before and after contact.
The skin microsphere used in the detection method carries corresponding signal channel activity reporting systems on the inner core and the outer shell respectively, and when the to-be-detected product is contacted with the skin microsphere, the skin microsphere can be used as a more accurate and visual detection model to reflect the effects of medicines and/or cosmetics on the epidermis layer and the dermis layer.
Additional aspects and advantages of the application will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practice of the application.
Drawings
FIG. 1 is a photograph of skin microspheres prepared in example 1 of the present application.
FIG. 2 shows the results of the reaction of skin microspheres to drugs in example 2 of the present application.
FIG. 3 is a schematic diagram of a plasmid carrying a signaling pathway activity reporter system and a photograph of a skin microsphere carrying a signaling pathway activity reporter system in example 3 of this application.
Detailed Description
The conception and the technical effects produced by the present application will be clearly and completely described in conjunction with the embodiments below to fully understand the objects, features and effects of the present application. It is apparent that the described embodiments are only some embodiments of the present application, but not all embodiments, and that other embodiments obtained by those skilled in the art without inventive effort are within the scope of the present application based on the embodiments of the present application.
The following detailed description of embodiments of the application is exemplary and is provided merely to illustrate the application and is not to be construed as limiting the application.
In the description of the present application, the meaning of a number is one or more, the meaning of a number is two or more, and greater than, less than, exceeding, etc. are understood to exclude the present number, and the meaning of a number is understood to include the present number. The description of the first and second is for the purpose of distinguishing between technical features only and should not be construed as indicating or implying relative importance or implicitly indicating the number of technical features indicated or implicitly indicating the precedence of the technical features indicated.
In the description of the present application, the descriptions of the terms "one embodiment," "some embodiments," "illustrative embodiments," "examples," "specific examples," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present application. In this specification, schematic representations of the above terms do not necessarily refer to the same embodiments or examples. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples.
Example 1
The embodiment provides a skin microsphere and a preparation method of the skin microsphere.
The experimental materials used in the preparation method are as follows:
rat tail type I collagen (Corning corporation, usa, product number 354236); f3100 oil phase (american 3M company); cisplatin (mikrin); DMEM (Corning corporation, usa); FBS (American BI Co.).
The preparation method comprises the following specific preparation steps:
(1) Taking rat tail type I collagen to prepare hydrogel with the concentration of 2mg/ml, uniformly mixing, and then placing the hydrogel into an ice box without regulating the pH.
(2) Human skin fibroblasts were digested, counted, and a certain amount of fibroblast suspension was taken and centrifuged.
(3) Adjusting the pH of the hydrogel to 7.0 with 0.1M NaOH, fully mixing, removing supernatant from the fibroblast suspension, mixing with the hydrogel with the pH of 7.0 to obtain a fibroblast mixed solution, evenly distributing the fibroblast mixed solution into eight connecting tubes, and placing the eight connecting tubes into an ice box.
(4) The F3100 oil phase was filter sterilized and added to 200ul of Teflon-96 well plates, respectively. Then, 4. Mu.l of the fibroblast mixture was sucked up in an eight-joint tube by a 10. Mu.l range of a row gun, dropped into the oil phase of a Teflon-96 well plate, then the Teflon-96 well plate was covered with Teflon-lad, and placed in an incubator for 15-20min to form dermal cell microspheres.
(5) After the dermal cell microspheres were formed, the well plate was removed from the incubator, 200. Mu.l of DMEM was added to each well, allowed to stand for 5min, and after the Teflon-lad was capped, the microspheres were collected by inversion, the upper liquid in Teflon-lad was poured into a 50ml beaker, the oil phase was prevented from being poured, washed 3 times with PBS, all the microspheres were resuspended in complete medium (DMEM+10% FBS), and all the liquid was transferred to a normal petri dish (non-treated) and placed into the incubator for cultivation for 24 hours.
(6) Pouring the dermal Cell microspheres and the culture medium into a Cell Spinner rotating bottle for 3D culture, digesting the immortalized epidermal cells HaCaT at the same time, taking a certain amount of epidermal Cell suspension, adding the epidermal Cell suspension into the rotating bottle which is running, fully mixing the dermal Cell microspheres and the epidermal cells, and co-culturing for 7 days to form the complete skin microspheres.
The photograph of the skin microsphere is shown in FIG. 1, A is a picture of the skin microsphere at a scale of 1000 μm, and B is a picture after bright field, different color channels (GFP and mCherry) and merger at a scale of 200 μm. As can be seen from fig. 1, the skin microsphere after 7 days of co-culture can be completely formed, has a regular shape and uniform size, has a double-layer skin structure, comprises dermal cell microspheres (mCherry marks) composed of fibroblasts and extracellular matrix (collagen), and has a composite structure formed by epidermal layers (green fluorescent protein GFP marks) wrapped by epidermal cells on the outer layer of the dermal cell microspheres, and the structure is quite similar to the skin structure of a human body, and can be well used as a detection model to simulate the skin of the human body.
Example 2
Reaction experiment of skin microsphere to drug
