CN114214284B - Method for culturing kidney tumor organoids - Google Patents

Method for culturing kidney tumor organoids Download PDF

Info

Publication number
CN114214284B
CN114214284B CN202111678865.XA CN202111678865A CN114214284B CN 114214284 B CN114214284 B CN 114214284B CN 202111678865 A CN202111678865 A CN 202111678865A CN 114214284 B CN114214284 B CN 114214284B
Authority
CN
China
Prior art keywords
kidney
culture
extracellular matrix
kidney tumor
tumor organoid
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
CN202111678865.XA
Other languages
Chinese (zh)
Other versions
CN114214284A (en
Inventor
包骥
步宏
王坤杰
朱星龙
金熙
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.)
West China Precision Medicine Industrial Technology Institute
West China Hospital of Sichuan University
Original Assignee
West China Precision Medicine Industrial Technology Institute
West China Hospital of Sichuan 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 West China Precision Medicine Industrial Technology Institute, West China Hospital of Sichuan University filed Critical West China Precision Medicine Industrial Technology Institute
Priority to CN202111678865.XA priority Critical patent/CN114214284B/en
Publication of CN114214284A publication Critical patent/CN114214284A/en
Application granted granted Critical
Publication of CN114214284B publication Critical patent/CN114214284B/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
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • 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
    • C12M23/08Flask, bottle or test tube
    • 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
    • C12M23/10Petri dish
    • 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
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue
    • 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)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Sustainable Development (AREA)
  • Clinical Laboratory Science (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Physics & Mathematics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Oncology (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention provides a method for culturing kidney tumor organoids, belonging to the organoid culture field. The invention provides a kidney tumor organoid culture container, which is a cell culture container with a kidney extracellular matrix imprinting block array on the inner bottom surface; the area of the bottom surface not covered by the extracellular matrix of the kidney is blocked by a blocking agent; the kidney extracellular matrix imprinting block array consists of circular lattice points with the diameter of 50-200 mu m, and the distance between each two circular lattice points is 25-300 mu m. The invention also provides a culture method of the kidney tumor organoids, which shortens the culture time, simplifies the culture mode, effectively controls the particle size and arrangement of the kidney tumor organoids, and improves the repeatability and stability of drug screening. The cultured kidney tumor organoids are used for drug screening, so that the method has higher efficiency and safety, and can be used for personalized treatment and guiding clinical medication.

