Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

Matrigel-based organoid culture of malignant mesothelioma reproduces cisplatin sensitivity through CTR1

verfasst von: Fumiya Ito, Katsuhiro Kato, Izumi Yanatori, Yuki Maeda, Toyoaki Murohara, Shinya Toyokuni

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Organoids are a three-dimensional (3D) culture system that simulate actual organs. Therefore, tumor organoids are expected to predict precise response to chemotherapy in patients. However, to date, few studies have studied the drug responses in organoids of malignant mesothelioma (MM). The poor prognosis of MM emphasizes the importance of establishing a protocol for generating MM-organoid for research and clinical use. Here, we established murine MM organoids from p53+/- or wild-type C57BL/6 strain by intraperitoneal injection either with crocidolite or carbon nanotube. Established MM-organoids proliferated in Matrigel as spheroids. Subcutaneous injection assays revealed that the MM-organoids mimicked actual tissue architecture and maintained the original histological features of the primary MM. RNA sequencing and pathway analyses revealed that the significant expressional differences between the 2D- and 3D-culture systems were observed in receptor tyrosine kinases, including IGF1R and EGFR, glycosylation and cholesterol/steroid metabolism. MM-organoids exhibited a more sensitive response to cisplatin through stable plasma membrane localization of a major cisplatin transporter, copper transporter 1/Slc31A1 (Ctr1) in comparison to 2D-cultures, presumably through glycosylation and lipidation. The Matrigel culture system facilitated the localization of CTR1 on the plasma membrane, which simulated the original MMs and the subcutaneous xenografts. These results suggest that the newly developed protocol for MM-organoids is useful to study strategies to overcome chemotherapy resistance to cisplatin.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-023-10966-4.
Fumiya Ito and Katsuhiro Kato contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BP
2-Bromopalmitate
CSC
Cancer stem cell
CTR1
Copper transporter 1 (SLC31A1)
EGF
Epidermal growth factor
EGFR
Epidermal growth factor receptor
EMT
Epithelial mesenchymal transition
ESC
Embryonic stem cell
FBS
Fetal bovine serum
FACS
Fluorescence-activated cell sorting
FFPE
Formalin-fixed paraffin-embedded
FGF
Fibroblast growth factor
GFP
Green fluorescent protein
GSEA
Gene set enrichment analysis
HA
Hyaluronic acid
H&E
Hematoxylin and eosin
HC
Hydrocortisone
IF
Immunofluorescence
IP
Intraperitoneal(ly)
iPSC
Induced pluripotent stem cell
MSLN
Mesothelin
MM
Malignant mesothelioma
PBS
Phosphate-buffered saline
PFA
Paraformaldehyde
SC
Subcutaneous(ly)
SDS-PAGE
Sodium dodecyl sulfate–polyacrylamide gel electrophoresis
TM
Tunicamycin

Introduction

Organoid technology bridges the gap between traditional two-dimensional (2D) cell line cultures and in vivo models [13]. Organoids are small 3D tissues that can be generated from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), adult stem cells and cancer cells [4]. Organoids maintain intrinsic properties of the primary tumors, including self-renewal, multilineage differentiation, signaling nodes and histological structures. In addition to displaying a specific morphology, such as microvilli and tight junction, organoids exhibit apical-basal cellular polarity. Regarding cancer biology, organoids are expected to present various applications in translational studies under precision medicine.
Malignant mesothelioma (MM) is an extremely lethal tumor [5], arising from the mesothelial cells of pleura, peritoneum or pericardium, which has been linked to asbestos exposure [6, 7]. Recently, we identified the iron-rich mutagenic milieu, resulting from the accumulation of asbestos-fed macrophages with myofibroblasts, leading to granuloma [8, 9]. This granuloma formation is accompanied by increased stiffness and extracellular matrix deposition [10], and the stiffness indeed contributes to the malignant phenotype in tumor biology [11, 12]. Therefore, experimental cultures may expect a similar stiffness for accurate results [13]. One of the major problems in cancer treatment is the presence of cancer stem cells (CSCs). CSCs are resistant to chemotherapy and are involved in cancer initiation and metastasis [14, 15]. Therefore, many studies have focused on the biology of CSCs to target them in cancer treatment. Although poor clinical prognosis of MM requires novel therapeutic approaches, not much is thus far known on the characteristics of both normal mesothelial and MM stem cells.
Cisplatin has been widely used in cancer therapy, including the first line chemotherapy for MM [5]. Homozygous deletion of the 9p21 locus in humans that harbors genes, encoding CDKN2A and CDKN2B, are the most common genetic alterations in MM [7]. Of note, miR-31 is located also at the same locus, which promotes cisplatin sensitivity through simultaneous loss of miR-31 [16]. CTR1 (SLC31A1) contributes to cisplatin uptake in the renal tubular cells during nephrotoxicity, a major side effect of cisplatin [17]. Mammalian CTR1 mRNA expression has been observed in all the examined tissues with kidney and liver at high levels and with brain and spleen at low levels. Reportedly, cisplatin is preferentially taken up through the basolateral membrane in the renal tubular cells [1820]. Cisplatin uptake is thus associated with cellular polarity. Accordingly, a model MM system, replicating cellular polarity, is essential to further understand the mechanisms of cisplatin resistance in MM. However, there are thus far no established protocols for organoid development in MM.
Here we started from establishing a protocol to generate MM organoids from mice. Mouse MM organoids is effective as pre-clinical models because MM is a rare tumor in Japan with the incidence of ~ 2,000 per year in 126 million population [7]. Fortunately, human and mouse CTR1 are highly homologous. After the first 37 residues, only four residues in mouse Ctr1 (mCtr1) are different from those of human CTR1 (hCTR1) [21]. These newly generated MM-organoids would potentially provide us with an understanding of cisplatin-transport dynamics in MM, thereby highlighting the merits of organoid culture to study clinical responses to cisplatin treatment and the chemotherapy resistance.

Materials and methods

Materials

The materials and kits used are as follows. EMEM (Wako, 051–07,615, Osaka, Japan), FBS (Biowest, S1810-500; Nuaillé, France), Tet system Approved FBS (TaKaRa, 631,107, Shiga, Japan), Autofluorescence Quenching Kit (Vector, SP-8400, Burlingame, CA), AurumTM Total RNA Mini Kit (BioRad, 64,337,836, Hercules, CA), NEBNext Ultra II Directional RNA Library prep kit for Illumina (BioLabs, E7760S, Boston, MA), NEBNext Multiplex Oligos for Illumina (BioLabs, E6440S), anti-CTR1 (CST, #13,086, Danvers, MA, Novus, #NBP2-36,573), anti-Flag (MBL, PM020, M185-3L, Nagoya, Japan), anti-golgin97 (CST, #13,192), anti-mouse mesothelin (IBL, #28,127, Gunma, Japan), anti-WT1 (Thermo Fischer Scientific, MA5-32,215), anti-ITGβ1 (CST, #9699), anti-IGFR (Proteintech, 20,254–1-AP, Chicago, IL), anti-β-catenin (CST, #8480 T), anti-GFP (MBL life science, #598), anti-mouse IgG Alexa488 (ThermoFischer Scientific, A-11108), anti-rabbit IgG Alexa 568 (Thermo Fischer Scientific, A-11011), anti-rabbit IgG Alexa 633 (Thermo Fischer Scientific, A-21070) CF®568 Phalloidin (biotinum,00064 T), Receptor Tyrosine Kinase Antibody Sampler Kit (CST, #42,344), Epithelial-Mesenchymal Transition (EMT) Antibody Sampler Kit (CST, #9782), anti-β-actin (Sigma Aldrich, P2120, St. Louis, MO) Advanced F12/DMEM (Thermo Fischer Scientific, 12,634,010), recombinant Murine R-Spondin-1 (PeproTech Inc., 315–32, Cranbury, NJ), recombinant Murine Noggin (PeproTech Inc., 250–38), recombinant Murine EGF (PeproTech Inc., 315–09), recombinant Wnt3A (PeproTech Inc., 315–20-2ug), recombinant FGF-basic (PeproTech Inc., 450–33), B-27™ Supplement (50x) (Thermo Fischer Scientific, 17,504,044), N-2 Supplement (100x) (ThermoFischer Scientific, 17,502,048), A8301 (Peprotech Inc., 9,094,360), N-Acetyl-L-cysteine (Wako, 017–05,131) CTS™ GlutaMAX™-I Supplement (ThermoFischer Scientific, A1286001), CultureSure® Y-27632 (Wako, 030–24,021) and Corning® Matrigel® Growth Factor Reduced (GFR) (Corning, 354,230) were used.