The skin microspheres prepared in example 1 were treated with drugs to test the proliferation and apoptosis changes of the skin microspheres before and after drug administration.
Experimental materials: bFGF (Sigma usa); mouse anti-E cadherin antibody (thermosusher, product No. 13-1700); rabbit anti-Ki 67 antibody (CST, product number 9662); goat anti-rabbit 555 antibody (CST, product number 3452); goat anti-mouse 647 antibody (CST, product number 4410).
The experimental procedure was as follows:
(1) The skin microspheres prepared in example 1 were placed in a 24-well plate without adherence for culture, 2ml of medium (containing 2% serum) was added to each well, then the drug was added, and the culture was continued in a 37-degree incubator for 48 hours.
(2) The skin microspheres are collected into a centrifuge tube by a thick mouth pipette, and after the microspheres are precipitated by a natural sedimentation method, the upper liquid is removed. After washing 2 times with PBS, 4% paraformaldehyde was added and the mixture was fixed at 4℃overnight according to the same method.
(3) Then, after washing with PBS for 2 times, the mixture was dehydrated with 10%, 20% and 30% sucrose solutions in sequence, and each step was dehydrated for 2 hours.
(4) The skin microspheres were then transferred to a tissue embedding cassette and excess water was removed by capillary siphoning with filter paper to minimize contact with the cells. Adding OCT embedding agent into the embedding box to immerse the skin microsphere, and rapidly freezing into blocks at-80deg.C. And (3) putting the frozen sample embedded blocks into a constant temperature frozen microtome of Leica for frozen section, wherein the section thickness is 8-10 microns. The temperature of the case of the frozen microtome is set to be minus 20 ℃, the temperature of the machine head is set to be minus 18 ℃, and the cut sample is put at minus 20 ℃ for standby.
(5) The frozen sections were taken out of the freezer at-20℃and left at room temperature for 15min, washed with PBS for 2 times, 10min each. After the slide was wiped dry, the samples were treated with an immunohistochemical pen for 30min with 0.25% Triton X-100 permeabilization along the edges of the samples. After absorbing Triton X-100 and washing 2 times with PBS, the mixture was blocked with PBS solution containing 3% Bovine Serum Albumin (BSA) or 10% goat serum at room temperature for 30min. After adding the primary antibody and incubating overnight at 4 ℃, the primary antibody was blotted off, washed 2 times with PBS, and the secondary antibody with fluorescent label added at the appropriate dilution and incubated for one hour at room temperature. Sucking the secondary antibody, washing for 2 times by using PBS, adding DAPI dye solution, dyeing for 20 minutes at room temperature, washing for 2 times by using PBS, sealing the tablet by using a sealing tablet, airing, and observing and photographing by using an inverted fluorescence microscope or a confocal microscope. Wherein the primary antibody is a mouse anti-E cadherin antibody and a rabbit anti-Ki 67 antibody, and the secondary antibody is a sheep anti-mouse 647 antibody and a sheep anti-rabbit 555 antibody.
As a result, as shown in fig. 2, it can be seen from the graph that the skin microspheres in the experimental group showed more cells expressing Ki67 than the control group, i.e., the number of proliferated cells was significantly increased, after 48 hours of treatment of the skin microspheres with the fibroblast growth factor bFGF.
Example 3
Referring to FIG. 3A, luciferase reporter plasmids 1 and 2 are constructed, wherein reporter plasmid 1 has thereon a TCF/LEF reporter gene comprising a TCF/LEF binding sequence and a corresponding luciferase Eluc sequence; and on reporter plasmid 2 there is an SRE reporter gene comprising an SRE binding sequence and a luciferase Fluc sequence. In addition, the reporter plasmids 1 and 2 were further provided with a green Renilla luciferase reporter gene (GrRenilla) and a Renilla luciferase reporter gene (Renilla), respectively, as internal controls. The reporter plasmid 1 and the reporter plasmid 2 are packaged by slow viruses and then are respectively infected with epidermis cells and dermis cells, and high-expression cell strains are separated through flow. Then, a skin microsphere having a dual luciferase reporter system was constructed according to the preparation method of example 1. The skin microsphere obtained by construction is shown as B in FIG. 3, the epidermis layer comprises epidermis cells with a Wnt signal channel activity reporting system, and the dermis microsphere comprises dermis cells with a MAPK/ERK signal channel activity reporting system.
After the skin microsphere is treated by the drug, the specific effect of the drug can be determined by detecting the luciferase activity. The specific detection process is as follows:
adding substrate D-luciferin for reaction 30s, measuring fluorescence spectrum in a corresponding wavelength region by using a filter disc (2 s) in the corresponding wavelength region to obtain the activity intensity of luciferase FLuc and ELuc, adding a quencher and substrate coelenterazine for reaction 7s, measuring the activity intensity of luciferase GrRenilla and Renilla by using a filter disc (1 s), and finally calculating the activation degree of a classical Wnt signal path and a MAPK/ERK signal path. The classical Wnt signal pathway is a key pathway related to promoting cell proliferation and regeneration, the MAPK/ERK signal pathway is a signal pathway related to promoting hair follicle papilla formation and hair follicle regeneration, and the activation of the two cell signal pathways by observing the drug can be used for discovering the active ingredients of the drug capable of promoting skin and hair follicle regeneration, so that the detection or screening purpose of the to-be-detected product is realized.
The present application has been described in detail with reference to the embodiments, but the present application is not limited to the embodiments, and various changes can be made within the knowledge of those skilled in the art without departing from the spirit of the present application. Furthermore, embodiments of the application and features of the embodiments may be combined with each other without conflict.

