This application claims priority to and the benefit of Korean Patent Application No. 10-2019-0113709, filed on Sep. 16, 2019, the disclosure of which is incorporated herein by reference in its entirety.
The present invention relates to a 3 dimensional mimetic tissue structure (Assembloid) which is defined by ‘Organoids derived from tissue stem/tumor cells reconstituted with other components of the tissue stroma/microenvironment based on patient-derived multiple cell types and a method of manufacturing the same, and more particularly, to a multicellular mimetic tissue structure based on various normal cells, which is manufactured by constituting epithelial or tumor cells with various cellular components of a microenvironment such as stromal cells, vascular cells, immune cells or muscle cells based on three-dimensional (3D) bioprinting, and a method of manufacturing the same.
For in vitro modeling of a human disease, efforts have been made to develop organoids, which are three-dimensional organ structures exhibiting structures and functions similar to in vivo tissues. Unlike conventional two-dimensional (2D) monolayer culture tending to use cells from original tissues due to genetic mutation, organoids mimic primary tissue cells and have the capacity of self-renewal to construct cellular components structures of in vivo tissues. Currently, numerous tissues such as the intestines, brain, kidneys, lungs, pancreas, stomach, liver, prostate, and bladder are modeled in vitro.
Recently, 3D culture systems for pancreatic, prostate, colon, colorectal, breast, liver and bladder tumors have been constructed due to the development of patient-derived tumor organoids derived from tumors of cancer patients. These tumor organoids retain similar pathological characteristics to actual tumors, improving the possibility of developing novel therapeutic methods for cancer treatment.
Organoids are mainly derived from tissue-restricted adult stem cells and pluripotent stem cells (PSCs), and tumor organoids are derived from tumor cells. The generation of the organoids derived from these three types of cells has been used as a method of modeling key characteristics of organs and tissues to understand various aspects of human diseases including cancer. Although these organoids represent the potential for a variety of biological studies on normal tissues and cancer, questions remain because numerous factors including a microenvironment associated with a disease in vivo cannot be explained.
Although it is possible to overcome this limitation in the microenvironment associated with an in vivo disease by using PSC-derived organoids, these are usually immature or in an embryonic state, and thus require transplantation to exhibit the normal characteristics of adult tissue. Also, there is a problem in that PSC-derived organoids cannot be applied to a tumor model.
To overcome these problems, recent studies have reported the structures of organotypic organoids including various cell components, but these organotypic organoids still do not precisely mimic in vivo tissue because they use a traditional co-culture system or a simple mixture of cellular components without an organized structure, and moreover, there is a limitation in which most of the cellular components that constitute a tissue microenvironment or stroma are lacking.
Meanwhile, in a recent study, a method of manufacturing bladder organoids (Proc Natl Acad Sci USA. 2019 Feb. 20) is actively being studied, but there are no studies on a stem cell- or tumor cell-based multicellular mimetic tissue structure realizing a microenvironment present in normal tissue or tumor tissue and a method of manufacturing the same.
Therefore, as a result of earnest research to construct an advanced mimetic tissue structure realizing a microenvironment present in normal tissue or tumor tissue in an organoid, the inventors reconstituted “Assembloid” by culturing organoids derived from stem cells or tumor cells along with the four major components of tissue stroma: stromal fibroblasts (cancer-associated fibroblasts in the case of tumor), endothelial cells, immune cells and muscle layers, first identifying a novel stem cell- or tumor cell-based 3D mimetic tissue structure, which includes organoids and microenvironment-constituting cells. Based on this, the present invention was completed.
Therefore, the present invention is directed to providing a method of manufacturing a 3D mimetic tissue structure (Assembloid), which includes the following steps:
(a) preparing an organoid;
(b) culturing the organoid in a medium containing fibroblasts and endothelial cells; and
(c) culturing the organoid cultured in step (b) in a medium containing muscle cells.
In addition, the present invention is directed to providing a mimetic tissue structure manufactured by the method.
However, technical problems to be solved in the present invention are not limited to the above-described problems, and other problems which are not described herein will be fully understood by those of ordinary skill in the art from the following descriptions.
To achieve the objects of the present invention, the present invention provides a method of manufacturing a 3D mimetic tissue structure, which includes the following steps:
(a) preparing an organoid;
(b) culturing the organoid in a medium containing fibroblasts and endothelial cells; and
(c) culturing the organoid cultured in step (b) in a medium containing muscle cells.
In one embodiment of the present invention, the organoid in step (a) may be manufactured by culturing any one of stem cells or tumor tissue.
In another embodiment of the present invention, the tumor tissue may be a bladder tumor tissue derived from a patient undergoing transurethral resection of bladder tumor (TURBT) or a cystectomy.
In still another embodiment of the present invention, the fibroblasts may be selected from the group consisting of mouse embryonic fibroblasts (MEFs) and cancer-associated fibroblasts (CAFs).
In yet another embodiment of the present invention, the muscle cells may be selected from the group consisting of primary bladder smooth muscle cells (BSMCs) and human smooth muscle cells (hSMCs).
In addition, the present invention provides a 3D mimetic tissue structure manufactured by the above method.
In one embodiment of the present invention, the tissue of the 3D mimetic tissue structure may be bladder tissue.
In another embodiment of the present invention, the tissue of the 3D mimetic tissue structure may be bladder tumor tissue.
The above and other objects, features and advantages of the present invention will become more apparent to those of ordinary skill in the art by describing in detail exemplary embodiments thereof with reference to the accompanying drawings, in which:
Hereinafter, the present invention will be described in detail.