Cell experiments

Mouse mesothelial cell lines were established from the C57BL/6 mice, using the previously described enzymatic method [2224]. Briefly, omentum and major intraperitoneal organs, including stomach, spleen and pancreas, were excised from 6 ~ 8-week-old mice, which was followed by careful separation of mesothelium, using scissors. Approximately 20 to 30 mesothelial fragments were isolated from each isolated serosal preparation, of which roughly 80% were found to attach to the Nunclon™ Delta-treated culture plate and to start proliferation with DMEM containing 10% FBS after overnight culture. The mesothelial cells were immortalized using SV40 as well as small and large T antigen (Addgene#22,298). Mouse MM cell lines were established also from p53-heterozygous knockout (p53+/- )mice as described below. All the cell lines were cultured under standard culturing conditions (37 °C, humidified atmosphere, 5% CO2) in EMEM containing 10% FBS and 1% antibiotic–antimycotic (Gibco). Cells were frozen in CellBanker (TakaraBio), thawed and used for experiments within two months.

MM carcinogenesis model in mice

MM carcinogenesis in mice were performed as previously described with some modifications [2527]. P53-heterozygous knockout (p53+/- mice were obtained from RIKEN BRC (RBRC01361 [28]) and were maintained by mating with normal wild-type C57BL/6 female mice (SLC, Shizuoka, Japan). Fourteen p53+/- and 10 p53+/+ male mice at the age of 6 ~ 8 weeks were injected intraperitoneally (ip) with a single dose either of 3.0 mg crocidolite (UICC) or 1.5 mg of multi-walled carbon nanotube of 50 nm-diameter (NT-50; VGCF-s) [29] in 3 ml of 0.5% bovine serum albumin/saline suspension. The vehicle solution (3 ml) was injected into 9 mice as negative controls. The mice were euthanized by cervical dislocation under isoflurane-anesthesia when they revealed massive ascites or 5% weigh loss in a week. Mouse that did not exhibit these phenotypes were euthanized after approximately 1y (365 d) had passed from the initial injection of fibers. All the animal experiments were approved by the animal experimental and ethics committee of Nagoya University Graduate School of Medicine (M220346-004, 2022/03/18).

Isolation of MM cells

Immediately after euthanizing the animals, tissue fragments of ~ 100–300 mm3 were excised from the induced MM. Non-necrotic areas with solid growth were selectively collected and processed as follows. Tissue fragments were cut into 2–3 mm pieces, washed with cold PBS several times and finally dissociated into small cell clusters or single cells through digestion with 2 U/ml dispase II and 1 mg/ml collagenase P (SigmaAldrich, 11,213,857,001) for 30 min at 37 °C. Dissociated cells were washed again with PBS and recovered as a pellet by centrifugation (500 × g for 5 min). If necessary, further digestion was performed using Accumax (Innovative Cell Technologies, San Diego, CA) for 5 min at 37 °C, washed with ice-cold PBS and recovered as a pellet by centrifugation (500 × g for 5 min). Erythrocytes were removed using hypotonic buffer (168 mM NH4Cl, 10 mM KHCO3, 81.8 µM EDTA-4Na). The number of wells to seed the MM cells was determined depending on the number of cell pellets recovered. Typically, 2 to 4 wells in a 48-well plate were used to initiate the primary organoid culture.

Organoid culture of MM cells

The culture media used for organoids was advanced DMEM/F12 (Thermo Fischer Scientific) supplemented with 50 ng/ml recombinant murine EGF (Peprotech), 100 ng/ml recombinant murine R-spondin1 (R&D, Minneapolis, MN) and 100 ng/ml recombinant murine Noggin (Peprotech), 10 µM Y-27632 (Wako). All the following experiments used this organoid medium. Organoids were cultured according to the general organoid-culture method established previously (Fig. 1f) [30]. The cells were resuspended in 250 µl of media and plated on 25 µl of solidified Matrigel (BD Bioscience, Franklin Lakes, NJ) per well in a 48-well plate and incubated overnight at 37 °C. Subsequently, floating non-viable cells were discarded along with the media. In instances when floating tissue fragments and cell aggregates appeared to contain numerous viable cells, they were recollected and digested with Accumax (Innovative Cell Technologies, San Diego, CA) for 5 min at 37 °C along with vigorous pipetting. For each passage, the cells were cultured until 70–80% confluency, from which 5 × 105 ~ 2 × 106 cells were typically harvested. Depending on the proliferation rate, the cells were diluted at 1:1 to 1:4 with culture media. For each passage, organoids, Matrigel and media were collected directly using a cell scraper, washed with PBS and then dissociated into single cells using Accumax treatment for 5 min at 37 °C along with vigorous pipetting. MM-derived organoids were cryopreserved in Bambanker (GC Lymphotec, Tokyo, Japan) supplemented with 10 µM Y-27632 and stored at − 80 °C until use. The organoids were thawed and used for experiments within two months (passenger times < 20).

Pathological analysis

Organoids were first isolated by de-polymerizing the Matrigel with Cell recovery Solution (BD Bioscience, Franklin Lakes, NJ) and then resuspended in iPGell (Genostaff, Tokyo, Japan). The iPGell-embedded organoids or the resected tumors were fixed in 4% paraformaldehyde (PFA), dehydrated and then embedded in paraffin. The formalin-fixed paraffin-embedded (FFPE) samples were cut into sections with 4-µm thickness and then stained with hematoxylin and eosin (H&E), alcian blue (pH 2.5) or mucicarmine staining. Hyaluronic acid was detected by hyaluronic acid binding protein using the manufacturer's protocol (Amsbio, AMS.HKD-BC41, Abingdon, UK). Immunohistochemical stainings were performed with a Leica BOND Max (Leica Microsystems, Wetzlar, Germany) as described [31].

Immunofluorescence staining

Cells were plated on coverslips with or without matrigel. After 18–24 h incubation, cells were fixed with 4% PFA for 10 min/4℃, permeabilized for 5 min in 0.2%Triton X-100 and then blocked with 3% BSA in PBS for 15 min. Cells were stained with specific antibodies namely anti-FLAG, anti-ITGB1, and anti-CTR1 at a dilution following the manufacture`s recommendation for 30 min at room temperature. Then, secondary antibodies conjugated to Alexa 488, Alexa 568, or Alexa 647 were used at 1:1000 for 30 min at room temperature with Hoechst 33,342 at 1:10,000. Following a final wash in H2O, glass coverslips were mounted with VECTASHIELD Antifade Mounting Medium (H-1900) (Vector Lab Inc., Newark, CA). Images were obtained using Olympus SpinSR10 confocal microscope and 100x/1.4NA oil immersion objective or IX81 inverted microscope (Olympus) and 60 × /1.42 NA oil objective lens. Images were analyzed using the image processing package, Fiji (https://​fiji.​sc/​).