Claims (5)

1. The preparation method of the skin microsphere is characterized by comprising the following steps:
mixing dermal cells with hydrogel uniformly to obtain dermal cell mixed solution, wherein the hydrogel is prepared from type I collagen;
dropwise adding the dermal cell mixture into an F3100 oil phase, and incubating for 15-20min to form dermal cell microspheres;
mixing the dermal cell microspheres with epidermal cells, and culturing in a rotary bottle for 7 days in a 3D mode to obtain the skin microspheres;
the dermal cells are fibroblasts separated from dermis of skin, the epidermal cells are immortalized keratinocytes HaCaT derived from skin or mucosa, the dermal cell microspheres are resuspended and cultured for 12-36 hours by adopting a culture medium before being mixed with the epidermal cells, and the diameter of the dermal microspheres is 0.02-10 mm.
2. The method of claim 1, wherein the dermal cells have a first signaling pathway activity reporter system, the first signaling pathway activity reporter system being a MAPK/ERK signaling pathway activity reporter system; the epidermal cells have a second signaling pathway activity reporting system that is a canonical Wnt signaling pathway activity reporting system.
3. The method of claim 2, wherein the dermal cells are transfected with an SRE reporter gene.
4. The method of claim 2, wherein the epidermal cells are transfected with a TCF/LEF1 reporter gene.
5. Use of the skin microsphere prepared by the preparation method of any one of claims 1 to 4 for detecting bFGF, for diagnosis or treatment of a non-disease.
CN202110419985.1A 2021-04-19 2021-04-19 Skin microsphere and preparation method and application thereof Active CN113201481B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202110419985.1A CN113201481B (en) 2021-04-19 2021-04-19 Skin microsphere and preparation method and application thereof
PCT/CN2022/086934 WO2022222848A1 (en) 2021-04-19 2022-04-15 Skin microspheres, preparation method therefor, and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110419985.1A CN113201481B (en) 2021-04-19 2021-04-19 Skin microsphere and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN113201481A CN113201481A (en) 2021-08-03
CN113201481B true CN113201481B (en) 2023-10-13