As a result of earnest research to construct an advanced mimetic tissue structure realizing a microenvironment present in normal tissue or tumor tissue in an organoid, the inventors reconstituted “Assembloid” by culturing organoids derived from stem cells or tumor cells along with the four major components of tissue stroma: stromal fibroblasts (cancer-associated fibroblasts in the case of tumor), endothelial cells, immune cells and muscle layers, and allowed tissue reconstruction of muscle cells by culture with a single cell suspension obtained from smooth muscle cells and endothelial cells, first identifying a novel stem cell- or tumor cell-based multicellular mimetic tissue structure, which includes organoids and microenvironment-constituting cells. Based on this, the present invention was completed.
Therefore, the present invention relates to a method of manufacturing a 3D mimetic tissue structure (Assembloid), which includes the following steps:
(a) preparing an organoid;
(b) culturing the organoid in a medium containing fibroblasts and endothelial cells; and
(c) culturing the organoid cultured in step (b) in a medium containing muscle cells.
The term “organoid” used herein refers to a three-dimensional (3D) organ-like structure formed by culturing cells derived from tissue or embryonic stem cells. An organoid is a suffix that has the same meaning as an “organ” and also means “similar to an organ.” Organoids have a better arrangement of cells and their functions and have shapes and functions like functional organs due to a 3D culture method. Organoids have attracted attention with stem cell research and the development of 3D cell culture and research on optimizing growth and differentiation factors capable of differentiating into various tissues.
The term “3D mimetic tissue structure (Assembloids)” used herein is an advanced organoid containing various cell types such as epithelial or cancer cells and microenvironment tissue cells, and is used in the sense of higher-order organoids constituting in vitro 3D tissue derived from stem or tumor cells and the stroma—a tissue component inducing cell differentiation and expression of functions of the cells such as, and includes stromal cells, vascular cells, immune cells and muscle cells, but the present invention is not limited thereto.
In the present invention, the organoids in step (a) is preferably manufactured by culturing any one of stem cells or tumor tissue, and more particularly, a normal tissue organoid or tumor cell organoid is manufactured by culturing stem cells or tumor tissue.
In the present invention, the step cells used in step (a) may be selected from adult stem cells and pluripotent stem cells, and preferably, urothelial stem cells.
In the present invention, the tumor tissue used in step (a) may be tumor tissue derived from various tumors, preferably, bladder tumor tissue, and more preferably, bladder tumor tissue derived from a patient receiving transurethral resection of bladder tumor (TURBT) or a cystectomy.
In the present invention, the fibroblasts in step (b) may be cells constituting the major component of fibrous connective tissue, and preferably any one selected from the group consisting of mouse embryonic fibroblasts (MEFs) and cancer-associated fibroblasts (CAFs), but the present invention is not limited thereto.
In the present invention, the muscle cells in step (c) are preferably smooth muscle cells, which are muscle cells surrounding internal organs constituting tubes such as the stomach, digestive tract, blood vessels and bladder, and more preferably any one selected from the group consisting of primary bladder smooth muscle cells (BSMCs) and human smooth muscle cells (hSMCs), but the present invention is not limited thereto.
In addition, the present invention relates to a 3D mimetic tissue structure manufactured by the above-described method, and the tissue may be any one of intestinal tissue, intestinal tumor tissue, brain tissue, brain tumor tissue, kidney tissue, kidney tumor tissue, lung tissue, lung tumor tissue, pancreatic tissue, pancreatic tumor tissue, stomach tissue, stomach tumor tissue, liver tissue, liver tumor tissue, prostate tissue, prostate tumor tissue, bladder tissue, and bladder tumor tissue, and preferably bladder tissue or bladder tumor tissue.
In addition, the present invention provides a mouse bladder tumor mimetic tissue structure manufactured by culturing a mouse bladder tumor organoid differentiated from a BBN-induced bladder tumor in a medium containing mouse embryonic fibroblasts (MEFs), endothelial cells and tumor-reactive T cells, and a method of manufacturing the same.
The term “three-dimensional (3D) bioprinting” used herein is a technique also called cell printing or organ printing, and enables manufacture of computer-designed 3D constructs using various types of cells, biomaterials and biomolecules (Murphy, S V et al., Nature Biotechnology, 2014; 32: 773-785). This technique is attracting a great deal of attention in the field of tissue engineering for the purpose of artificial tissue or organ regeneration. Organs or tissues constituting the human body may be composed of biomaterials constituting various types of cells and the extracellular matrix. Research on regenerating required functional tissues through the manufacture of a cell construct similar to tissue constituting the human body using a 3D bioprinting technique is actively progressing. Recently, this technique has attracted a great deal of attention for developing an artificial bio-model for testing toxicity and efficacy of a new drug (Poliniab, A et al., Expert Opinion on Drug Discovery, 2014; 9(4): 335-352). In this printing process, a bio-ink is the most important key factor for precise patterning and securing a survival rate of cells in a bio printing process, and the characteristics of the ink are directly related to the process.
The inventors established the 3D mimetic tissue structure according to the present invention, manufactured by culturing normal bladder organoids and bladder tumor organoids and performing tissue reconstitution of cells constituting a microenvironment according to an embodiment, and a method of manufacturing the same.
In one embodiment of the present invention, a tissue structure similar to a reconstituted three-layered miniature bladder was confirmed in vitro, in vivo bladder-like physiological activity of the miniature bladder was confirmed, and it was confirmed that the pharmacological inhibition of a stromal Hedgehog (Hh) response using Vismodegib in the miniature bladder reduces the proliferation of epithelial cells and stromal cells by reconstituting a muscle layer of the miniature bladder, and upregulation of the stromal Hh activity using SAG increases the proliferation of epithelial cells and stromal cells, demonstrating that in vivo interactions between the epithelium and the stroma are repeated to induce cell proliferation by the three-layered miniature bladder (see Example 3).