Cloning of mCTR1

The amino acid coding regions of mouse CTR1, was amplified using cDNA from mouse normal mesothelial cells. mCTR1 was PCR-cloned into pCIG3 modified vector as the same of vector construction section. Additionally, mCTR1 was PCR-amplified to insert a FLAG epitope (NH2-DYKDDDDK-COOH) at the amino terminus of mCTR1. Mutagenic primers were designed to introduce single amino acid substitution at amino acids N23D and N23Q (Table 1). Mesothelial cells, MM cells and MEF cells with CTR1 overexpression or mutated CTR1 were established, using either a pCIG3 modified lentiviral vector (Addgene, #78,264) or pcDNA3.2.
Table 1
Primer sequences for cloning and mutagenesis of mCTR1
Primer
Primer orientation
Nucleotide sequence
N-terminal Flag mCTR1
Forward
5'-GAATTCGCCACCATGGACTACAAAGACGATGACGACAAGAACCA-3'
Reverse
5'-GGATCCTCAATGGCAGTGCTCTGTGA-3'
mCTR1
Forward
5'-GAATTCGCCACCATGAACCATATGGGGATGAACCATATGG-3'
Reverse
5'-GGATCCTCAATGGCAGTGCTCTGTGA-3'
N23D
Forward
5'-ATTACCATGCCACCTCACCACCAC-3'
Reverse
5'-GTCGTCGTCCGTGTGGTTCATACCCATATG-3'
N23Q
Forward
5'-ATTACCATGCCACCTCACCACCAC-3'
Reverse
5'-CTGGTCGTCCGTGTGGTTCATACCCATATG-3'
Primers were designed for generating different mouse CTR1 constructs, which were ligated into the appropriate epitope-tag sequences and then cloned into pcDNA3.2 vector

Cell proliferation and drug sensitivity assays

Regarding in vitro assays, organoids were digested in Accumax for 5 min at 37 ℃ and thereafter in cell recovery solution for 20 min on ice. The dissociated cells were counted using the Countess II FL (Thermo Fischer Scientific). For the cell proliferation assay, 2 × 104 cells were plated in duplicate in a 24-well plate containing solidified Matrigel to form organoids. Data were collected at day 7 and normalized to the corresponding value of day 1. Regarding the drug sensitivity assay, 5 × 103 single cells were plated into a 96-well plate in triplicate. At 48 h after plating, cisplatin (Wako) was added at five serially diluted concentrations, varying from 5 to 100 nM or from 1 to 100 µM. Cell viability was analyzed using the AlamarBlue® Cell Viability Reagent (Thermo Fischer Scientific) after 24 h-incubation.

Tumorigenicity assay

Immunodeficient nude mice KSN/Slcnu/nu were purchased from SLC Japan (4 ~ 6 weeks old; Shizuoka, Japan). The tumorigenicity assay was conducted as described previously [30]. Briefly, tumor-derived organoids were collected at 70–80% confluency, and an aliquot of 1/10 the volume was completely dissociated using Accumax for cell counting with the Countess FL Automated Cell Counter. For each organoid, 1 × 106 cells were resuspended in 100 µl of advanced DMEM/F12, mixed with 100 µl of Matrigel at 1:1 ratio and thereafter injected subcutaneously (sc) into the dorsal skin of the nude mice. Tumor development was monitored for 4 weeks.

RNA sequencing and data analysis

2D-culture cells were cultured in appropriate media and harvested by trypsinization. 3D-culture cells were cultured in organoid culture conditions described above. Total RNA was isolated using the AurumTM Total RNA Mini Kit (BioRad) according to the manufacturer’s protocol. RNA quality was assessed using a 2100 BioAnalyzer (Agilent). In total, 100 ng of RNA was used to prepare sequencing libraries, using NEBNext Ultra II Directional RNA Library Prep with Sample Purification Beads (NEB), according to the manufacturer’s instructions. Libraries were validated using the BioAnalyzer and quantified by qPCR and Qubit Fluorometric Quantitation (Thermo Fischer). Novaseq 6000 (Illumina), which generates 150-bp pair-end reads, was used. Data analysis of RNA-seq was performed as previously described with some modifications [9, 32]. Quality of raw sequence data was assessed using FastQC (Version: FastQC 0.10.0). FASTQ files were trimmed for adaptors and Phred score (> 20) using TrimGalore! (0.6.4). Trimmed sequenced reads were aligned to the mouse genome assembly (GRCm39 Ensembl release 103) using STAR (2.7.9a). Aligned reads were used to quantify mRNA with HTSeq-count (version 0.12.4). Differential gene expression analysis of paired-comparison between 2D-culture and 3D-culture was performed using edgeR package on protein-coding genes. Genes were considered as differentially expressed when the FDR-adjusted P value was below 0.05. Gene ontology analysis was performed using the online bioinformatic tool, Database for Annotation, Visualization and Integrated Discovery (DAVID, v6.8) and Enrichr (https://​maayanlab.​cloud/​Enrichr/​). Heat maps were generated using either R package pheatmap or heatmap3.

Immunoblot analysis

Protein extraction and sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) were performed according to the previously established protocol [33]. Cell line samples were lysed using the RIPA buffer (150 mM NaCl, 50 mM Tris–HCl (pH 7.4), 1% Nonidet P-40, 1% SDS, 0.5% deoxycholate), containing PierceTM Protease and Phosphatase inhibitor (Thermo Scientific). Subsequently, they were homogenized by super-sonication (UD-100, TOMY, Tokyo, Japan) and clarified by centrifugation at 18,000 × g for 30 min at 4 °C. The total protein concentration of the lysates was quantified using the Protein Assay Bicinchoninate kit (Nacalai tesque, Kyoto, Japan).

Statistical analyses

Statistical analyses were performed using the Graphpad Prism software (version 8.4.3) and the R statistical environment (http://​r-project.​org). All data are presented as means ± SEM, unless indicated otherwise. Data were tested for normal distribution with unpaired two-tailed Student’s t-test (if two samples have equal variances) or Welch's t-test (if two samples have unequal variances) to determine statistical significance between the two groups. Statistical significance among more than two groups were analyzed using one-way ANOVA along with Tukey's multiple comparison test to assess statistical significance with a 95% confidence interval. P < 0.05 was considered statistically significant, unless otherwise specified. All the experiments were independently repeated at least three times, unless otherwise mentioned.

Results

Establishment of organoids from mouse MM

MM organoids were generated using mouse MM models by intraperitoneally injecting mice with either asbestos (crocidolite) [2527] or carbon nanotubes (NT-50) [29] as described previously. MM incidence was 42.9% in crocidolite-treated p53+/- mice (Table 2), which was similar to the result of a previous report [34]. MM was assessed to be the main cause of death in the crocidolite-treated mice whereas ileus caused severe peritonitis with adhesion, primarily leading to death, in the NT-50-treated mice (Fig. 1a–c). Attempts to establish MM organoids from the MM tissue or ascites resulted in an establishment and successful propagation of organoids in 7 of the 11 MM cases (Table 2 and Fig. 1f, g). To test whether the organoids have a self-renewal potential, we performed single-cell seeding assay, where single-cell suspensions could generate MM organoids (Fig. 1d, e). The MM organoids revealed two distinct patterns. Most organoids formed a spherical pattern whereas only one MM-organoid (m12) formed a budding pattern (Fig. 1d–g). MM-organoids derived from MM tissue were confirmed to propagate at least for 6 months, tolerating freeze and thaw (data not shown). These results indicated that mouse MM-derived organoids can function as a useful resource for research on MM (Table 3).
Table 2
MM incidence and organoid lines derived from p53+/- and wild-type C57BL6 mouse
Mouse genotype
Material
Total number
Mesothelioma incidence
Established organoids
Number
%
Number
%
P53 +/-
Crocidolite
14
6
42.9
5
83.3
NT50
10(3)
3
42.9
1
33.3
NT
8
0
0.0
0
ND
P53 + / +
Crocidolite
10(1)
1
11.1
1
100.0
NT50
4(2)
1
50.0
0
0
NT
7
0
0.0
0
ND
The total number includes mice died from ileus resulting from peritonitis in parentheses. Mesothelioma incidence: mesothelioma/[total number-ileus] (%). Established organoid: the number of established organoids/number or mesothelioma incidence (%). NT Carbon nanotube, NT No treatment, ND Not determined. Refer to text for details
Table 3
Histological summary of mice with exposed materials and the days of tumor development
Organoid ID
Material
mouse genotype
Mesothelioma incidence
Histology
Days
Subtype
MSLN
WT1
m5
Crocidolite
P53 +/-
211
Biphasic
 + 
 + 
m12
Crocidolite
P53 +/-
156
Biphasic
-
-
m28
Crocidolite
P53 +/-
221
Sarcomatoid
 + 
 + 
m54
Crocidolite
P53 +/-
173
Sarcomatoid
 + 
-
m60
Crocidolite
P53 +/-
194
Biphasic
-
-
m67
Crocidolite
P53 + / +
139
ND(Ascites)
ND(Ascites)
ND(Ascites)
m107
NT50
P53 +/-
174
Sarcomatoid
 + 
 + 
NT Carbon nanotube, ND Not determined. Refer to text for details