Description

Method for culturing kidney tumor organoids
Technical Field
The invention belongs to the field of organoid culture, and particularly relates to a method for culturing kidney tumor organoids.
Background
Urinary system tumors are one of the most common malignant tumors in the world, including prostate cancer, bladder cancer and renal cell carcinoma. Renal cell carcinoma (renal cell carcinoma, RCC) is abbreviated as renal carcinoma, and has the third most serious incidence among urinary system tumors. Renal cancer is a malignant tumor that originates from the urinary tract epithelial system of the kidney parenchyma, severely affects the renal function of the patient, and can metastasize to the outside of the kidney with the progression of the condition. At present, the pathogenesis and progress of kidney cancer is not completely clear, and is mostly related to genetics, environment, related chronic medical history and bad life habit. About 55% of patients with kidney cancer have no typical early manifestation, and more symptomatic patients are found in lumbago and hematuria than in healthy physical examination, and fewer patients are diagnosed due to the fact that the abdomen can touch the tumor. At present, the treatment measures about the kidney cancer mainly comprise comprehensive treatment including operation, radiotherapy and chemotherapy and targeted treatment. With the development of accurate medicine, tumor treatment has entered the individualization age: firstly, using high-flux gene sequencing to find out tumor related mutation and searching a drug target; and secondly, the sensitivity of the targeted drug is tested by using a tumor model, so that personalized treatment is realized. However, when kidney cancer is diagnosed, it is often too late, and it is important how to efficiently and briefly build an in vitro model capable of reflecting the heterogeneity of patients. To solve the above problems, researchers have focused on organoid technology.
Organoid technology is the most rapidly developing in vitro culture technology in the last decade, and has been honored as one of the most important scientific advances in 2013. The technology can induce and differentiate in vitro into functional cell clusters with the basic characteristics of organs derived from the cells, the embryonic stem cells and the adult stem cells by virtue of the tissues, the embryonic stem cells and the adult stem cells, and provides a new in vitro model for simulating living organs. In modern biomedical research, the most commonly used cell culture and animal models only partially simulate in-vivo environments, and cannot fully show the real situation of a research target in a human body. Researchers have therefore given great attention to 3D culture techniques, which they wish to better mimic the microenvironment of in vivo cell growth, especially tumor cells, based on the material structure basis that they retain the cell microenvironment. The human tumor organoids (PDO) retain individual tumor heterogeneity, and the screening of targeted drugs by PDO guides clinical drugs, thus showing great value in personalized treatment. The stability and fidelity of tumor organoids as drug screening models can be divided into three aspects. First, the tumor organoids retain the heterogeneity of the primary tumor during long-term culture, wherein the results of different generations of tumor organoids on anti-tumor drug screening are substantially consistent. Second, the results of drug screening of the tumor organoids are consistent with clinical medication of patients, which indicates that the tumor organoids have great potential value for accurate treatment. Thirdly, the tumor organoid can be regarded as a 'cell line' in the long-term culture process, and the same tumor organoid cell line is subjected to drug screening, so that the same tumor organoid has consistent drug screening results.
The tumor organoids can be passaged for a long time without losing or changing genetic information during subculture. The heterogeneity between intratumoral and external is preserved. At present, colorectal cancer, non-small cell lung cancer, breast cancer and pancreatic cancer tumor organoid models have been widely applied to drug evaluation, biomarker identification, biological research and personalized treatment. Patent CN113186165A, CN111607495A discloses a method for culturing kidney tumor organoids, which is mainly obtained by mixing kidney cancer cells or kidney cancer tissues with matrigel, or wrapping the cells or tissues with matrigel and then culturing the cells or tissues in a specific culture medium. However, the existing method for culturing kidney tumor organoids has the defects of complicated operation, higher cost, small scale, different organoids with different particle sizes, different planes, difficult analysis and observation, and the like, and cannot be used for large-scale mass production. Meanwhile, the existing method for culturing kidney tumor organoids has long culture period and may cause delay of patient's illness.
Therefore, the method for quickly searching the kidney tumor organoids has the advantages of being quick, simple and convenient to operate and low in cost, being uniform in culture size, growing on the same plane and being convenient to analyze and observe, and achieving the quick large-scale culture of the kidney tumor organoids has important significance.
Disclosure of Invention
The invention aims to provide a method for culturing kidney tumor organoids.
The invention provides a kidney tumor organoid culture container, which is a cell culture container with a kidney extracellular matrix imprinting block array on the inner bottom surface;