In another embodiment of the present invention, to confirm the in vivo tumor response of a reconstituted bladder tumor organoid to a stroma-mediated, subtype-dependent anticancer agent, an in vivo tumor-like response of a reconstituted bladder tumor organoid to a stroma-mediated, subtype-dependent anticancer agent was confirmed by the stromal Hh activity effect of the bladder tumor organoid, and it was confirmed that in vivo responses of tumor organoids are precisely exhibited to various chemotherapeutic drugs due to a chemotherapeutic drug response of tumor stroma (see Example 6).
In still another embodiment of the present invention, tumor invasion and immune cell infiltration of reconstituted bladder tumor organoids having tumor microenvironments into a muscle layer were confirmed, thereby demonstrating that in vitro reconstituted organoids with stroma form platforms capable of modeling various biological aspects of tumors including muscular invasion and immune cell infiltration (see Example 8).
Hereinafter, to help in understanding the present invention, exemplary examples will be suggested. However, the following examples are merely provided to more easily understand the present invention, and not to limit the present invention.
1-1. Mice
For lineage tracing experiments, CK5CreERT2 (JAX: 018394) mice were crossed with R26Rainbow/Rainbow mice to obtain CK5CreERT2; R26Rainbow/Rainbow mice. Unless particularly stated, in all other experiments, C57BL/6 mice were used. In each experiment, mice were randomly selected from a cage for drug treatment. Procedures were performed under isoflurane anesthesia using a standard vaporizer. All procedures were performed according to the protocols approved by the Institutional Animal Care and Use Committee at POSTECH (IACUC number: POSTECH-2019-0055).
1-2. Human Bladder Tumor Samples
Human bladder tumor samples were obtained from the tissue bank of Seoul National University Hospital (SNUH). 0.5 to 1 cm3 specimens of bladder tumor tissue were acquired from patients undergoing TURB or a cystectomy according to a protocol approved by the SNUH Institutional Review Board (IRB). The tumor samples were transported to POSTECH after cryopreservation in 90% fetal bovine serum (FBS) containing 10% DMSO.
1-3. Culture of Normal Bladder Organoids
To isolate basal urothelial cells, mouse bladders were collected and inverted. The inverted bladders were incubated in DMEM supplemented with 10% FBS (Millipore) containing 500 U/mL collagenase/dispase (Sigma) at 37° C. for 2 hours, and the surface was scraped with a blade every 30 minutes. Isolated tissues were filtered through a 100 μm cell strainer (Falcon), a single-cell suspension was obtained after the lysis of red blood cells in ACK lysis buffer (Gibco), and cells were counted using a hemacytometer (Marienfeld). Urothelial cells were mixed with cold Matrigel (Growth Factor Reduced, Corning), layered onto a 24-well tissue culture dish (a 40 uL drop containing 6,000 cells) and incubated at 37° C. for 15 minutes. A pre-warmed organoid medium [advanced DMEM/F-12 (Gibco) supplemented with 10 mM HEPES (pH 7.4, Sigma), 10 mM nicotinamide (Sigma), 1 mM N-acetyl-L-cysteine (Sigma), GlutaMAX (Gibco), 1% penicillin/streptomycin (Gibco), 50 ng/mL mouse EGF (Peprotech), 0.5×B-27 (Gibco), 1 mM A8301 and 10 mM Y-27632] was added to make cells grown until analysis. In the case of subcultured organoids, bladder organoids in Matrigel were released by physical pipetting and collected in 15 mL tubes by centrifugation at 1,500 rpm and 4° C. for 5 minutes. Afterward, organoids were separated into single cells by incubation in 0.25% trypsin-EDTA (Welgene) containing Y-27632 at 37° C. for 10 minutes, followed by grinding for 1 to 2 minutes. Single cells were then seeded in Matrigel and cultured as described above. For Matrigel replacement, organoids in Matrigel were released by physical pipetting, collected in 15 mL tubes through centrifugation at 1500 rpm for 5 minutes at 4° C., mixed with fresh cold Matrigel and reseeded in the medium. The organoid medium was replaced with a fresh medium every 2 to 3 days, and Matrigel was replaced every 7 to 9 days.
1-4. Culture of Bladder Tumor Organoids
Tumor tissues obtained from patients were washed with DPBS, and then washed twice with DMEM supplemented with 10% FBS. The washed tumors were minced and incubated in DMEM containing 10% FBS and collagenase I and II (20 mg/mL each) at 37° C. for 1 hour and ground for 5 minutes every 30 minutes. The isolated tissues were spun down at 1,500 rpm for 5 minutes, resuspended in ACK lysis buffer, and then incubated for 5 minutes at room temperature. Dissociated clusters were washed with DMEM containing 10% FBS and filtered through a 100 μm cell strainer. The dissociated cell clusters were spun down, resuspended in Matrigel (Growth Factor reduced) and plated in the middle of one well of a 6-well plate. The plated drop was solidified in an incubator at 37° C. for 15 minutes, and a 2.5 mL organoid medium [advanced DMEM/F-12 supplemented with 10 mM HEPES (pH 7.4), 10 mM nicotinamide, 1 mM N-acetyl-L-cysteine, GlutaMAX, 1% penicillin/streptomycin, 50 ng/mL mouse EGF, 0.5×B-27, 1 mM A8301 and 10 mM Y-27632] was added to the well. The medium was replaced once every 2 to 3 days. For passaging, 1 mg/mL collagenase/dispase was added to the medium, followed by incubation for 1 hour at 37° C. to digest the Matrigel. Subsequently, organoids were centrifuged at 1,500 rpm for 3 minutes, washed with PBS, and then spun down. Then, 0.05% trypsin-EDTA (Welgene) was added, organoids were incubated at 37° C. for 5 minutes, followed by mechanical grinding into small cells by pipetting. The organoids were passaged in a 1:3 or 1:4 ratio every 1 to 2 weeks.