Epithelial growth factors regulate MM stemness and proliferation in organoids

In order to test the dependence of MM-derived mesothelial organoids on the crucial growth factors supplemented in the organoid growth medium, organoids were dissociated to single cells and resuspended in organoid media either with or without the crucial growth factors. EGF, Noggin and R-spondin1 are widely accepted as most essential factors in the organoid medium. We also tested additional factors such as Wnt3a, FGF and hydrocortisone (HC) [3538]. Compared to the m5 organoid line (Table 3), the m12 organoid line grew faster, independent of all the growth factors except epidermal growth factor (EGF) (Fig. 2a, b, S2a, b). In contrast, the m5 organoid line required both EGF and R-spondin1 (Fig. 2a, b). EGF-receptor (EGFR) is overexpressed in various epithelial malignancies and in pleural MM [39, 40]. In order to study the differences between these newly established MM cell lines and normal mesothelial cells, we performed immunoblotting analysis on the MM-associated oncoproteins and the tumor suppressor proteins [41]. Expression of β-catenin and insulin growth factor receptor 1 (IGFR) was not altered in mouse MM cell lines. However, EGFR was expressed in some MM cell lines (Fig. 2c). Furthermore, p53 tumor suppressor protein was at the undetectable level in five MM cell lines (Fig. 2d). These results suggest that most of the newly derived MM cells depends on EGFR pathway activation and/or loss of p53 and MM cells required the EGF to grow as organoid.

Implants of MM-organoids maintain characteristics of primary MM

We then evaluated the capacities of mouse MM organoids to retain mesothelial characteristics. Secretion of hyaluronic acid (HA) is one of the characteristics of MM [42]. The retention of HA secretion by MM organoid was confirmed by detection with acidic mucin staining or HA binding protein in a similar fashion between the mouse MM-derived organoids and MM tissues (Fig. S3). In order to gain further insight into the MM organoid culturing system, the MM-organoids and 2D MM cell lines were simultaneously injected into the bilateral dorsal skin of immunodeficient mice (Fig. 3a). We used m5 and m107 cell line because these two lines are typical mouse MM by the expression of WT1 and mesothelin (MSLN) (Table 3) and EGFR or P53 expression status were completely opposite in the two cell lines (Fig. 2c, d). At 36 days after the sc injection, histology of both the implants revealed a similar solid growth pattern (Fig. 3b). However, proliferation of the MM organoids was significantly faster than that of the 2D MM lines (Fig. 3a, c). Immunohistochemical analyses of the implants revealed the expression of WT1 and MSLN, confirming the maintenance of the original characteristics (Fig. 3d). These results suggest that MM organoids can capture the in vivo characteristics of MM developed by crocidolite or CNT-50 injection in mice.

RNA sequencing analysis revealed expressional alteration between MM organoids and 2D-culture

We further investigated the differences between MM organoids and 2D MM cell lines with RNA sequencing analysis. MM organoids and 2D MM cell lines revealed genes in distinct clusters (Fig. 4a). Gene Ontology analysis for biological processes and cellular components linking the gene expressional changes between MM organoids (3D) cells and 2D culture-associated genes indicated that molecular function and Protein Protein-Interaction (PPI) Hub Proteins were associated with the receptor tyrosine kinases, such as IGFR and EGFR (Fig. 4b, c). These results suggest that the different culture systems strongly alter gene expression in MM.

Sensitivity of MM organoids to cisplatin depends on CTR1

In order to assess the usefulness of MM organoids as a pre-clinical model to evaluate the sensitivity to chemotherapeutic agents, we performed cell viability assays after cisplatin treatment (Fig. 5a-c). MM organoids were significantly more sensitive to cisplatin treatment than the corresponding 2D MM cell lines (Fig. 5b, c, S2c, d). A SLC31 family copper transporter, CTR1, is a major cisplatin transporter for uptake and its suppression contributes to chemoresistance [17]. However, Slc31a1 (Ctr1) mRNA expression was not significantly different between MM organoids and 2D MM cell lines based on RNA-sequence analysis (Fig. 5d) and the cellular localization of CTR1 was not significantly different as well (Fig. 5e). We found, however, a significant difference in the band number and band-shift on the immunoblot of CTR1 between MM organoids and MM 2D cultures, suggesting difference in glycosylation or possibly lipidation, which revealed a similar pattern between m5 and m107 (Fig. 5f). Our pathway analysis showed that protein glycosylation was strongly affected between MM organoids and 2D MM cell lines (Fig. 5g, h). These results suggest that posttranslational modification of CTR1 protein but not expressional level regulates cisplatin sensitivity.

Glycosylation and extracellular stiffness control the CTR1 localization

One major differences in organoid and 2D-culture is stiffness, which may affect cell metabolism [43]. Protein glycosylation and lipidation regulate its cellular localization [44, 45]. We thus studied the CTR1 localization between MM-organoid and 2D cultures. For this purpose, we established the murine MM cells, stably expressing flag-mCTR1 (Fig. 6a) and used the two different culture systems, high-stiffness culture with plastic bottom and low stiffness culture with Matrigel layer (Fig. 6b). MβCD, a known endocytosis inhibitor, eventually induces membrane localization of CTR1 [46]. After 24 h incubation, CTR1 expression decreased in the low stiffness culture but the MβCD induced no alteration (Fig. 6c). Actin filament (F-actin) is known as a protein localizing to plasma membrane, thus we used F-actin as a marker of plasma membrane (Fig. 6d). Surprisingly, CTR1 localized in the cytosol with 2D MM cell lines whereas 2D MM cell lines plated on Matrigel exhibited CTR1 localization on the plasma membrane in a similar fashion to MβCD application (Fig. 6d), suggesting that extracellular stiffness regulates CTR1 localization. Then, we used the tunicamycin (TM) as a N-glycosylation inhibitor and 2-bromopalmitate (BP) as a lipidation inhibitor. We found that TM mediated the band shift of CTR1 from 30–50 kDa to 25 kDa in MM but not with BP (Fig. 6e, f, S4a-c). Further, as we expected, TM also inhibited the CTR1-plasma membrane localization (Fig. 6g, S4d) and the mutant of CTR1-N-glycosylation site (N23) localized to Golgi and the mutation also inhibited the band shift by glycosylation similar to TM treatment (Fig. S5a-c). These results suggested that glycosylation regulates the sub-cellular localization of CTR-1.

