Abstract
Background/Aim: Fucoxanthin (Fx), a dietary marine xanthophyll, exerts potent anticancer effects in various colorectal cancer (CRC) animal models. However, therapeutic effects of Fx in human cancer tissues remain unclear. A patient-derived xenograft (PDX) mouse model transplanted with cancer tissues from patients is widely accepted as the best preclinical model for evaluating the anticancer potential of drug candidates. Materials and Methods: Herein, we investigated the anticancer effects of Fx in PDX mice transplanted with cancer tissues derived from a patient with CRC (CRC-PDX) using LC-MS/MS- and western blot-based proteome analysis. Results: The tumor in the patient with CRC was a primary adenocarcinoma (T3N0M0, stage II) showing mutations of certain genes that were tumor protein p53 (TP53), AT-rich interaction domain 1A (ARID1A), neuroblastoma RAS viral oncogene homolog (NRAS), and PMS1 homolog 2 (PMS2). Administration of Fx significantly suppressed the tumor growth (0.6-fold) and tended to induce differentiation in CRC-PDX mice. Fx up-regulated glycanated-decorin (Gc-DCN) expression, and down-regulated Kinetochore-associated protein DSN1 homolog (DSN1), phospho(p) focal adhesion kinase (pFAK)(Tyr397), pPaxillin(Tyr31), and c-MYC involved in growth, adhesion, and/or cell cycle, in the tumors of CRC-PDX mice than in control mice. Alterations in the five proteins were consistent with those in human CRC HT-29 and HCT116 cells treated with fucoxanthinol (FxOH, a major metabolite of Fx). Conclusion: Fx suppresses development of human-like CRC tissues, especially through growth, adhesion, and cell cycle signals.
Fucoxanthin (Fx) (Figure 1) is a non-provitamin A carotenoid involved in photosynthesis and photoprotection in aquatic organisms (1). This natural compound (C42H58O6 658.91 g/mol) is estimated to account for >10% of total biogenic carotenoids (2). Fx is abundantly found in many dietary brown algae [0.1-18.6 mg Fx/g alga dry weight (dw)], and in certain microalgae that easily concentrate Fx (8.6-26.6 mg Fx/g alga dw) (3). In humans and rodents, orally administrated Fx is rapidly converted to cis-Fx, fucoxanthinol (FxOH), amarouciaxanthin A (Amx A) and cis-Amx A inside humans and rodent bodies (4-6). In particular, FxOH is a major metabolite of Fx in human blood (4, 5). No serious adverse events have been reported related to Fx administration in animal toxicity tests (7, 8). Fx supplementation exerts multifunctional effects, such as, anti-inflammatory, anti-obesity, anti-diabetic, anti-angiogenetic, anti-oxidative, and other effects (9-13). Notably, a well-investigated research topic is the one on anticancer effects of Fx or FxOH. To date, no epidemiological data demonstrating the suppressive effects of Fx or FxOH on cancer are available. However, Fx promotes apoptosis-inducing cell death in many organ types of cancer cells, such as cervical, gastric, lung, brain, and colorectal cancer cells. The mechanisms underlying induction of apoptosis in these cancer cells include the alterations of cell cycle, phosphatidylinositol 3 kinase/protein kinase B (PI3K/AKT), mitogen-activated protein kinase (MAPK), signal transducer and activator of transcription (STAT) signals, p38, and nuclear factor- B (NF-
B) signals, up-regulations of TP53 and p21, down-regulation of Bcl-2, and activation of caspase-3 (14-18). FxOH also induces apoptosis and anoikis in breast, colorectal, and pancreatic cancer cells through mechanisms similar to those described above (19-22). In animal experiments, most anticancer effects of Fx have been reported using colorectal cancer (CRC) murine models. Fx administration attenuated tumorigenesis and tumor development in a CRC murine model, azoxymethane/dextran sodium sulfate (AOM/DSS) mouse, through alterations of colorectal tumor microenvironment (TME), gut microbiota, and many signal transductions, such as adhesion, cell cycle, chemokine receptors, interleukins, MAPK, PI3K/AKT, STAT, transforming growth factor-β (TGF-β), Wingless/integrated (WNT)/β-catenin (23-26). Furthermore, Fx administration exerted anticancer effects in a 1,2-dimethylhydrazine-initiated murine CRC model and ApcMin/+ CRC murine model, although the detailed anticancer mechanisms are not known (27, 28).
A chemical structure of fucoxanthin. Molecular weight, 658.91.