For the culture of mouse bladder tumor organoids, BBN-induced bladder tumors were ground and then incubated in DMEM containing collagenase I and II (20 mg/mL each) and thermolysin (250 KU/mL, Millipore) at 37° C. for 2 hours, and ground for 5 minutes every 30 minutes. A single-cell suspension was obtained through filtration through a 100 μm cell strainer. After the lysis of red blood cells in ACK lysis buffer, the cells were washed with DMEM containing 10% FBS and counted using a hemacytometer (Marienfeld). Single tumor cells were embedded in Matrigel, and incubated in an organoid culture medium [advanced DMEM/F-12 supplemented with 10 mM HEPES (pH 7.4), 10 mM nicotinamide, 1 mM N-acetyl-L-cysteine, GlutaMAX, 1% penicillin/streptomycin, 50 ng/mL mouse EGF, 0.5×B-27, 1 mM A8301 and 10 mM Y-27632]. To generate stocks, dissociated organoids were frozen in 90% FBS/10% DMSO, and stored in liquid nitrogen. Cryopreserved stocks were successfully recovered after freezing. It was confirmed that 40 to 50% of the cell clusters form organoids.
1-5. Reconstitution of Three-Layered Miniature Bladders
To generate normal bladder organoids with stroma, normal bladder organoids cultured for a long time (>6 months) were mixed with mouse embryonic fibroblasts (MEFs) and endothelial cells (HULEC, ATCC) at 3.75×104 cells/μL and 5×103 cells/μL, respectively, along with 3 to 4 μL of Matrigel. Before additional analysis, these bladder organoids with stroma were cultured in an organoid medium in a spinning bioreactor for 7 days. The 12-well version of the spinning bioreactor was manufactured by 3D printing according to the method described in Qian, X. et al. Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell 165, 1238-1254, doi:10.1016/j.cell. 2016.04.032 (2016). In brief, individual components such as a plate cover (Project MJP 2500, 3D Systems, USA), a rotating shaft (Single Plus, Cubicon, Korea), a fixed rod (Single Plus, Cubicon, Korea) and a gear (Single Plus, Cubicon, Korea) were printed using a 3D printer in a stackable version, and the components were assembled to manufacture a spinning bioreactor. To obtain an outer muscle layer for the development of three-layered miniature bladders, primary bladder smooth muscle cells (BSMCs) were isolated from mice. For isolation, mouse bladders were collected and inverted, and after a urothelial surface was removed by scraping with a blade, outer muscle walls were incubated in DMEM with 10% FBS containing collagenase I and II (2 mg/mL each) for 1 hour at 37° C. and ground every 15 minutes. A dissociated BSMC suspension by additional culture in DMEM with 10% FBS was filtered through a 100 μm cell strainer and centrifuged, thereby obtaining a single-cell suspension. To generate three-layered miniature bladders, each bladder organoid with stroma cultured for 7 days was put into 3 to 4 μL of a Matrigel medium containing BSMCs and HULECs at 3×104 cells/μL and 3×103 cells/μL, respectively. The miniature bladders generated as described above were cultured in a spinning bioreactor for 3 to 4 days, and then used for the experiments. To generate Rainbow miniature bladders, the bladder tissues derived from CK5CreERT2; R26Rainbow/Rainbow mice were cultured for a long time, and reconstituted with MEFs and HULECs. The reconstituted organoids were reconstituted with BSMCs derived from CK5CreERT2; R26Rainbow/Rainbow mice.
1-6. In Vivo and In Vitro UTI Models
For UTI models, UT189, which is a uropathogenic strain of E. coli (UPEC) isolated from human patients with UTI, was cultured for 4 hours with shaking, and a bacterial pellet was generated by centrifugation at 4,200 rpm for 8 minutes at room temperature. The pellet was washed twice with 5 mL PBS, centrifuged and resuspended in 1 mL physiological saline. To produce an in vivo UTI mouse model, anesthetized female mice between the age of 6 to 9 weeks were subjected to transurethral injection of UPEC at a concentration of 2×1010 CFU/mL in 50 μL. All transurethral injection procedures were performed under isoflurane anesthesia with a standard vaporizer. To generate an in vitro UTI mouse model, the organoid culture medium of the miniature bladder was replaced with an antibiotic-free fresh medium one day before microinjection. A bacterial culture was prepared as described above, and UPEC was microinjected directly into the lumen of miniature bladders using a microinjector (FemotoJet4i; Eppendorf) at an injection pressure of 40 hPa, an injection time of 0.5 s and a compensation pressure of 11 hPa. The injected miniature bladders were maintained with shaking and analyzed at indicated time points.
1-7. Lineage Marking and Tracing Experiments
To permanently label Ck5-expressing cells in vivo, 8 mg TM (based on 30 g of body weight) was orally administered to CK5CreERT2; R26Rainbow/Rainbow female mice three times a day. Five days after the last TM administration, UTI189 was transurethrally injected into the mice. At the indicated time points after injection, mice were sacrificed and bladders were collected. The collected bladders were fixed in 10% neutral-buffered formalin for 6 hours, washed with PBS three times, incubated overnight in 30% sucrose, and embedded in an OCT compound (Tissue-Tek). Tissue sections were mounted with a Prolong Gold mounting reagent (Invitrogen), and analyzed with four-color fluorescence. For in vitro lineage tracing, miniature bladders derived from CK5CreERT2; R26Rainbow/Rainbow mice were treated with 2 μM 4-hydroxytamoxifen (4-OHT) for 2 days to permanently label Ck5-expressing cells. After 4-OHT treatment, UT189 was microinjected into miniature bladders, and then the miniature bladders were collected at the indicated time points. The collected miniature bladders were fixed in 10% neutral-buffered formalin for 4 hours, washed with PBS for three times, incubated in 30% sucrose overnight, and embedded in an OCT compound. Sections were mounted with a Prolong Gold mounting reagent and analyzed with four-color fluorescence. Four-color fluorescence images were analyzed by confocal microscopy (Leica SP5 or Olympus FV1000).