Discussion

In the present study, we established a protocol for the Matrigel-based mouse MM-organoid generation as 3D culture. There are a few preceding studies on human MM, such as detection of MM stem cell markers [47, 48], recognition of transglutaminase as MM stem cell survival protein [49] and 2 human MM case studies of organoid formation by the use of photopolymerizable hyaluronic acid and gelatin hydrogen precursor to screen for appropriate chemotherapeutic drugs [50]. However, due to the rarity of human MM, it is not easy to confirm the reproducibility. Here we for the first time established the MM-organoid, using Matrigel and complete organoid medium containing basic stem cell factors (EGF, Noggin, R-spondin-1) but without FBS [51, 52]. This strategy exhibits various advantages, including EMT inhibition (Fig. S2e), preservation of CSCs and maintenance of polarity and stiffness similar to those of the actual human tumor microenvironment [12, 13]. It has been indeed established that 3D culture systems, especially CSC culture, simulate physiological conditions. Thus, our new findings could be applied to the high throughput drug screening and organoids-on-a-chip technology for targeting therapy of MM-CSC [13].
This study indeed showed an important finding that CTR1, a major cisplatin importer, localizes to the plasma membrane both in the Matrigel culture system and the implanted tumor but not in the 2D system (Figs. 5e and 6d and S2f). MβCD, a known endocytosis inhibitor, could eventually correct and induce membrane localization of CTR1. Cellular localization of CTR1 as a Cu(II) transporter dynamically changes in enterocytes where CTR1 is highly expressed [46]. Organoid culture study on CTR1 localization in intestinal cells clarified an unknown link between intestinal Cu metabolism and dietary fat processing [53]. Alterations in protein-localization are regulated at least partially by protein modifications [44, 45]. Our data also showed that plasma membrane localization of CTR1 regulates cisplatin sensitivity (Figs. 5g-h and 6e-g). Recently, it was reported that BAP1 loss in MM may modulate survival after cisplatin and/or pemetrexed treatment [54, 55]. Our RNA sequencing data showed no differences in Bap1 gene expression between 2D culture and organoid (data not shown) but how the BAP1 plays a role in organoid culture is still unknown. Our organoid model may contribute to revealing the association between BAP1 status and cisplatin sensitivity through CTR1 localization in the near future. These findings may incite the studies not only on cisplatin-drug sensitivity through CTR1 but also on other plasma membrane proteins associated with cancer drug metabolism, including immunotherapy. In 2020, FDA approved drug combination therapy using nivolumab which can decrease tumor growth by enhancing cytotoxic T-cell function (https://​www.​fda.​gov/​news-events/​press-announcements/​fda-approves-drugcombination-treating-mesothelioma). For tumor cell killing, these immune cells recognize the specific ligand of immune checkpoint receptor in cancer cells [56]. These stromal cells also exist in MM tissues such as myofibroblast and macrophages (Fig. S6a). However, our organoid culture could not retain the CD68-positive macrophage population (Fig. S6b) and possibly other immune cells in the MM-organoid. A recent study showed a possibility for organoid co-culture with the immune cells to test for immunotherapy [57, 58]. For preclinical test in immunotherapy, further studies are necessary. RNA-sequence analysis revealed that the immune cells also affect to mesothelioma cells especially cytokine stimulation (Fig. S6c-d). Organoid culture, however, holds merits to quantify the ligand protein such as PD-L1, CD80, MHC and Ceacam1 because these ligand proteins are delivered to cell surface through the glycosylation, which can be demonstrated in the organoid culture. Therefore, we believe that Matrigel-based cultures would change the paradigm and improve our knowledge of cell behavior in MM.
Our RNA-sequencing data was compared in two different conditions between 2D culture in medium with FBS and antibiotics and 3D organoid under complete medium. Therefore, there is a limitation on the gene expression of our study that we cannot completely rule out the possibility that the differences might be influenced by the culture media and additives. Various culture strategies have been undertaken to establish MM cell lines [23, 24, 59]. However, MM cell lines have generally presented a problem of changing morphology into spindle-shaped via EMT [60, 61]. According with each passage, the MM cells became more spindle-shaped (data not shown). The organoid culture system could solve this issue because this system uses no FBS, thus inhibiting the EMT (Fig. S2e, f). The present protocol may be applied to the organoid culture of non-tumorous mesothelial cells as well, where the mesothelial cells can be immortalized using SV40 and small T antigen. Therefore, the formation of mesothelial organoids would be eventually possible. We first attempted to establish mesothelial organoids from mesentery (Fig. S1a, b), but the mesothelial cells without immortalization in the Matrigel failed to proliferate with essential organoid medium, resulting in a failure in passage. Further studies are in progress to focus on the ingredient requirements of the mesothelial stem-like cells and the embryonic origin of the mesothelial stem-like cells, which can be ground-breaking in this research area.
There are several reports on the MM models in mice. In this study, we used the p53+/- mice. CDKN2A encodes p16INK4A and p14ARF (p19ARF in mice), two tumor suppressors that regulate the Rb and p53 pathways, respectively. P14ARF is a component of the p53 pathway and p53 mutation has also been reported in MM [62, 63]. In this p53+/- mouse, crocidolite injection produced MM. However, carcinogenic carbon nanotube NT-50 sometimes caused ileus due to peritonitis in addition to MM (Fig. 1b and Table 3). We used 1.5 mg NT-50 for ip injection, which might have been an excess dose. Previous reports on NT-50 showed that MM incidence is dose-dependent and less than 0.3 mg ip injection would be successful to cause MM incidence without causing peritonitis and ileus [64]. We further found that some of MM from the mouse model expressed EGFR. Three MM-cell lines from the present study were TP53-deficient and revealed EGFR expression whereas two MM-cell lines had no change in TP53 and expressed EGFR (Fig. 2d). On the other hand, our RNA-sequencing analysis showed that the tendency of Egfr, Ctnnb1(beta-catenin) and Igf1r expression was not different between organoid culture and the original tissue MM but rather intensified the tendency (Fig. S6f-h). This means that some genes are influenced on transcriptional regulation with organoid culture but others are not. EGF was also essential for MM-organoids (Fig. 2a, b) and our RNA-sequencing analysis revealed that organoid culture enriched receptor ligand activity and growth factor pathways (Fig. 4b, c). These results indicate that the organoid culture is adaptive for this tyrosine receptor kinase, including EGFR in MM.
In conclusion, we have established a novel protocol for mouse MM-organoid generation using Matrigel and the MM-organoid lines based thereon. The culture protocol would contribute to the future MM studies in mice, where EGF is required for the maintenance of MM stem-like cells in mouse MM-organoid. Finally, we believe that MM organoids can provide a pre-clinical model of humans for drug discovery and translational research in the near future.

Acknowledgements

The authors thank Nobuaki Misawa (Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine) for excellent technical assistance, and also Division for Medical Research Engineering, Nagoya University Graduate School of Medicine for technical assistance.