the area of the bottom surface not covered by the extracellular matrix of the kidney is blocked by a blocking agent;
the kidney extracellular matrix imprinting block array consists of circular lattice points with the diameter of 50-200 mu m, and the distance between each two circular lattice points is 25-300 mu m.
Further, the kidney extracellular matrix blot block array is composed of circular lattice points with a diameter of 100 μm, and the interval between each circular lattice point is 50 μm.
Further, the area of the kidney extracellular matrix blotting block array is 1-10 cm 2
Preferably, the area of the kidney extracellular matrix blotting block array is 4cm 2
Further, the container is a culture dish, a culture box, a culture plate or a culture bottle;
and/or the blocking agent is Pluronic F-127;
preferably, the mass fraction of Pluronic F-127 is 1-5%.
Further, the kidney extracellular matrix is porcine kidney extracellular matrix;
preferably, the preparation method of the pig kidney extracellular matrix comprises the following steps:
(1) Preparing a pig whole kidney decellularized scaffold by adopting a Triton-SLES-Triton perfusion method;
(2) The obtained decellularized kidney stent is obtained by freeze-drying and crushing the decellularized kidney stent and then carrying out enzymolysis by pepsin.
Preferably, the Triton-SLES-Triton infusion method is to take fresh pig kidney, cannulate renal artery, freeze and thaw the kidney after removing blood, infuse with 1% Triton X-100 for 3 hours, then 1% sodium laureth sulfate (SLES) for 6 hours, infuse with 1% Triton X-100 for 3 hours, and finally infuse with PBS for 2 hours to balance the decellularized kidney, thus obtaining the decellularized kidney scaffold. The flow rate throughout the course was 15mL/min during the perfusion.
Preferably, during enzymolysis, the concentration of the extracellular matrix of the kidney obtained after crushing is 10mg/mL; the mass ratio of pepsin to extracellular matrix powder is 1:10. In addition, the pH was 2 during the enzymolysis, the temperature was room temperature, and the enzymolysis time was 72 hours.
Further, the concentration of the kidney extracellular matrix is 0.05-0.5 mg/mL;
preferably, the concentration of the kidney extracellular matrix is 0.1-0.5 mg/mL.
The kidney tumor organoid culture container is prepared by adopting a PDMS micro-pattern printing technology: firstly, a silicon wafer with a specific pattern is etched by using laser as a template, a micro-array PDMS stamp with a convex circular micro-pattern with a specific size is obtained by reverse molding, kidney extracellular matrix glue is coated on the surface of the PDMS stamp, the kidney extracellular matrix glue is stamped on the bottom surface of a culture container, an extracellular matrix micro-pattern array is obtained, and then the rest of the culture container is sealed by using a sealing agent, so that the kidney tumor organoid culture container is obtained.
The invention also provides a kidney tumor organoid culture method, which comprises the following steps:
inoculating the kidney cancer cells into the culture container, culturing for 4-6 h, washing off unattached cells, and culturing for 3-5 days to obtain the kidney tumor organoid.
Further, the inoculation density of the kidney cancer cells is 10 4 ~10 6 A personal/culture container;
and/or, the kidney cancer cells are derived from a human;
preferably, the inoculation density of the kidney cancer cells is 10 5 A culture vessel.
The invention also provides a kidney tumor organoid which is prepared by the method.
The invention also provides application of the kidney tumor organoid in constructing a kidney cancer model or screening medicines.
The invention provides a method for culturing kidney tumor organoids, which cultures kidney cancer cells by adopting kidney ECM pattern microarrays with specific lattice shapes, sizes and densities, prepares the kidney tumor organoids, shortens the in-vitro amplification culture time of the kidney tumor organoids, simplifies the culture mode of the kidney tumor organoids, and effectively overcomes the defects of complicated operation, higher cost, small scale, different organoid particle sizes, difficult analysis and observation and the like of the traditional tumor organoids. Effectively controls the particle size and arrangement of kidney tumor organoids and improves the repeatability and stability of drug screening. The kidney decellularized ECM provides a tumor microenvironment for kidney tumor organoid culture, is favorable for maintaining tumor heterogeneity, and simulates in vivo kidney cancer tissues. The kidney tumor organoid is used for drug screening, has higher efficiency and safety, and simultaneously PDO can be used for personalized treatment to guide clinical medication.
It should be apparent that, in light of the foregoing, various modifications, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined by the appended claims.
The above-described aspects of the present invention will be described in further detail below with reference to specific embodiments in the form of examples. It should not be understood that the scope of the above subject matter of the present invention is limited to the following examples only. All techniques implemented based on the above description of the invention are within the scope of the invention.
Drawings
FIG. 1 shows the course of culture of kidney tumor organoids according to the invention.
Fig. 2 shows the results after decellularization of whole pig kidneys: a is the general appearance of the kidneys before and after decellularization; b is an electron microscope image before and after decellularization; c is DNA content detection before and after decellularization; d is H & E staining before and after decellularization; e is the dyeing of type I collagen fibers before and after decellularization; f is the staining of type IV collagen fibers before and after decellularization; g is fibronectin staining before and after decellularization; h is laminin staining before and after decellularization; scale = 20 μm; in the figure Native is non-decellularized and DLSs is decellularized.