Although the anticancer effects of certain agents have been thoroughly verified in cultured cancer cells and animal models of carcinogenesis, such effects have not always been observed in humans. In fact, many anticancer candidates have failed the late-stage clinical development and missed regulatory approval (29). Immortalized cancer cell lines, such as NCI-60, established from tumors of patients, their cells-derived xenograft animals, carcinogen-induced model animals, and transgenic cancer model animals have been used for the research on anticancer effects as useful tools for decades. However, their models are largely lacking primarily and tumor heterogeneity of individual patients. Therefore, these models readily lead to clinical failure (30, 31). Currently, three models closely resembling patient tumors have been developed: patient-derived cells (PDCs), patient-derived organoids (PDOs), and patient-derived xenografts (PDXs). PDCs are cells isolated from fresh patient tumors that undergo minimal genetic shifts after a small number of passages in culture, unlike traditional immortalized cancer cell lines. PDOs are three-dimension cell culture models that recapitulate the clonal heterogeneity. Both PDCs and PDOs are convenient tools for screening anticancer agents; however, they lack complex TME, such as cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) (32, 33). PDX models created by engraftment of tumor tissue of patients into immunodeficient animals have been developed as the best preclinical models, which resemble genetic profiles, pathological hallmarks, and TME of primary tumors. The response rate of PDX to anticancer agents correlates closely to clinical outcomes (34-36). If Fx shows anticancer effect in PDX animals, it could be used for human cancer prevention and treatment. In addition, the molecular mechanism of action in PDX animals may be similar to that in patient administered with Fx. Therefore, this study is meaningful for translational research. However, little information is available regarding the anti-cancer effects of Fx in PDX mice.
In the present study, we investigated anticancer effects of Fx in a colorectal cancer tissue-engrafted PDX (CRC-PDX) mouse model to investigate its use as a preventive and therapeutic agent against cancer in humans in the future.
Materials and Methods
Chemicals. A high-purity Fx powder (≥99%) was kindly donated by PATH Co., Ltd. (Tokyo, Japan), ALNUR Co., Ltd. (Tokyo, Japan), and Kampoikagakukenkyujyo Co., Ltd. (Osaka, Japan). All-trans-FxOH (purity, ≥98%), a metabolite of Fx, was enzymatically prepared from marine brown algal extracts by Dr. Hayato Maeda (Hirosaki University, Japan). RPMI-1640 cell culture medium (cat.no. 183-02165), Hank’s balanced salt solution (HBSS) (cat.no. 082-08961), dimethylsulfoxide (DMSO) (cat.no. 041-07217), isoflurane (cat.no. 099-06571), poly(oxyethylene 20 sorbitan) monolaurate (Tween 20) (cat.no. 166-21213), 2-amino-2-hydroxymethyl-1,3-propanediol (Tris) (cat.no. 013-16385), sodium dodecyl sulfate (SDS) (cat.no. 194-13985), and neutral buffered formaldehydes (37%) (cat.no. A16163) and (10%) (cat.no. 060-01667) were purchased from FUJIFILM Wako Pure Chemicals (Osaka, Japan). Fetal bovine serum (FBS) (cat.no.10437), RNAlater (cat.no. AM7021), GlutaMAX (cat.no. 35050061), Lipofectamine RNAiMAX (cat.no. 13-778-075), Opti-MEM I reduced serum medium (cat.no. 31985062), and penicillin/streptomycin (cat.no. 15-140-122) were purchased from Thermo Fisher Scientific (Carlsbad, CA, USA). Bovine serum albumin (BSA) (cat.no. 01281-97) and premix water-soluble tetrazolium salt cell proliferation assay system (WST-1) (cat.no. MK400) were obtained from Nacalai Tesque (Kyoto, Japan) and Takara Bio (Shiga, Japan), respectively. DynaMarker RNA High for Easy Electrophoresis (cat.no. DM170) was purchased from BioDynamics Laboratory (Tokyo, Japan). Supporting information (source, cat.no., and brand name) for each primary antibody was indicated to Table I. Goat anti-rabbit (cat.no. #7074) and anti-mouse (cat.no. SA00001-1) secondary antibodies conjugated with horseradish peroxidase (HRP) were obtained from Cell Signaling Technology (Danvers, MA, USA) and Proteintech (Rosemont, IL, USA), respectively. Recombinant human decorin (DCN or DCN-A, about 40 kDa) (cat.no. 143-DE) was obtained from R&D Systems/BioTechne (Minneapolis, MN, USA). Human colorectal cancer HT-29 (ATCC No. HTB-38) and HCT116 (ATCC No. CCL-247) cells were purchased from the American Type Culture Collection (Manassas, VA, USA). All other chemicals and solvents were of high-grade quality.