Declarations

All the animal experiments were approved by the animal experimental and ethics committee of Nagoya University Graduate School of Medicine (M220346-004, 2022/03/18). We confirm that all methods were carried out in accordance with relevant guidelines and regulations, and we also confirm that all methods are reported in accordance with ARRIVE guidelines (https://​arriveguidelines​.​org) for the reporting of animal experiments. This study does not include patient participation or analysis of patient data.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Steele NG, Chakrabarti J, Wang J, Biesiada J, Holokai L, Chang J, Nowacki LM, Hawkins J, Mahe M, Sundaram N, et al. An Organoid-Based Preclinical Model of Human Gastric Cancer. Cell Mol Gastroenterol Hepatol. 2019;7(1):161–84.PubMedCrossRef Steele NG, Chakrabarti J, Wang J, Biesiada J, Holokai L, Chang J, Nowacki LM, Hawkins J, Mahe M, Sundaram N, et al. An Organoid-Based Preclinical Model of Human Gastric Cancer. Cell Mol Gastroenterol Hepatol. 2019;7(1):161–84.PubMedCrossRef
2.
Zurück zum Zitat Almagro J, Messal HA, Zaw Thin M, van Rheenen J, Behrens A. Tissue clearing to examine tumour complexity in three dimensions. Nat Rev Cancer. 2021;21(11):718–30.PubMedCrossRef Almagro J, Messal HA, Zaw Thin M, van Rheenen J, Behrens A. Tissue clearing to examine tumour complexity in three dimensions. Nat Rev Cancer. 2021;21(11):718–30.PubMedCrossRef
3.
Zurück zum Zitat Hofer M, Lutolf MP. Engineering organoids. Nat Rev Mater. 2021;6(5):402–20. Hofer M, Lutolf MP. Engineering organoids. Nat Rev Mater. 2021;6(5):402–20.
6.
Zurück zum Zitat IARC, WHO: Asbestos (chrysotile, amosite, crocidolite, tremolite, actinolite, and anthophyllite). In: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans A Review of Human Carcinogens; Part C: Arsenic, Metals, Fibres, and Dusts. Volume 100C. Lyon; 2012. p. 219–309. IARC, WHO: Asbestos (chrysotile, amosite, crocidolite, tremolite, actinolite, and anthophyllite). In: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans A Review of Human Carcinogens; Part C: Arsenic, Metals, Fibres, and Dusts. Volume 100C. Lyon; 2012. p. 219–309.
7.
Zurück zum Zitat Toyokuni S. Iron addiction with ferroptosis-resistance in asbestos-induced mesothelial carcinogenesis: Toward the era of mesothelioma prevention. Free Radic Biol Med. 2019;133:206–15.PubMedCrossRef Toyokuni S. Iron addiction with ferroptosis-resistance in asbestos-induced mesothelial carcinogenesis: Toward the era of mesothelioma prevention. Free Radic Biol Med. 2019;133:206–15.PubMedCrossRef
8.
Zurück zum Zitat Ito F, Yanatori I, Maeda Y, Nimura K, Ito S, Hirayama T, Nagasawa H, Kohyama N, Okazaki Y, Akatsuka S, et al. Asbestos conceives Fe(II)-dependent mutagenic stromal milieu through ceaseless macrophage ferroptosis and β-catenin induction in mesothelium. Redox Biol. 2020;36:101616. Ito F, Yanatori I, Maeda Y, Nimura K, Ito S, Hirayama T, Nagasawa H, Kohyama N, Okazaki Y, Akatsuka S, et al. Asbestos conceives Fe(II)-dependent mutagenic stromal milieu through ceaseless macrophage ferroptosis and β-catenin induction in mesothelium. Redox Biol. 2020;36:101616.
9.
Zurück zum Zitat Ito F, Kato K, Yanatori I, Murohara T, Toyokuni S. Ferroptosis-dependent extracellular vesicles from macrophage contribute to asbestos-induced mesothelial carcinogenesis through loading ferritin. Redox Biol. 2021;47: 102174.PubMedPubMedCentralCrossRef Ito F, Kato K, Yanatori I, Murohara T, Toyokuni S. Ferroptosis-dependent extracellular vesicles from macrophage contribute to asbestos-induced mesothelial carcinogenesis through loading ferritin. Redox Biol. 2021;47: 102174.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Evans S, Butler JR, Mattila JT, Kirschner DE. Systems biology predicts that fibrosis in tuberculous granulomas may arise through macrophage-to-myofibroblast transformation. PLoS Comput Biol. 2020;16(12): e1008520.PubMedPubMedCentralCrossRef Evans S, Butler JR, Mattila JT, Kirschner DE. Systems biology predicts that fibrosis in tuberculous granulomas may arise through macrophage-to-myofibroblast transformation. PLoS Comput Biol. 2020;16(12): e1008520.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891–906.PubMedPubMedCentralCrossRef Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891–906.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Funfak A, Bouzhir L, Gontran E, Minier N, Dupuis-Williams P, Gobaa S. Biophysical Control of Bile Duct Epithelial Morphogenesis in Natural and Synthetic Scaffolds. Front Bioeng Biotechnol. 2019;7:417.PubMedPubMedCentralCrossRef Funfak A, Bouzhir L, Gontran E, Minier N, Dupuis-Williams P, Gobaa S. Biophysical Control of Bile Duct Epithelial Morphogenesis in Natural and Synthetic Scaffolds. Front Bioeng Biotechnol. 2019;7:417.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Lv H, Li L, Sun M, Zhang Y, Chen L, Rong Y, Li Y. Mechanism of regulation of stem cell differentiation by matrix stiffness. Stem Cell Res Ther. 2015;6:103.PubMedPubMedCentralCrossRef Lv H, Li L, Sun M, Zhang Y, Chen L, Rong Y, Li Y. Mechanism of regulation of stem cell differentiation by matrix stiffness. Stem Cell Res Ther. 2015;6:103.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Nassar D, Blanpain C. Cancer Stem Cells: Basic Concepts and Therapeutic Implications. Annu Rev Pathol. 2016;11:47–76.PubMedCrossRef Nassar D, Blanpain C. Cancer Stem Cells: Basic Concepts and Therapeutic Implications. Annu Rev Pathol. 2016;11:47–76.PubMedCrossRef
15.
16.
17.
Zurück zum Zitat Pabla N, Murphy RF, Liu K, Dong Z. The copper transporter Ctr1 contributes to cisplatin uptake by renal tubular cells during cisplatin nephrotoxicity. Am J Physiol Renal Physiol. 2009;296(3):F505-511.PubMedPubMedCentralCrossRef Pabla N, Murphy RF, Liu K, Dong Z. The copper transporter Ctr1 contributes to cisplatin uptake by renal tubular cells during cisplatin nephrotoxicity. Am J Physiol Renal Physiol. 2009;296(3):F505-511.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Endo T, Kimura O, Sakata M. Carrier-mediated uptake of cisplatin by the OK renal epithelial cell line. Toxicology. 2000;146(2–3):187–95.PubMedCrossRef Endo T, Kimura O, Sakata M. Carrier-mediated uptake of cisplatin by the OK renal epithelial cell line. Toxicology. 2000;146(2–3):187–95.PubMedCrossRef
19.
Zurück zum Zitat Kolb RJ, Ghazi AM, Barfuss DW. Inhibition of basolateral transport and cellular accumulation of cDDP and N-acetyl- L-cysteine-cDDP by TEA and PAH in the renal proximal tubule. Cancer Chemother Pharmacol. 2003;51(2):132–8.PubMedCrossRef Kolb RJ, Ghazi AM, Barfuss DW. Inhibition of basolateral transport and cellular accumulation of cDDP and N-acetyl- L-cysteine-cDDP by TEA and PAH in the renal proximal tubule. Cancer Chemother Pharmacol. 2003;51(2):132–8.PubMedCrossRef
20.
Zurück zum Zitat Ludwig T, Riethmuller C, Gekle M, Schwerdt G, Oberleithner H. Nephrotoxicity of platinum complexes is related to basolateral organic cation transport. Kidney Int. 2004;66(1):196–202.PubMedCrossRef Ludwig T, Riethmuller C, Gekle M, Schwerdt G, Oberleithner H. Nephrotoxicity of platinum complexes is related to basolateral organic cation transport. Kidney Int. 2004;66(1):196–202.PubMedCrossRef
21.
Zurück zum Zitat Eisses JF, Kaplan JH. Molecular characterization of hCTR1, the human copper uptake protein. J Biol Chem. 2002;277(32):29162–71.PubMedCrossRef Eisses JF, Kaplan JH. Molecular characterization of hCTR1, the human copper uptake protein. J Biol Chem. 2002;277(32):29162–71.PubMedCrossRef
22.