Fig. 3 is a kidney decellularized ECM powder (left) kidney decellularized ECM gel (right).
FIG. 4 shows the results of the dead and alive staining of kidney tumor organoids after the action of each drug.
Fig. 5 shows a decellularized renal extracellular matrix micropattern array chip prepared by micropattern printing technology and a kidney tumor organoid cultured: a is PDMS seal; b is an extracellular matrix micropattern array; scale = 100 μm.
FIG. 6 shows the kidney tumor organoids formed after 3 days of culture.
Detailed Description
Unless otherwise indicated, the materials and equipment used in the embodiments of the present invention are all known products and are obtained by purchasing commercially available products.
The culture route of the kidney tumor organoid of the invention is shown in figure 1, and comprises the following steps:
(1) Preparation of kidney decellularized ECM: porcine whole kidney decellularized scaffold was prepared using Triton-SLES-Triton perfusion. The obtained decellularized kidney scaffold (kidney extracellular matrix) was further lyophilized, and after pulverization using a ball mill, the decellularized kidney extracellular matrix (ECM) gel was obtained by pepsin enzymolysis.
(2) Preparing a pattern micro-array master plate by adopting a PDMS micro-pattern printing technology: a silicon wafer with a specific pattern is etched by using laser as a template, the etching precision reaches 1 mu m, the etching area reaches 12 inches at maximum, and a Polydimethylsiloxane (PDMS) seal is obtained by reverse molding. The extracellular matrix gel (called extracellular matrix gel or matrigel) is coated on the surface of a seal, then a PDMS micro-pattern printing technology is utilized to lay an extracellular matrix gel pattern micro-array on the bottom of a cell culture dish which is subjected to non-attachment treatment, pluronic F-127 seals the space at the bottom of the rest culture dish, so that the added cells can only grow on the extracellular matrix gel pattern in an attached mode. The kidney decellularized ECM pattern microarray chip was obtained.
(3) Culture of kidney tumor organoids: the kidney decellularized ECM pattern microarray chip is used for culturing kidney tumor organoids, so that the in-vitro amplification culture time is shortened, and the particle size and arrangement of the kidney tumor organoids can be effectively controlled.
EXAMPLE 1 method of culturing kidney tumor organoids of the invention
The method for culturing the kidney tumor organoid comprises the following steps:
(1) Preparation of pig kidney decellularized extracellular matrix glue: after taking fresh pig kidneys, renal arteries were cannulated and flushed with PBS for 10 minutes to remove blood. Kidneys were then frozen and thawed, perfused with 1% triton X-100 for 3 hours, followed by 1% sodium laureth sulfate (SLES) for 6 hours, further perfused with 1% triton X-100 for 3 hours, and finally perfused with PBS for 2 hours to equilibrate the decellularized kidneys to give a decellularized renal stent (renal extracellular matrix). The flow rate throughout the course was 15mL/min during the perfusion. Decellularized kidney scaffolds (DLSs) were then cut into cubes of 1X 1cm and lyophilized. Further crushing the mixture by a ball mill to obtain kidney extracellular matrix powder, and carrying out enzymolysis on the kidney extracellular matrix powder by using pepsin to obtain the decellularized kidney extracellular matrix gel. During enzymolysis, extracellular matrix powder is dissolved in water, the concentration is 10mg/mL, the mass ratio of pepsin to extracellular matrix powder is 1:10, the pH is 2 during enzymolysis, the temperature is room temperature, and the enzymolysis time is 72 hours.
The pig kidney is white and transparent after decellularization, and after immunofluorescence, histochemical and enzyme-linked immunosorbent assay, extracellular matrix protein is well reserved (figure 2), and further, decellularized kidney extracellular matrix gel is obtained through crushing and enzymolysis (figure 3).
(2) A silicon wafer with a specific pattern is etched by using laser as a template, and a PDMS stamp is obtained by reverse molding, so that a pattern microarray with convex circular array points (each circular array point has a diameter of 100 μm, the distance between each circular array point is 50 μm, and the microarray size is 4 cm) 2 ) And a PDMS stamp.
(3) The matrigel concentration is regulated by PBS, 1mL of pig kidney decellularized extracellular matrigel with the concentration of 0.1mg/mL is coated on the surface of a PDMS seal, after incubation for 20 minutes at room temperature, the superfluous matrigel is washed off, and the mixture is dried in an incubator at 37 ℃.
(4) The PDMS stamp was pressed against the inner bottom surface of the cell non-attached culture dish, and a force of 0.2 newton was applied for 10 minutes, and the stamp was removed after the pattern was formed on the bottom surface of the culture dish.
(5) The mass fraction of 1% Pluronic F-127 (Prulinix F-127, a polypropylene glycol and ethylene oxide addition polymer (polyether)) was added and incubated at room temperature for 2 hours, and the space on the inner bottom surface of the remaining dish was closed to obtain an "extracellular matrix pattern microarray" having an adhesive effect on cells. Before the prepared microarray culture dish is used, PBS is used for cleaning for 3 times, so that the culture dish substrate cannot be completely dried, and then cells are inoculated.