Family

ID=77027617

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110419985.1A Active CN113201481B (en) 2021-04-19 2021-04-19 Skin microsphere and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN113201481B (en)
WO (1) WO2022222848A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113201481B (en) * 2021-04-19 2023-10-13 清华大学深圳国际研究生院 Skin microsphere and preparation method and application thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005059119A2 (en) * 2003-12-15 2005-06-30 The University Of Durham Differentiated cells
JP2008237045A (en) * 2007-03-26 2008-10-09 Shiseido Co Ltd Three-dimensional cultured pigmentation skin model, method for producing the same and method for evaluating localization, excretion, metabolism and degradation of melanin using three-dimenensional cultured pigmentation skin model
CN101912636A (en) * 2010-05-19 2010-12-15 深圳市冠威海事服务有限公司 Miniature skin structure, preparation method and use thereof
WO2013092480A1 (en) * 2011-12-20 2013-06-27 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. In vitro 3d-reporter skin model
WO2016138182A1 (en) * 2015-02-24 2016-09-01 Nodality, Inc. Methods and compositions for immunomodulation
CN107802891A (en) * 2017-11-09 2018-03-16 清华大学深圳研究生院 Organization engineering skin and preparation method thereof
CN108525021A (en) * 2018-04-17 2018-09-14 山西医科大学 Contain blood vessel and hair follicle structure organization engineering skin and preparation method thereof based on 3D printing
WO2018207952A1 (en) * 2017-05-12 2018-11-15 国立大学法人九州大学 Composition for hair growth and/or hair restoration
CN111269834A (en) * 2020-02-19 2020-06-12 清华大学深圳国际研究生院 3D voxel printing device and method based on cell soft sphere

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9210574D0 (en) * 1992-05-18 1992-07-01 Ca Nat Research Council Biotherapeutic cell-coated microspheres for wound/burn and prothesis implant applications
US20030022835A1 (en) * 1998-04-29 2003-01-30 Genesis Research And Development Corporation Limited Compositions isolated from skin cells and methods for their use
EP1130086A1 (en) * 2000-02-18 2001-09-05 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Genetically engineered keratinocytes and toxicity assay using said keratinocytes
US7741116B2 (en) * 2002-03-06 2010-06-22 University Of Cincinnati Surgical device for skin therapy or testing
DE102004039537A1 (en) * 2004-08-13 2006-02-23 Phenion Gmbh & Co. Kg Crosslinked collagen matrix for the preparation of a skin equivalent
US7700284B2 (en) * 2004-11-10 2010-04-20 Attagene, Inc. Reporter transcription unit populations and kits comprising same
US20130040302A1 (en) * 2011-07-11 2013-02-14 Thomas J. Burke Methods for cell reprogramming and genome engineering
US20180282690A1 (en) * 2015-10-05 2018-10-04 Max-Planck-Gesellschaft zur Förderung der Wissenchaften e.V. Method and culture medium for ex vivo culturing of epidermis-derived stem cells
EP3492583A4 (en) * 2016-07-29 2019-06-19 FUJIFILM Corporation Cell laminate, method for producing cell laminate, method for evaluating epidermal proliferation or differentiation, method for evaluating test substance, and biological implantation material
CN113201481B (en) * 2021-04-19 2023-10-13 清华大学深圳国际研究生院 Skin microsphere and preparation method and application thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005059119A2 (en) * 2003-12-15 2005-06-30 The University Of Durham Differentiated cells
JP2008237045A (en) * 2007-03-26 2008-10-09 Shiseido Co Ltd Three-dimensional cultured pigmentation skin model, method for producing the same and method for evaluating localization, excretion, metabolism and degradation of melanin using three-dimenensional cultured pigmentation skin model
CN101912636A (en) * 2010-05-19 2010-12-15 深圳市冠威海事服务有限公司 Miniature skin structure, preparation method and use thereof
WO2013092480A1 (en) * 2011-12-20 2013-06-27 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. In vitro 3d-reporter skin model
WO2016138182A1 (en) * 2015-02-24 2016-09-01 Nodality, Inc. Methods and compositions for immunomodulation
WO2018207952A1 (en) * 2017-05-12 2018-11-15 国立大学法人九州大学 Composition for hair growth and/or hair restoration
CN107802891A (en) * 2017-11-09 2018-03-16 清华大学深圳研究生院 Organization engineering skin and preparation method thereof
CN108525021A (en) * 2018-04-17 2018-09-14 山西医科大学 Contain blood vessel and hair follicle structure organization engineering skin and preparation method thereof based on 3D printing
CN111269834A (en) * 2020-02-19 2020-06-12 清华大学深圳国际研究生院 3D voxel printing device and method based on cell soft sphere