Zurück zum Zitat Dauleh S, Santeramo I, Fielding C, Ward K, Herrmann A, Murray P, Wilm B. Characterisation of Cultured Mesothelial Cells Derived from the Murine Adult Omentum. PLoS ONE. 2016;11(7): e0158997.PubMedPubMedCentralCrossRef Dauleh S, Santeramo I, Fielding C, Ward K, Herrmann A, Murray P, Wilm B. Characterisation of Cultured Mesothelial Cells Derived from the Murine Adult Omentum. PLoS ONE. 2016;11(7): e0158997.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Kawai N, Ouji Y, Sakagami M, Tojo T, Sawabata N, Yoshikawa M, Taniguchi S. Isolation and culture of pleural mesothelial cells. Exp Lung Res. 2019;45(5–6):151–6.PubMedCrossRef Kawai N, Ouji Y, Sakagami M, Tojo T, Sawabata N, Yoshikawa M, Taniguchi S. Isolation and culture of pleural mesothelial cells. Exp Lung Res. 2019;45(5–6):151–6.PubMedCrossRef
25.
Zurück zum Zitat Jiang L, Akatsuka S, Nagai H, Chew SH, Ohara H, Okazaki Y, Yamashita Y, Yoshikawa Y, Yasui H, Ikuta K, et al. Iron overload signature in chrysotile-induced malignant mesothelioma. J Pathol. 2012;228:366–77.PubMedCrossRef Jiang L, Akatsuka S, Nagai H, Chew SH, Ohara H, Okazaki Y, Yamashita Y, Yoshikawa Y, Yasui H, Ikuta K, et al. Iron overload signature in chrysotile-induced malignant mesothelioma. J Pathol. 2012;228:366–77.PubMedCrossRef
26.
Zurück zum Zitat Funahashi S, Okazaki Y, Akatsuka S, Takahashi T, Sakumi K, Nakabeppu Y, Toyokuni S. Mth1 deficiency provides longer survival upon intraperitoneal crocidolite injection in female mice. Free Radic Res. 2020;54(2–3):195–205.PubMedCrossRef Funahashi S, Okazaki Y, Akatsuka S, Takahashi T, Sakumi K, Nakabeppu Y, Toyokuni S. Mth1 deficiency provides longer survival upon intraperitoneal crocidolite injection in female mice. Free Radic Res. 2020;54(2–3):195–205.PubMedCrossRef
27.
Zurück zum Zitat Okazaki Y, Misawa N, Akatsuka S, Kohyama N, Sekido Y, Takahashi T, Toyokuni S. Frequent homozygous deletion of Cdkn2a/2b in tremolite-induced malignant mesothelioma in rats. Cancer Sci. 2020;111(4):1180–92.PubMedPubMedCentralCrossRef Okazaki Y, Misawa N, Akatsuka S, Kohyama N, Sekido Y, Takahashi T, Toyokuni S. Frequent homozygous deletion of Cdkn2a/2b in tremolite-induced malignant mesothelioma in rats. Cancer Sci. 2020;111(4):1180–92.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Tsukada T, Tomooka Y, Takai S, Ueda Y, Nishikawa S, Yagi T, Tokunaga T, Takeda N, Suda Y, Abe S, et al. Enhanced proliferative potential in culture of cells from p53-deficient mice. Oncogene. 1993;8(12):3313–22.PubMed Tsukada T, Tomooka Y, Takai S, Ueda Y, Nishikawa S, Yagi T, Tokunaga T, Takeda N, Suda Y, Abe S, et al. Enhanced proliferative potential in culture of cells from p53-deficient mice. Oncogene. 1993;8(12):3313–22.PubMed
29.
Zurück zum Zitat Nagai H, Okazaki Y, Chew S, Misawa N, Yamashita Y, Akatsuka S, Yamashita K, Ishihara T, Yoshikawa Y, Jiang L, et al. Diameter of multi-walled carbon nanotubes is a critical factor in mesothelial injury and subsequent carcinogenesis. Proc Natl Acad Sci U S A. 2011;108(49):E1330-1338.PubMedPubMedCentralCrossRef Nagai H, Okazaki Y, Chew S, Misawa N, Yamashita Y, Akatsuka S, Yamashita K, Ishihara T, Yoshikawa Y, Jiang L, et al. Diameter of multi-walled carbon nanotubes is a critical factor in mesothelial injury and subsequent carcinogenesis. Proc Natl Acad Sci U S A. 2011;108(49):E1330-1338.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Maru Y, Tanaka N, Ebisawa K, Odaka A, Sugiyama T, Itami M, Hippo Y. Establishment and characterization of patient-derived organoids from a young patient with cervical clear cell carcinoma. Cancer Sci. 2019;110(9):2992–3005.PubMedPubMedCentralCrossRef Maru Y, Tanaka N, Ebisawa K, Odaka A, Sugiyama T, Itami M, Hippo Y. Establishment and characterization of patient-derived organoids from a young patient with cervical clear cell carcinoma. Cancer Sci. 2019;110(9):2992–3005.PubMedPubMedCentralCrossRef
31.
32.
Zurück zum Zitat Kato K, Dieguez-Hurtado R, Park DY, Hong SP, Kato-Azuma S, Adams S, Stehling M, Trappmann B, Wrana JL, Koh GY, et al. Pulmonary pericytes regulate lung morphogenesis. Nat Commun. 2018;9(1):2448.PubMedPubMedCentralCrossRef Kato K, Dieguez-Hurtado R, Park DY, Hong SP, Kato-Azuma S, Adams S, Stehling M, Trappmann B, Wrana JL, Koh GY, et al. Pulmonary pericytes regulate lung morphogenesis. Nat Commun. 2018;9(1):2448.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Yanatori I, Richardson DR, Dhekne HS, Toyokuni S, Kishi F. CD63 is regulated by iron via the IRE-IRP system and is important for ferritin secretion by extracellular vesicles. Blood. 2021;138(16):1490–503.PubMedPubMedCentralCrossRef Yanatori I, Richardson DR, Dhekne HS, Toyokuni S, Kishi F. CD63 is regulated by iron via the IRE-IRP system and is important for ferritin secretion by extracellular vesicles. Blood. 2021;138(16):1490–503.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Takagi A, Hirose A, Nishimura T, Fukumori N, Ogata A, Ohashi N, Kitajima S, Kanno J. Induction of mesothelioma in p53+/- mouse by intraperitoneal application of multi-wall carbon nanotube. J Toxicol Sci. 2008;33(1):105–16.PubMedCrossRef Takagi A, Hirose A, Nishimura T, Fukumori N, Ogata A, Ohashi N, Kitajima S, Kanno J. Induction of mesothelioma in p53+/- mouse by intraperitoneal application of multi-wall carbon nanotube. J Toxicol Sci. 2008;33(1):105–16.PubMedCrossRef
35.
Zurück zum Zitat Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72.
36.
Zurück zum Zitat Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, Ohta Y, Toshimitsu K, Nakazato Y, Kawasaki K, et al. A Colorectal Tumor Organoid Library Demonstrates Progressive Loss of Niche Factor Requirements during Tumorigenesis. Cell Stem Cell. 2016;18(6):827–38.PubMedCrossRef Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, Ohta Y, Toshimitsu K, Nakazato Y, Kawasaki K, et al. A Colorectal Tumor Organoid Library Demonstrates Progressive Loss of Niche Factor Requirements during Tumorigenesis. Cell Stem Cell. 2016;18(6):827–38.PubMedCrossRef
37.
Zurück zum Zitat Date S, Sato T. Mini-gut organoids: reconstitution of the stem cell niche. Annu Rev Cell Dev Biol. 2015;31:269–89.PubMedCrossRef Date S, Sato T. Mini-gut organoids: reconstitution of the stem cell niche. Annu Rev Cell Dev Biol. 2015;31:269–89.PubMedCrossRef
38.
Zurück zum Zitat Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 2018; 172(1–2):373–386 e310. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 2018; 172(1–2):373–386 e310.
39.
Zurück zum Zitat Destro A, Ceresoli GL, Falleni M, Zucali PA, Morenghi E, Bianchi P, Pellegrini C, Cordani N, Vaira V, Alloisio M, et al. EGFR overexpression in malignant pleural mesothelioma. An immunohistochemical and molecular study with clinico-pathological correlations. Lung Cancer. 2006;51(2):207–15. Destro A, Ceresoli GL, Falleni M, Zucali PA, Morenghi E, Bianchi P, Pellegrini C, Cordani N, Vaira V, Alloisio M, et al. EGFR overexpression in malignant pleural mesothelioma. An immunohistochemical and molecular study with clinico-pathological correlations. Lung Cancer. 2006;51(2):207–15.
40.
Zurück zum Zitat Brevet M, Shimizu S, Bott MJ, Shukla N, Zhou Q, Olshen AB, Rusch V, Ladanyi M. Coactivation of receptor tyrosine kinases in malignant mesothelioma as a rationale for combination targeted therapy. J Thorac Oncol. 2011;6(5):864–74.PubMedCrossRef Brevet M, Shimizu S, Bott MJ, Shukla N, Zhou Q, Olshen AB, Rusch V, Ladanyi M. Coactivation of receptor tyrosine kinases in malignant mesothelioma as a rationale for combination targeted therapy. J Thorac Oncol. 2011;6(5):864–74.PubMedCrossRef
41.
Zurück zum Zitat Sekido Y. Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 2013;34(7):1413–9.PubMedCrossRef Sekido Y. Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 2013;34(7):1413–9.PubMedCrossRef
42.
Zurück zum Zitat Fujimoto N, Gemba K, Asano M, Fuchimoto Y, Wada S, Ono K, Ozaki S, Kishimoto T. Hyaluronic acid in the pleural fluid of patients with malignant pleural mesothelioma. Respir Investig. 2013;51(2):92–7.PubMedCrossRef Fujimoto N, Gemba K, Asano M, Fuchimoto Y, Wada S, Ono K, Ozaki S, Kishimoto T. Hyaluronic acid in the pleural fluid of patients with malignant pleural mesothelioma. Respir Investig. 2013;51(2):92–7.PubMedCrossRef