(6) Human kidney cancer cells (os-rc-2) were inoculated in 1.5X10-dose in a petri dish 5 Dish) is cultured for 6 hours at 37 ℃, unattached cells are washed off, and the kidney tumor organoids which are visible with naked eyes can be formed after 3 days of culture. The kidney tumor organoids are all on the same plane, and have uniform size and controllable quantity.
Human kidney cancer cells are defined to attach to "extracellular matrix patterned microarrays" for ingrowth. The medium used in this step was 1640 medium containing 10% fetal bovine serum and 1% diabody.
The decellularized renal extracellular matrix pattern microarray chip is prepared by a micropattern printing technology, so that renal tumor cells are cultured, and the renal tumor organoid can be formed after 3 days of culture.
The beneficial effects of the present invention are demonstrated by specific test examples below.
Test example 1 screening of the kidney tumor organoids of the present invention for drugs
Experimental method
The kidney tumor organoids of 100 μm diameter cultured in example 1 were selected and screened with different concentrations (5. Mu.g/ml, 10. Mu.g/ml, 50. Mu.g/ml, 100. Mu.g/ml) of sunitinib, sirolimus and everolimus, respectively, and incubated for 24h and 48h. Dead and alive staining analysis was performed using calcein and propidium iodide fluorescent double staining and imaging analysis using confocal microscopy.
Experimental results
From the results of fig. 4, it can be seen that: the kidney tumor organoids cultured by the invention can effectively screen the influence of medicine types, medicine doses and medicine action time on kidney tumors. The results show that the activity of the kidney tumor organoids is related to the drug species, the drug dosage and the culture time. Sunitinib has the greatest inhibitory activity on kidney tumor organoids, and as sunitinib concentration and culture time increase, kidney tumor organoids stop growing. Sirolimus and everolimus have some inhibitory activity on kidney tumor organoids, but have weaker inhibitory activity than sunitinib. It was found that the organoids can be better analyzed and the results of drug screening observed after size control and alignment. The kidney tumor organoid pattern microarray chip is applied to personalized treatment and potential value of antineoplastic drug screening.
Test example 2 screening of the method for culturing kidney tumor organoids of the present invention
Early-stage researches show that if rectangular lattice points are used in the pattern microarray, the effect of preparing kidney tumor organoids is poor, and even kidney tumor organoids cannot be generated; while kidney tumor organoids prepared using circular lattice points are significantly better than rectangular lattice points. Thus, an extracellular matrix patterned microarray was prepared by the method described in example 1, and the size of the circular array spots (the diameters of the circular array spots were 50 μm, 100 μm, 150 μm, 200 μm, respectively) in the patterned microarray was changed, the interval between each circular array spot was 50 μm, and the microarray size was 4cm 2 . Each pattern microarray is shown in fig. 5.
According to realityExample 1A kidney tumor organoid was cultured separately using the above extracellular matrix patterned microarray, which was washed 1 time with PBS before use to ensure that the culture substrate was not completely dried, inoculated with human kidney cancer cells (os-rc-2, inoculum size 1.5X10 5 /dish). The patterned microarray was grown on spots defined in the extracellular matrix gel patterned microarray by restricting the growth and adhesion space of cells on the bottom of the cell culture dish, and kidney tumor organoids were formed by culturing for 3 days (fig. 6).
The optimal size of circular array spots in the microarray was selected based on the morphology of the kidney tumor organoids, the dead-alive function. The research finds that: when the diameter of the circular lattice point is too small (50 μm), kidney tumor organoids cannot be cultivated; with the increase of the diameter of the circular lattice points, dead cells in the kidney tumor organoids are increased, and the functional activity is poor, which is unfavorable for evaluation as a model. Considering comprehensively, the circular array point with the diameter of 100 μm is finally selected as the optimal array point for culturing the kidney tumor organoid pattern microarray, and the prepared kidney tumor organoid has optimal morphology and function.
In summary, the invention provides a method for culturing kidney tumor organoids, which cultures kidney cancer cells by adopting kidney ECM pattern microarrays with specific lattice shapes, sizes and densities, prepares the kidney tumor organoids, shortens the in-vitro amplification culture time of the kidney tumor organoids, simplifies the culture mode of the kidney tumor organoids, and effectively overcomes the defects of complicated operation, higher cost, small scale, different particle sizes of organoids, difficult analysis and observation and the like of the conventional tumor organoids. Effectively controls the particle size and arrangement of kidney tumor organoids and improves the repeatability and stability of drug screening. The kidney decellularized ECM provides a tumor microenvironment for kidney tumor organoid culture, is favorable for maintaining tumor heterogeneity, and simulates in vivo kidney cancer tissues. The kidney tumor organoid is used for drug screening, has higher efficiency and safety, and simultaneously PDO can be used for personalized treatment to guide clinical medication.