1-8. Reconstitution of Bladder Tumor Organoids with Stroma
To generate patient-derived tumor organoids with stroma, human tumor organoids were cultured for 10 days, and mixed with stromal components containing patient-derived CAFs and HULECs at concentrations of 5×104 cells/μL and 4×103 cells/μL, respectively, in Matrigel. And then, 5 μL drops of tumor organoids with stroma were cultured in an organoid medium in a spinning bioreactor for 7 days. To reconstitute patient-derived bladder tumor organoids with stroma and an outer muscle layer, the drops of tumor organoids with stroma were added to 5 μL Matrigel containing human smooth muscle cells at a concentration of 5×104 cells/μL. The generated tumor organoids were cultured in an organoid medium in a spinning bioreactor until analysis.
To generate mouse bladder tumor organoids with stroma, mouse bladder tumor organoids derived from BBN-induced bladder tumors were cultured for 10 days, and mixed with stromal components containing MEFs and HULECs at concentrations of 5×104 cells/μL and 4×103 cells/μL, respectively. Subsequently, 5 μL drops of tumor organoids with stroma were cultured in an organoid medium in a spinning bioreactor for 7 days.
To generate mouse bladder tumor organoids containing stroma and tumor-reactive T cells, mouse bladder tumor tissues were cultured for 10 days, and mixed with a stromal component containing MEFs, HULECs and tumor-reactive T cells at 5×104 cells/μL, 4×103 cells/μL and 2×104 cells/μL, respectively. Subsequently, 5 μL drops of tumor organoids with stroma and tumor-reactive T cells were cultured in an organoid medium supplemented with 50 ng/mL IL-2 (Peprotech) in a spinning bioreactor for 7 days.
1-9. Drug Treatment and Response of Bladder Organoids
Normal bladder organoids were treated with 50 μM Vismodegib (Abmole), 100 nM SAG (Millipore) or DMSO and cultured in a spinning bioreactor for 7 days. Bladder tumor organoids were treated with 300 nM SAG, 5 μM FK506 (Cayman Chemical) or DMSO, and cultured in a spinning bioreactor for 7 days. Human bladder tumor organoids were cultured in an organoid medium for 7 days in a spinning bioreactor before treatment with chemotherapeutic drugs. Subsequently, tumor organoids were cultured for 48 hours in the presence of cisplatin (0, 0.2, 0.5, 1, 2, 5, 10, 50 and 100 μm, Sigma), gemcitabine (0, 0.2, 0.5, 1, 2, 5, 10, 50 and 100 μM, Sigma) or mitomycin C (0, 0.2, 0.5, 1, 2, 5, 10, 50 and 100 μM, Sigma). To produce dose-response curves for the three chemotherapeutic drugs, cell viability was assayed using a CellEvent™ Caspase-3/7 Green Detection Reagent (Thermo Fisher) according to the manufacturer's instructions. Forty-eight hours after treatment with cell therapeutic agent, bladder tumor organoids were fixed in 4% PFA at 4° C. for 15 minutes, washed with PBS three times, cultured in 30% sucrose overnight, and embedded in an OCT compound. Capase-3/7-stained cells in the bladder tumor organoids were counted based on green fluorescence signals, and dose-response curves were generated using GraphPad Prism ver.6.
1-10. Differentiation of Human Smooth Muscle Cells
H9 human embryonic stem cells (hESCs) were cultured to 30% confluence in an mTeSR (STEMCELL) medium on a Matrigel-coated plate with daily medium replacement. Mesodermal lineage differentiation was initiated by culturing the cells with DMEM/F12 (Gibco) supplemented with knockout Serum Replacement (Invitrogen), 1×MEM non-essential amino acids (Gibco), β-mercaptoethanol (Gibco), 1×Glutamax, 1% penicillin/streptomycin, 10 μM Y-27632, 10 ng/mL activin A (Peprotech) and 20 ng/mL BMP4 (Peprotech). Afterward, human smooth muscle cells (hSMCs) were differentiated by culturing the cells in DMEM/F12 containing 5% FBS, 1% penicillin/streptomycin, 5 ng/mL PDGF-BB (Peprotech) and 2.5 ng/mL TGFβ (Peprotech) for 3 to 14 days. The medium was replaced every 2 days.
1-11. Reconstitution of Bladder Tumor Organoids by 3D Bioprinting
Tumor cells derived from patient-derived bladder tumor organoids were embedded in 40 μL Matrigel at a density of 5×105 cells/mL, and cultured in an organoid culture medium for 10 days. To prepare a bio-ink mixture for the reconstitution of tumor organoids with stroma by 3D bioprinting, 6 wells containing the embedded tumor organoids were treated with 0.5 mg/mL of a collagenase/dispase solution at 37° C. for 30 minutes, washed and centrifuged. The collected organoids were resuspended in 1 mL Matrigel (Growth Factor Reduced) containing patient-derived CAFs and HULECs at 4×107 cells/mL and 4×106 cells/mL, respectively. The reconstituted bio-ink was put into a sterilized 10 mL syringe with a 20-gauge needle, and stored on ice for less than 5 minutes before printing. A jet-type 3D printer (in vivo, Rokit, Korea) was used to reconstitute tumor organoids with stroma as described above. A G-code based script was written by Creator K software (Rokit, Korea) to print a 5×6 array of 10 μL drops. Each drop was dispensed on a parafilm at 8-mm intervals by extrusion. The temperature of a syringe insulator was set to 4° C. throughout the process. To avoid sedimentation of cells in the bio-ink mixture, each array was printed within 3 minutes. After dispensing, drops were inverted and crosslinked in an incubator (37° C., 5% CO2) for 15 minutes. To exclude unstable products, the first two drops were discarded, and the remaining product was separately transferred to a 12-well spinning bioreactor containing a 2 mL organoid culture medium. The culture plate was incubated at 37° C., and the culture medium was replaced every 2 days until analysis.