Supporting information of each antibody.
Human cancer tissue. An experimental scheme is shown in Figure 2. A Japanese PDX (J-PDX) library was prepared at the National Cancer Center Hospital in Japan using a part of cancer tissue resected from a patient with CRC, who provided written informed consent and agreed to give the sample collection and the information transmission, as described in a previous study (37). The protocol was conducted according to the principles outlined in the Declaration of Helsinki and approved by the Institutional Review Board of National Cancer Center (identification code, 2015-123 and 2021-163; authorization, 2015 and 2021). The surgical specimen of the patient’s cancer tissue was conveyed to a suitable facility for pathological and multi-omics analyses, and to establish PDX-derived tissue. The primary tissue represented as F0 was characterized by race, age, sex, location of sample collection, clinical characteristics of the disease, pathological type, TNM status, clinical stage, and prior treatment history (chemotherapy, radiotherapy and immunotherapy).
Experimental scheme. In the first step, an establishment of Japanese patient-derived xenograft (J-PDX) tumor was conducted with analyses of pathological finding in primary tumor and gene alteration of a Trans generation 3 (TG3) PDX tumor. In the second step, a preclinical trial of fucoxanthin (Fx) administration was performed using a PDX murine model transplanted a colorectal cancer J-PDX tumor (CRC-PDX). The anticancer effects of Fx were assessed by body weight and tumor size changes, pathological characterization, proteome profiles using LC-MS/MS, and protein expressions and activations using western blot. In last step, the molecular mechanisms suggesting the anticancer effects of Fx in the mice were confirmed using human colorectal cancers cells treated with fucoxanthinol that was a main metabolite for dietary Fx. The effects were evaluated by cell viability, protein treatment and gene knockdown assays, and protein expressions and activations using western blot.
Establishment of PDX tumor from human colorectal cancer tissue. The Trans Generation 3 (TG3) PDX tumor tissue was prepared using the F0 primary CRC tissue from the patient described in the Section Human cancer tissue in the Materials and Methods. Six-week-old female NOD.Cg-Prkdcscid Il2rgtm1Sug/ShiJic (NOG) mice were obtained from In-Vivo Science (Kanagawa, Japan) and acclimated for 1 week under controlled temperature, humidity and light/dark cycle (12 h light/dark). Standard solid chow and tap water were given ad libitum. The F0 CRC tissue was cut into 2 mm3 pieces and implanted subcutaneously in the right flank of the mice (7-week-old) (designated hereafter as CRC-PDX mice) using a 13G transplantation needle (Natsume Seisakusyo, Tokyo, Japan). Estimated tumor volumes in the mice were measured weekly. The estimated tumor volume (mm3) was expressed as the formula: a (mm) × b2 (mm)/2 (a, long range; b, short range). After the tumor volume was reached the range of 200 to 2,000 mm3, it was excised, cut into 2 mm3 pieces, and transferred to another NOG mouse using the same protocol (TG1). TG3 CRC-PDX tumors were prepared using the same protocol and stored in a J-PDX library in Japan (37). To analyze genetic variations in the tissue, the DNA and RNA from the TG3 tissue were extracted and purified using the KingFisher Cell and Tissue DNA kit (cat.no. 97030196) (Thermo Fisher Scientific, Waltham, MA, USA) and MagMAX mirVana Total RNA Isolation kit (cat.no. A27828) (Thermo Fisher Scientific, Waltham, MA, USA), respectively. Genetic profiles of the tissue for 161 cancer-related genes were analyzed using a next generation sequencing panel with Oncomine Comprehensive Assay v3 and the Ion Torrent Genexus System (software version 6.6) (cat.no. A46296) (Thermo Fisher Scientific) was performed in the tissue. Animal experiments were performed according to the guidelines of the Institute for Laboratory Animal Research, National Cancer Center Research Institute (identification codes, T17-020, T17-073 and T19-008; authorization, 2017 and 2021).