Claims (13)

1. A kidney tumor organoid culture container, characterized in that: the cell culture container is provided with a kidney extracellular matrix imprinting block array on the inner bottom surface;
the area of the bottom surface not covered by the extracellular matrix of the kidney is blocked by a blocking agent;
the kidney extracellular matrix imprinting block array consists of circular lattice points with the diameter of 100 mu m, and the interval between each circular lattice point is 50 mu m;
the concentration of the kidney extracellular matrix is 0.05-0.5 mg/mL.
2. A kidney tumor organoid culture vessel according to claim 1, wherein: the area of the kidney extracellular matrix imprinting block array is 1-10 cm 2
3. A kidney tumor organoid culture vessel according to claim 2, wherein: the area of the kidney extracellular matrix imprinting block array is 4cm 2
4. A kidney tumor organoid culture vessel according to any one of claims 1 to 3, wherein: the container is a culture dish, a culture box, a culture plate or a culture bottle;
and/or the blocking agent is Pluronic F-127.
5. A kidney tumor organoid culture vessel according to claim 4, wherein: the mass fraction of Pluronic F-127 is 1-5%.
6. A kidney tumor organoid culture vessel according to any one of claims 1 to 3, wherein: the kidney extracellular matrix is porcine kidney extracellular matrix.
7. A kidney tumor organoid culture vessel according to claim 6, wherein: the preparation method of the pig kidney extracellular matrix comprises the following steps:
(1) Preparing a pig whole kidney decellularized scaffold by adopting a Triton-SLES-Triton perfusion method;
(2) The obtained decellularized kidney stent is obtained by freeze-drying and crushing the decellularized kidney stent and then carrying out enzymolysis by pepsin.
8. A kidney tumor organoid culture vessel according to claim 1, wherein: the concentration of the kidney extracellular matrix is 0.1-0.5 mg/mL.
9. A kidney tumor organoid culture method, which is characterized in that: the method comprises the following steps:
inoculating the kidney cancer cells into the culture container according to any one of claims 1-8, culturing for 4-6 h, washing off unattached cells, and culturing for 3-5 days to obtain the kidney tumor organoid.
10. The culture method according to claim 9, wherein: the inoculation density of the kidney cancer cells is 10 4 ~10 6 A personal/culture container;
and/or, the kidney cancer cells are of human origin.
11. The culture method according to claim 10, wherein: the inoculation density of the kidney cancer cells is 10 5 A culture vessel.
12. A kidney tumor organoid, characterized in that: a kidney tumor organoid prepared by the method of any one of claims 9 to 11.
13. Use of the kidney tumor organoid of claim 12 in constructing a kidney cancer model or in drug screening.
CN202111678865.XA 2021-12-31 2021-12-31 Method for culturing kidney tumor organoids Active CN114214284B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111678865.XA CN114214284B (en) 2021-12-31 2021-12-31 Method for culturing kidney tumor organoids