1-12. Generation of Tumor-Reactive T Cells
Total lymph nodes and spleens were isolated from C57/BL6 mice. Tissues were ground with 3 mL PBS and filtered through a 100 μm cell strainer. Cell suspensions were collected in a 15 mL tube, and cell pellets were generated by centrifugation at 1,500 rpm and 4° C. Red blood cells in the lymphocyte pellets were removed with ACK lysis buffer. The isolated lymphocytes were cultured overnight with 10% FBS-containing RPMI (Welgene) supplemented with 5 ng/mL IL-2 in a 96-well tissue culture plate pre-coated with 0.5 μg/mL anti-CD3 (BD) and 0.5 μg/mL anti-CD28 (BD) at 37° C. To prepare the tumor cells for stimulating lymphocytes, BBN-induced mouse bladder tumor organoids from isogenic C57BL/6 mice were cultured for 7 days and treated with 200 ng/mL IFNγ (Peprotech) overnight at 37° C. The next day, the stimulated tumor organoids were separated into single cells and resuspended in a lymphocyte medium. The dissociated tumor cells and lymphocytes were co-cultured at a 1:20 ratio for 2 weeks in the presence of anti-CD28, IL-2 and 20 μg/mL anti-PD-1 (Bio X Cell) antibodies.
1-13. Flow Cytometry Analysis
The lymphocytes co-cultured with the tumor cells were collected, and re-stimulated with a cell stimulation cocktail for 4 hours in the presence of a protein transport inhibitor (eBioscience). Afterward, the cells were washed twice with FACS, and a surface marker was stained for 30 minutes at 4° C. For intracellular staining, a single-cell suspension was fixed, and permeability was maintained with an eBioscience staining buffer set. The following antibodies were used: anti-CD4-BUV395 (BD, GK1.5), anti-Zombie-Aqua (BioLegend), anti-CD8a-BV650 (BD, 53-6.7), anti-CD45.2-BV605 (BD, 104), anti-B220-BV710 (BD, RA3-6B2), anti-CD11b-PerCP-Cy5.5 (BD, M1/70), anti-TCRβ-APC-Cy7 (BD, H57-597), anti-CD69-PacificBlue (BioLegend, H1.2F3) and anti-IFNγ-PE (eBioscience, XMG1.2). The cells were analyzed on LSR II (BD) and data was processed with FlowJo software (Tree Star).
1-14. Quantitative RT-PCR
Tissues were homogenized by grinding and trypsin treatment, and RNA was extracted using an RNeasy Plus Mini Kit (Qiagen). The RNA samples were dissolved in RNase-free water, and then their concentrations and purities were measured using a spectrophotometer. For quantitative RT-PCR of mRNA transcripts, primary cDNA was synthesized using a High-Capacity cDNA Reverse Transcriptase Kit (Applied Biosystems) containing oligo DT. Quantitative RT-PCR was performed using SYBR Green Supermix (Applied Biosystems) and a one-step cycler (Applied Biosystems). Gene expression was normalized to a housekeeping gene HPRT.
1-15. Histological Analysis
Tissue specimens were pre-fixed in 10% neutral-buffered formalin for 24 hours and embedded in paraffin. To form agarose blocks, organoids were embedded in a 3% agarose gel, fixed in 10% neutral-buffered formalin for 24 hours and embedded in paraffin. The paraffin block was cut into 4-μm sections using a microtome. Slides were stained with hematoxylin and then counter-stained with eosin for histological analysis.
1-16 Immunohistochemistry
Tissues and organoids were fixed in 4% PFA, embedded in an OCT compound, and cut into 10 to 25-μm sections using a Microm cryostat (Leica). For immunohistochemistry, frozen sections were post-fixed in 4% PFA at 4° C. for 20 minutes. The sections were washed with PBS three times, blocked with 2% goat serum and PBS containing 0.25% Triton X-100 (PBS-T) for 1 hour, and incubated with primary antibodies diluted in blocking buffer in a humidified chamber overnight at 4° C. Primary antibodies diluted in blocking buffer are as follows: rat anti-Ck18 (1:500, DSHB); rabbit anti-Ck5 (1:500, Abcam); chicken anti-vimentin (1:500, Millipore); mouse anti-uroplakin 3 (1:50, Fitzgerald); mouse anti-alpha smooth muscle actin (1:200, Abcam); hamster anti-CD31 (1:200, Abcam), rabbit anti-Ki67 (1:500, Abcam) and rabbit anti-caspase 3 (1:200, Cell Signaling). Afterward, the sections were washed three times with 0.25% PBS-T, and incubated with Alexa Fluor 488, 594, 633 or 647-conjugated secondary antibodies, which were suitably diluted 1:1,000, in 0.25% PBS-T, along with DAPI, at room temperature for 1 hour. The conjugated antibodies, anti-CD8a-BV510 (1:200, BD), were incubated in 0.25% PBS-T along with DAPI at room temperature for 1 hour. The sections were washed twice with 0.25% PBS-T, and mounted on slides with a Prolog Gold mounting reagent.