43.
44.
Zurück zum Zitat Chen B, Sun Y, Niu J, Jarugumilli GK, Wu X. Protein Lipidation in Cell Signaling and Diseases: Function, Regulation, and Therapeutic Opportunities. Cell Chem Biol. 2018;25(7):817–31.PubMedPubMedCentralCrossRef Chen B, Sun Y, Niu J, Jarugumilli GK, Wu X. Protein Lipidation in Cell Signaling and Diseases: Function, Regulation, and Therapeutic Opportunities. Cell Chem Biol. 2018;25(7):817–31.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Clifford RJ, Maryon EB, Kaplan JH. Dynamic internalization and recycling of a metal ion transporter: Cu homeostasis and CTR1, the human Cu+ uptake system. J Cell Sci. 2016;129(8):1711–21. Clifford RJ, Maryon EB, Kaplan JH. Dynamic internalization and recycling of a metal ion transporter: Cu homeostasis and CTR1, the human Cu+ uptake system. J Cell Sci. 2016;129(8):1711–21.
47.
Zurück zum Zitat Ghani FI, Yamazaki H, Iwata S, Okamoto T, Aoe K, Okabe K, Mimura Y, Fujimoto N, Kishimoto T, Yamada T, et al. Identification of cancer stem cell markers in human malignant mesothelioma cells. Biochem Biophys Res Commun. 2011;404(2):735–42.PubMedCrossRef Ghani FI, Yamazaki H, Iwata S, Okamoto T, Aoe K, Okabe K, Mimura Y, Fujimoto N, Kishimoto T, Yamada T, et al. Identification of cancer stem cell markers in human malignant mesothelioma cells. Biochem Biophys Res Commun. 2011;404(2):735–42.PubMedCrossRef
48.
Zurück zum Zitat Varghese S, Whipple R, Martin SS, Alexander HR. Multipotent cancer stem cells derived from human malignant peritoneal mesothelioma promote tumorigenesis. PLoS ONE. 2012;7(12): e52825.PubMedPubMedCentralCrossRef Varghese S, Whipple R, Martin SS, Alexander HR. Multipotent cancer stem cells derived from human malignant peritoneal mesothelioma promote tumorigenesis. PLoS ONE. 2012;7(12): e52825.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Adhikary G, Grun D, Alexander HR, Friedberg JS, Xu W, Keillor JW, Kandasamy S, Eckert RL. Transglutaminase is a mesothelioma cancer stem cell survival protein that is required for tumor formation. Oncotarget. 2018;9(77):34495–505.PubMedPubMedCentralCrossRef Adhikary G, Grun D, Alexander HR, Friedberg JS, Xu W, Keillor JW, Kandasamy S, Eckert RL. Transglutaminase is a mesothelioma cancer stem cell survival protein that is required for tumor formation. Oncotarget. 2018;9(77):34495–505.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Mazzocchi AR, Rajan SAP, Votanopoulos KI, Hall AR, Skardal A. In vitro patient-derived 3D mesothelioma tumor organoids facilitate patient-centric therapeutic screening. Sci Rep. 2018;8(1):2886.PubMedPubMedCentralCrossRef Mazzocchi AR, Rajan SAP, Votanopoulos KI, Hall AR, Skardal A. In vitro patient-derived 3D mesothelioma tumor organoids facilitate patient-centric therapeutic screening. Sci Rep. 2018;8(1):2886.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Lugli N, Kamileri I, Keogh A, Malinka T, Sarris ME, Talianidis I, Schaad O, Candinas D, Stroka D, Halazonetis TD. R-spondin 1 and noggin facilitate expansion of resident stem cells from non-damaged gallbladders. EMBO Rep. 2016;17(5):769–79.PubMedPubMedCentralCrossRef Lugli N, Kamileri I, Keogh A, Malinka T, Sarris ME, Talianidis I, Schaad O, Candinas D, Stroka D, Halazonetis TD. R-spondin 1 and noggin facilitate expansion of resident stem cells from non-damaged gallbladders. EMBO Rep. 2016;17(5):769–79.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Bigaeva E, Bomers JJM, Biel C, Mutsaers HAM, de Graaf IAM, Boersema M, Olinga P. Growth factors of stem cell niche extend the life-span of precision-cut intestinal slices in culture: A proof-of-concept study. Toxicol In Vitro. 2019;59:312–21.PubMedCrossRef Bigaeva E, Bomers JJM, Biel C, Mutsaers HAM, de Graaf IAM, Boersema M, Olinga P. Growth factors of stem cell niche extend the life-span of precision-cut intestinal slices in culture: A proof-of-concept study. Toxicol In Vitro. 2019;59:312–21.PubMedCrossRef
54.
Zurück zum Zitat Guazzelli A, Meysami P, Bakker E, Demonacos C, Giordano A, Krstic-Demonacos M, Mutti L. BAP1 Status Determines the Sensitivity of Malignant Mesothelioma Cells to Gemcitabine Treatment. Int J Mol Sci. 2019;20(2):429. Guazzelli A, Meysami P, Bakker E, Demonacos C, Giordano A, Krstic-Demonacos M, Mutti L. BAP1 Status Determines the Sensitivity of Malignant Mesothelioma Cells to Gemcitabine Treatment. Int J Mol Sci. 2019;20(2):429.
55.
Zurück zum Zitat Louw A, Panou V, Szejniuk WM, Meristoudis C, Chai SM, van Vliet C, Lee YCG, Dick IM, Firth T, Lynggaard LA, et al. BAP1 Loss by Immunohistochemistry Predicts Improved Survival to First-Line Platinum and Pemetrexed Chemotherapy for Patients With Pleural Mesothelioma: A Validation Study. J Thorac Oncol. 2022;17(7):921–30.PubMedCrossRef Louw A, Panou V, Szejniuk WM, Meristoudis C, Chai SM, van Vliet C, Lee YCG, Dick IM, Firth T, Lynggaard LA, et al. BAP1 Loss by Immunohistochemistry Predicts Improved Survival to First-Line Platinum and Pemetrexed Chemotherapy for Patients With Pleural Mesothelioma: A Validation Study. J Thorac Oncol. 2022;17(7):921–30.PubMedCrossRef
60.
Zurück zum Zitat Kajiyama H, Shibata K, Terauchi M, Ino K, Nawa A, Kikkawa F. Involvement of SDF-1alpha/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int J Cancer. 2008;122(1):91–9.PubMedCrossRef Kajiyama H, Shibata K, Terauchi M, Ino K, Nawa A, Kikkawa F. Involvement of SDF-1alpha/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int J Cancer. 2008;122(1):91–9.PubMedCrossRef
61.
Zurück zum Zitat Yoshihara M, Kajiyama H, Yokoi A, Sugiyama M, Koya Y, Yamakita Y, Liu W, Nakamura K, Moriyama Y, Yasui H, et al. Ovarian cancer-associated mesothelial cells induce acquired platinum-resistance in peritoneal metastasis via the FN1/Akt signaling pathway. Int J Cancer. 2020;146(8):2268–80.PubMedPubMedCentralCrossRef Yoshihara M, Kajiyama H, Yokoi A, Sugiyama M, Koya Y, Yamakita Y, Liu W, Nakamura K, Moriyama Y, Yasui H, et al. Ovarian cancer-associated mesothelial cells induce acquired platinum-resistance in peritoneal metastasis via the FN1/Akt signaling pathway. Int J Cancer. 2020;146(8):2268–80.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Cote RJ, Jhanwar SC, Novick S, Pellicer A. Genetic Alterations of the P53 Gene Are a Feature of Malignant Mesotheliomas. Can Res. 1991;51(19):5410–6. Cote RJ, Jhanwar SC, Novick S, Pellicer A. Genetic Alterations of the P53 Gene Are a Feature of Malignant Mesotheliomas. Can Res. 1991;51(19):5410–6.
63.
Zurück zum Zitat Altomare DA, Vaslet CA, Skele KL, De Rienzo A, Devarajan K, Jhanwar SC, McClatchey AI, Kane AB, Testa JR. A mouse model recapitulating molecular features of human mesothelioma. Cancer Res. 2005;65(18):8090–5.PubMedCrossRef Altomare DA, Vaslet CA, Skele KL, De Rienzo A, Devarajan K, Jhanwar SC, McClatchey AI, Kane AB, Testa JR. A mouse model recapitulating molecular features of human mesothelioma. Cancer Res. 2005;65(18):8090–5.PubMedCrossRef
64.
Zurück zum Zitat Takagi A, Hirose A, Futakuchi M, Tsuda H, Kanno J. Dose-dependent mesothelioma induction by intraperitoneal administration of multi-wall carbon nanotubes in p53 heterozygous mice. Cancer Sci. 2012;103(8):1440–4.PubMedCrossRef Takagi A, Hirose A, Futakuchi M, Tsuda H, Kanno J. Dose-dependent mesothelioma induction by intraperitoneal administration of multi-wall carbon nanotubes in p53 heterozygous mice. Cancer Sci. 2012;103(8):1440–4.PubMedCrossRef
Metadaten
Titel
Matrigel-based organoid culture of malignant mesothelioma reproduces cisplatin sensitivity through CTR1
verfasst von
Fumiya Ito
Katsuhiro Kato
Izumi Yanatori
Yuki Maeda
Toyoaki Murohara
Shinya Toyokuni
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10966-4

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.