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111678865.XA CN114214284B (en) 2021-12-31 2021-12-31 Method for culturing kidney tumor organoids

Publications (2)

Publication Number Publication Date
CN114214284A CN114214284A (en) 2022-03-22
CN114214284B true CN114214284B (en) 2024-01-26

Family

ID=80707553

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111678865.XA Active CN114214284B (en) 2021-12-31 2021-12-31 Method for culturing kidney tumor organoids

Country Status (1)

Country Link
CN (1) CN114214284B (en)

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104208751A (en) * 2014-08-19 2014-12-17 温州医科大学 Preparation method for novel kidney acellularized biological scaffold
CN104841017A (en) * 2015-05-28 2015-08-19 四川大学华西医院 Cellularization biological liver stent with anticoagulation property and preparation method of cellularization biological liver stent
CN108102913A (en) * 2016-11-25 2018-06-01 中国科学院苏州纳米技术与纳米仿生研究所 Three-dimensional cell culture chip, its preparation method and application based on soft lithography
CN109182125A (en) * 2018-09-25 2019-01-11 中国科学院苏州纳米技术与纳米仿生研究所 Three-dimensional single cell source cell ball production chip, preparation method and application
CN110249044A (en) * 2016-11-18 2019-09-17 洛桑联邦理工学院 Organoid organizational project
CN111494718A (en) * 2020-04-24 2020-08-07 四川大学华西医院 Preparation method of animal decellularization lung biological scaffold material
CN111826338A (en) * 2019-12-27 2020-10-27 四川大学华西医院 Method for rapidly culturing liver bud organoid
CN112210536A (en) * 2020-09-03 2021-01-12 电子科技大学 2D and 3D cell co-culture system capable of being continuously harvested without enzyme digestion and construction method and application thereof
CN113151177A (en) * 2021-05-21 2021-07-23 四川大学华西医院 Mammary gland or breast cancer tissue acellular matrix and preparation method and application thereof
CN113186165A (en) * 2021-04-15 2021-07-30 北京大学 Kidney cancer related organoid combination and application thereof
CN114214282A (en) * 2021-12-31 2022-03-22 四川大学华西医院 Method for culturing lung tumor organoid
CN114214283A (en) * 2021-12-31 2022-03-22 四川大学华西医院 Method for culturing liver tumor organoid

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2210589B1 (en) * 2009-01-22 2015-05-06 Ludwig-Maximilians-Universität München Vesicular phospholipid gels comprising proteinaceous substances
WO2014169031A1 (en) * 2013-04-10 2014-10-16 Tufts University Two and three dimensional decellularized ecm constructs and uses therefor
WO2017024206A1 (en) * 2015-08-05 2017-02-09 Colorado State University Research Foundation Engineered platforms to stabilize both hepatocytes and endothelial cells in vitro