1-17. Data Analysis
Statistical analysis was performed using GraphPad Prism ver.6. All data is expressed as +/−s.e.m. The comparison between groups was performed by a two-tailed Student's test. P<0.05 was considered statistically significant.
2-1. Confirmation of Mature Bladder Tissue Differentiation of Urothelial Stem Cell
To confirm urothelial stem cells capable of forming a bladder tissue structure in vitro, and confirm whether organoids generated by the stem cells develop into mature bladder tissue including multiple layers of similarly differentiated epithelial cells as mature bladder tissue in vivo, as shown in
In addition, the bladder organoids cultured for 9 days were further cultured for 9 days, and the bladder organoids cultured for 18 days were analyzed by immunostaining for the basal epithelial marker Ck5 and the luminal epithelial marker Ck18.
As a result, as shown in
2-2. Confirmation of Long-Term Growth of Bladder Cell Membrane Organoid without Serial Passaging
As shown in
In addition, as shown in
From this result, it was confirmed that the long-term cultured bladder organoids recapitulate the normal bladder development from an embryonic state to an adult state.
In addition, it was confirmed that the long-term cultured bladder organoids represent the mature urothelial cells of the bladder having multiple layers of epithelial cells as well as Upk+ and Ck18 differentiated from luminal cells in an inner layer that is the line of a luminal space and an outer layer that contains a Ck5+ basal layer.
From the result of Example 2, a long-term culture system of normal bladder organoids that continuously expand normal bladders, self-organize a urothelial structure, and mimic mature urothelial cells was confirmed.
3-1. Confirmation of In Vivo Bladder-Like Tissue Structure of In Vitro Reconstituted Three-Layered Miniature Bladder
The inventors intended to determine that tissue stroma is a critical tissue component serving as a gap between stem cells for stimulating cell proliferation and differentiation and providing structural support, and to develop bladder organoids including tissue stroma. As shown in
The cultured organoids were analyzed by immunohistochemical analysis, and as shown in
In addition, similar to the reconstituted normal urothelial cells and the long-term urothelial organoids (
From this result, it was confirmed that bladder organoids showed smooth and flat Ck5+ epithelial layers (
3-2. Confirmation of In Vivo Bladder-Like Physiological Activity of In Vitro Reconstituted Three-Layered Miniature Bladder
Cell proliferation in the bladder requires a stromal Hedgehog (Hh) response, and is mediated by Hh/Wnt signaling feedback between the epithelium and stroma. To confirm whether the reconstituted bladder organoids showed stroma-mediated cell proliferation as the in vivo bladder, reconstituted bladder organoids were treated with Vismodegib or SAG, which pharmacologically inhibit the reconstituted bladder organics, was treated to activate the substrate Hh reaction
As a result, as shown in
From this result, it was confirmed that the reconstituted bladder organoids physiologically recapitulate the interaction between the epithelium and stroma of the normal bladder by maintaining functional stroma required for reciprocal Hh/Wnt signaling feedback.
3-3. Development of Muscle Layer of In Vitro Reconstituted Three-Layered Miniature Bladder
To further develop miniature bladders that precisely mimic the tissue structure of the adult bladder with the tissue structure of an outer muscle layer, as shown in
As a result, as shown in
From this result, as shown in
3-4. Confirmation of Hh Pathway Activity Response of In Vitro Reconstituted Three-Layered Miniature Bladder
To further confirm that the three-layered miniature bladders respond to stromal Hh pathway activity, similar experiments as described in Example 3-2 were performed using the reconstituted bladder organoids with stroma.
As a result, as shown in
4-1. Confirmation of Recapitulation of Injury-Induced In Vivo Interaction Between Epithelial Cells and Stromal Cells of Three-Layered Miniature Bladder
To confirm the possibility of establishing an in vitro UTI model and recapitulating injury-induced the in vivo interaction between epithelial cells and stromal cells in a three-layered miniature bladder, as shown in
The Ck18+ umbrella cells of a wild-type bladder were exfoliated by bacterial infection, and as shown in
As a result, as shown in
4-2. Confirmation of Regulation of Regenerative Response of Miniature Bladder In Vitro by Injury Induction Between Endothelial Cells and Stromal Cells
To confirm whether injury induction between endothelial cells and stromal cells whose activity are increased by Hh/Wnt signaling feedback regulates the regenerative response of a miniature bladder in vitro, the expression of various genes involved in Hh/Wnt signaling activity was investigated.
As a result, as shown in
In addition, as shown in
4-3. Confirmation of Tissue Dynamics of Injury-Induced Urothelial Cell Regeneration by Clonal Analysis Using In Vivo and In Vitro Models of UTI
To investigate stem cells and tissue dynamics for urothelial regeneration, as shown in
Accordingly, as shown in
As shown in
4-4. Confirmation of Reproduction of Clonal Expansion of Basal Cells Regenerating Urothelial Cells in In Vitro UTI Model Developed Using Three-Layered Miniature Bladder
To confirm clonal expansion reproduction of basal cells regenerating urothelial cells in an in vitro UTI model developed using a three-layered miniature bladder, as shown in
As shown in
As a result, as shown in
From this result, as shown in
5-1 Immunohistochemical Analysis of Bladder Tumor Organoids
As shown in Table 1, in addition to normal bladder organoids, 8 invasive urothelial carcinoma patient-derived bladder tumor tissues were acquired from fresh patient TURB or radical cystectomy samples.