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
Advances in generation of three-dimensional skin equivalents;Subholakshmi Choudhury等;《Cytotherapy》;20210131;第23卷(第01期);第1-9页 *
CK-794致小鼠毛发生长发育异常动物模型的建立及其基因表达谱的分析;周宏元;《中国博士学位论文全文数据库(医药卫生科技辑)》;20060930(第09期);第E059-1页 *
In vitro skin three-dimensional models and their applications;Julia Klicks等;《Journal of Cellular Biotechnology》;20171113;第03卷(第01期);摘要,第21页第1段至第35页第1段 *
Julia Klicks等.In vitro skin three-dimensional models and their applications.《Journal of Cellular Biotechnology》.2017,第03卷(第01期), *
SFRP5对黑色素细胞黑素合成的抑制及其机制探究;邹道佩等;《中国细胞生物学学报》;20200515;第42卷(第05期);第741-750页 *
Tissue engineering with HaCaT cells and a fibroblast cell line;Karin Kehe等;《archives of dermatological research》;19991130;第291卷(第11期);第600-605页 *
人外泌汗腺细胞的生物学特性及其iPS细胞向汗腺分化的研究;梁含思;《万方学位论文》;20151203;第1-84页 *
人胚胎成纤维细胞作为组织工程皮肤种子细胞的可行性研究;刘柳等;《中国美容医学》;20110420;第20卷(第04期);第605-608页 *
毛囊形态发生变化过程分子调控研究进展;张燕军等;《安徽农业科学》;20100201;第38卷(第04期);第1695-1697页 *

Also Published As

Publication number Publication date
CN113201481A (en) 2021-08-03
WO2022222848A1 (en) 2022-10-27

Similar Documents

Publication Publication Date Title
Derr et al. Fully three-dimensional bioprinted skin equivalent constructs with validated morphology and barrier function
JP7018875B2 (en) Induction of human skin organoids from pluripotent stem cells
Nguyen-Ngoc et al. 3D culture assays of murine mammary branching morphogenesis and epithelial invasion
Lim et al. Investigation of the influence of keloid-derived keratinocytes on fibroblast growth and proliferation in vitro
Castro-Muñozledo Corneal epithelial stem cells, their niche and wound healing
Xie et al. Development of a three-dimensional human skin equivalent wound model for investigating novel wound healing therapies
Aizawa et al. The role of endothelial cells in the retinal stem and progenitor cell niche within a 3D engineered hydrogel matrix
US20070269792A1 (en) Cross-linked collagen matrix for producing a skin equivalent
Takezawa et al. Development of a human corneal epithelium model utilizing a collagen vitrigel membrane and the changes of its barrier function induced by exposing eye irritant chemicals
JP2004505614A (en) 3D skin model
BR112015003839B1 (en) METHOD TO PRODUCE RETINAL PIGMENTAL EPITHELIUM CELL SLAM
JP2019514370A (en) In vitro complete skin model containing three-dimensional cell culture model of sweat gland
Egles et al. Three-dimensional human tissue models of wounded skin
Meyers et al. Shape change controls supporting cell proliferation in lesioned mammalian balance epithelium
CN113201481B (en) Skin microsphere and preparation method and application thereof
KR101770983B1 (en) Manufacturing method of 3D human skin model and method for estimating Human toxicoid chemicals using the same
Ayata et al. Behaviour of endothelial cells in a tridimensional in vitro environment
Sun et al. In situ image analysis of interactions between normal human keratinocytes and fibroblasts cultured in three-dimensional fibrin gels
Murakami et al. Effect of 3D-fibroblast dermis constructed by layer-by-layer cell coating technique on tight junction formation and function in full-thickness skin equivalent
JP2017113030A (en) Reconstructed scalp model and process for screening active molecules
CN109722410B (en) 3D full-layer skin model, culture medium for forming same and preparation method
Leelahavanichkul et al. Oral and pharyngeal epithelial keratinocyte culture
TW202135779A (en) Composition for promoting hair regrowth, mitochondria rich plasma, manufacturing method thereof, and use thereof
WO2015152025A1 (en) Method for evaluating quality of sheet-shaped cell culture
Abreu et al. Gellan gum-based hydrogels support the recreation of the dermal papilla microenvironment

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
CB03 Change of inventor or designer information
CB03 Change of inventor or designer information

Inventor after: Wu Yaojiong

Inventor after: Xie Jundong

Inventor after: Yang Qingyang

Inventor before: Wu Yaojiong

Inventor before: Xie Jundong

GR01 Patent grant
GR01 Patent grant