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104208751A (en) * 2014-08-19 2014-12-17 温州医科大学 Preparation method for novel kidney acellularized biological scaffold
CN104841017A (en) * 2015-05-28 2015-08-19 四川大学华西医院 Cellularization biological liver stent with anticoagulation property and preparation method of cellularization biological liver stent
CN110249044A (en) * 2016-11-18 2019-09-17 洛桑联邦理工学院 Organoid organizational project
CN108102913A (en) * 2016-11-25 2018-06-01 中国科学院苏州纳米技术与纳米仿生研究所 Three-dimensional cell culture chip, its preparation method and application based on soft lithography
CN109182125A (en) * 2018-09-25 2019-01-11 中国科学院苏州纳米技术与纳米仿生研究所 Three-dimensional single cell source cell ball production chip, preparation method and application
CN111826338A (en) * 2019-12-27 2020-10-27 四川大学华西医院 Method for rapidly culturing liver bud organoid
CN111494718A (en) * 2020-04-24 2020-08-07 四川大学华西医院 Preparation method of animal decellularization lung biological scaffold material
CN112210536A (en) * 2020-09-03 2021-01-12 电子科技大学 2D and 3D cell co-culture system capable of being continuously harvested without enzyme digestion and construction method and application thereof
CN113186165A (en) * 2021-04-15 2021-07-30 北京大学 Kidney cancer related organoid combination and application thereof
CN113151177A (en) * 2021-05-21 2021-07-23 四川大学华西医院 Mammary gland or breast cancer tissue acellular matrix and preparation method and application thereof
CN114214282A (en) * 2021-12-31 2022-03-22 四川大学华西医院 Method for culturing lung tumor organoid
CN114214283A (en) * 2021-12-31 2022-03-22 四川大学华西医院 Method for culturing liver tumor organoid

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Adam D. McInnes等.Preparation and Use of Decellularized Extracellular Matrix for Tissue Engineering.Journal of Functional Biomaterials.2022,第13卷(第4期),1-54. *
Yi Li等.Decellularization of porcine whole lung to obtaina clinical-scale bioengineered scaffold.J Biomed Mater Res A.第109卷(第9期),第1624段左栏第3段. *
刘春晓等.灌注法制备大鼠全肾脏脱细胞基质的研究.南方医科大学学报.2021,第29卷(第5期),979-982. *
许洁.大鼠子宫去细胞支架及其细胞外基质凝胶的制备.生物医学工程学杂志.2018,第35卷(第2期),第238页左栏第1段. *

Also Published As

Publication number Publication date
CN114214284A (en) 2022-03-22

Similar Documents

Publication Publication Date Title
CN108624561A (en) Primary tumor cell culture medium, cultural method and application
CN106574242A (en) Single cell-derived organoids
CN110475860A (en) Use the dimensional culture of the primary cancer cell of tumor tissues
WO2020187081A1 (en) Method for preparing three-dimensional biological construct, and three-dimensional biological construct and use thereof
CN102250840B (en) Human pancreatic cancer cell line and its application
TWI666448B (en) Method for predicting effect of drug
CN104531620A (en) Method for culturing lung cancer stem cells under 3D culture conditions
CN112592897A (en) Preparation method of tumor organoid
CN102329775B (en) A kind of human pancreatic cancer cell to gemcitabine resistance and application thereof
CN110878285A (en) Chip organ model for screening bladder tumor chemotherapy drugs and manufacturing method thereof
Li et al. On-chip modeling of tumor evolution: Advances, challenges and opportunities
CN114214282B (en) Method for culturing lung tumor organoids
CN104673743A (en) Tissue block culture method for obtaining primary cell from animal tissue
US20200270557A1 (en) Human in vitro orthotopic and metastatic models of cancer
CN114214283B (en) Method for culturing liver tumor organoids
Nair et al. Multi compartmental 3D breast cancer disease model–recapitulating tumor complexity in in-vitro
CN106676074B (en) Method for in-vitro amplification of liver cancer stem cells
CN114214284B (en) Method for culturing kidney tumor organoids
CN114958756B (en) Culture solution for culturing prostate cancer organoid and preparation method thereof
EP3684915A1 (en) Fabrication of a biomimetic platform system and methods of use
EP4098740A1 (en) Decellularized heart extracellular matrix-derived scaffold for culturing and transplanting heart organoid, and preparation method therefor
Leighton et al. The propagation of cancer, a process of tissue remodeling: studies in histophysiologic gradient culture
Kolahi Azar et al. The progressive trend of modeling and drug screening systems of breast cancer bone metastasis
Liu et al. Breast tumor-on-chip: from the tumor microenvironment to medical applications
Chaji Bioprinting of three-dimensional models to evaluate adipocyte breast cancer cell interactions and migration

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
GR01 Patent grant
GR01 Patent grant