As shown in
As shown in
As shown in
In addition, as shown in
5-2. Confirmation of Pathophysiology and Mimetic Structure of Parenteral Tumor of Bladder Tumor Organoid
To establish patient-specific tumor organoid models precisely mimicking the pathophysiology and structure of parenteral tumors, as shown in
A tumor structure in reconstituted bladder tumor organoids was analyzed, and responses of the reconstituted bladder tumor organoids were compared with orthotopically transplanted xenografts and parenteral tumors by H&E assay and immunostaining for Ck5 and Ck18 to mark tumor cells, vimentin for CAFs and CD31 for endothelial cells.
As a result, as shown in
In addition, as shown in
In addition, as shown in
As shown in
As shown in
As shown in
As shown in
As shown in
From this result, it was confirmed that tumor organoids which matched CAFs and three-dimensionally reconstituted from different subtypes of invasive urothelial carcinoma show the intrinsic structure and histopathology of parental tumors, which were not observed in an organoid-only culture condition.
6-1. Confirmation of Stromal Hh Activating Effect of Bladder Tumor Organoid
As shown in
To examine the effect of an increased Bmp response in tumor cells, bladder tumor organoids with or without tumor stroma were treated with FK506, known to stimulate a Bmp response.
As a result, as shown in
To further examine the effect of stromal Hh activity on bladder tumor growth, bladder tumor organoids were treated with SAG, which is a Hh pathway agonist.
As a result, as shown in
This result revealed that reconstituted bladder tumor organoids show an in vivo tumor-like response to a stroma-mediated, subtype-dependent anticancer agent, demonstrating that a stromal Hh response induced stromal expression of Bmp to elicit a tumor-suppressing effect in basal tumors, and a tumor-suppressing effect by subtype conversion from invasive tumors to less aggressive luminal subtypes.
6-2. Confirmation of Chemotherapeutic Response of Tumor Stroma
As shown in
As shown in
In addition, as shown in
From this result, it was confirmed that chemotherapeutic drugs are poorly delivered to a cancer tissue site due to the surrounding stroma, which is more extensive and denser than normal tissues in many solid tumors, and tumor organoids can precisely exhibit in vivo responses to various chemotherapeutic drugs.
7-1. Confirmation of Histopathology and Tumor Structure of 3D Bioprinting-Based Reconstituted Bladder Tumor Organoid
To develop a high-throughput platform for reconstituted tumor organoids, a method of automatically generating multiple reproducible tumor organoids with stroma was designed using a 3D bioprinting technique, and to this end, as shown in
As a result, as shown in
7-2. Confirmation of In Vivo Tumor Responses of 3D Bioprinting-Based Reconstituted Bladder Tumor Organoid to Stroma-Mediated Subtype-Dependent Anticancer Agent
In addition to examining histopathology and tumor structure, it was confirmed whether bioprinted bladder tumor organoids exhibit in vivo tumor responses to tumor stroma-mediated, subtype-dependent anticancer agents.
As a result, as shown in
In addition, as shown in
As a result, as shown in
From this result, it was confirmed that 3D bioprinting-based reconstitution of patient-derived bladder tumor organoids formed functional tumor organoids that exhibit various characteristics of parental tumors, such as original tumor structures, tumor stroma-mediated anticancer effects and responses to chemotherapeutic drugs.
8-1. Confirmation of Reconstitution of Muscle Layer of Reconstituted Tumor Organoid Platform
A muscle layer was reconstructed to exhibit muscle invasion of tumor cells in an in vivo tumor organoid model. Tumor organoids derived from two different lines, consisting of a P-7 line for the luminal type at the T1 stage and a P-3 line for the basal type at the T2 stage were reconstituted by matching with patient-derived CAFs.
As shown in
As a result, as shown in
8-2. Confirmation of Repopulation of T Cell-Based Immune Microenvironment in Reconstituted Tumor Organoid Platform
To confirm whether a T cell-based immune microenvironment was able to be repopulated in a reconstituted tumor organoid platform, reconstitution of tumor organoids including tumor-specific T cells was performed. To confirm reconstitution of T cells in the tumor organoids, mouse tumor organoids were reconstituted in a bladder cancer mouse model such as an N-butyl-N-4-hydroxybutyl nitrosamine (BBN)-induced mouse model, and comparative analysis was performed.
As shown in
In addition, as shown in
8-3. Confirmation of Reconstitution of Tumor-Reactive T Cells in Mouse Tumor Organoids
In the mouse tumor organoids, to confirm further reconstitution of tumor-reactive T cells, tumor-reactive T cells were prepared by co-culturing urothelial carcinoma-derived mouse tumor cells with isogenic primary lymphocytes in a BBN-induced mouse for 2 weeks.
As shown in
As a result, as shown in
In addition, as shown in
From this result, it was confirmed that in vitro organoids with stroma were reconstituted to form a potent platform for modeling various biological aspects of tumors including muscle invasion and immune cell infiltration.
It is confirmed that a stem cell- or tumor cell-based multicellular mimetic tissue structure designed according to the present invention mimics physiological and pathological characteristics of in vivo tissues by realizing the major factors of a tissue microenvironment such as stromal cells, vascular cells, immune cells, and muscle cells in the existing organoid through tissue reconstruction, and the mimetic tissue structure is expected to be effectively used as a platform for new drug development and a disease model by culturing normal tissue and tumor tissue.
It should be understood by those of ordinary skill in the art that the above description of the present invention is exemplary, and the exemplary embodiments disclosed herein can be easily modified into other specific forms without departing from the technical spirit or essential features of the present invention. Therefore, the exemplary embodiments described above should be interpreted as illustrative and not limited in any aspect.
Number | Date | Country | Kind |
---|---|---|---|
10-2019-0113709 | Sep 2019 | KR | national |