Animal experiments of Fx administration. The experimental design is shown in Figure 2. The TG3 CRC-PDX tumor tissue was provided by the National Cancer Center Research Institute (Tokyo, Japan). A 4-weeks-old female CB17.Cg-Prkdcscid Lystbg-J/CrlCrlj (SCID-Beige) mouse was purchased from Oriental Yeast (Tokyo, Japan) and acclimated for one week under controlled temperature, humidity, and 12 h light/dark cycle. Standard solid food (Grade: MF, Oriental Yeast Co. Ltd.) and tap water were given ad libitum. Two pieces of the tumors (size, 2 mm3) were implanted subcutaneously into the right flank of the mouse (designated hereafter as TG4-CRC-PDX) using a 13G transplantation needle (Natsume Seisakusyo, Tokyo, Japan). The mouse was observed twice a week for clinical signs and estimated tumor volume. After three months, the mouse was sacrificed under 2% isoflurane anesthesia (air, 2.0 l/min), and the tumor (TG4 CRC-PDX) was resected and cut into many pieces of 2 mm3 size tumors under HBSS (4°C) to generate TG5 CRC-PDX mice.
Female SCID-Beige mice (4-weeks-old) were purchased from Oriental Yeast, randomly assigned to three groups (3-4 mice/cage, 6-12 mice per group), and maintained in similar temperature, humidity, and 12 h light/dark cycle conditions. Standard solid MF and tap water were given ad libitum. After a week of acclimation, two pieces of the TG4 CRC-PDX tumors (2 mm3) were implanted subcutaneously in the right flank of the mice (5-week-old) (designated as TG5 CRC-PDX mice) using a 13G transplantation needle on day 0. Fx powder was mixed with standard MF powder (Grade: MF, Oriental Yeast Co. Ltd.) at a concentration of 0.3% Fx (3.0 mg Fx/g MF powder, Fx-high) and 0.1% Fx (1.0 mg Fx/g MF powder, Fx-low). The control diet contained only MF powder (control diet). Groups 1 and 2 mice were administered Fx-high (n=11) and Fx-low (n=6) diets, respectively, ad libitum for 20 days from the day 0, and then implanted with TG4 PDX tumors until sacrificed. Mice in Group 3 mice were given control diet (n=12) ad libitum during the same period. The animals were inspected routinely for clinical signs, mortality, dietary intake amount, body weight, and estimated tumor volume. The estimated tumor volume (mm3) was expressed as the formula: a (mm) × b2 (mm)/2 (a, long range; b, short range). Mice were sacrificed under 2% isoflurane anesthesia (air, 2.0 l/min). The entire tumor from each mouse was excised, washed with cold phosphate-buffered saline (PBS), and cut into several pieces for pathological, proteome, and western blot analyses. For histopathological examination, one piece of the tumor was fixed in 10% formalin for 2 days and prepared the hematoxylin and eosin (HE)-stained sections. Histopathological findings were examined to determine the degree of the tumor lesions in the HE sections prepared by Morphotechnology (Hokkaido, Japan). Histopathological findings of the tumor lesions were evaluated by an expert pathologist. Other tumor pieces were immediately permeated with RNAlater (500 μl) overnight at 4°C, added to equal volume of cold-PBS and supernatants were then removed by centrifugation. Next, the tumor tissues were frozen at −80°C until proteome and western blot analyses. The experiments were performed in compliances with the Institutional Ethics Review Boards of the National Cancer Center Research Institute (identification code, 2021-163; authorization, 2021) and of the Health Sciences University of Hokkaido (identification code, 21P003; authorization, 2021), according to the Institutional Guidelines for Animal Care and Use in the Health Sciences University of Hokkaido (identification code, 22-003; authorization, 2021).
Proteome analysis. Whole tumor tissues from Groups 1 and 3 were taken, sonicated in SDS lysis buffer (50 mM Tris-HCl, 1%SDS, pH 6.8), and denatured. Protein concentrations in tumor or cell samples were determined photometrically using the Bradford assay (Bio-Rad, Hercules, CA, USA) and the proteins were then denatured at 95°C for 5 min. Total proteins from Group 1 and 3 were prepared as a mixed sample (total 75 μg) with equivalent amount of proteins taken from five mice. The samples were digested with trypsin and subjected to LC-MS/MS-based proteomics analysis using a quantitative LC-MS/MS system (UltiMate 3000 RSLCnano LC System) equipped with a Q Exactive HF-X mass spectrometer (Thermo Fisher Scientific, Carlsbad, CA, USA) at the Kazusa DNA Research Institute (Chiba, Japan) (Analysis Registration number: KK1657). Human tissue proteins were identified using the Human UniProtKB/Swiss-Prot database (downloaded, 26, Nov, 2021) and the Prosit spectral library (https://www.proteomicsdb.org/prosit/). The proteins were filtered by the cutoffs related to peptide false discovery rate (FDR) (≤1.0%) and protein FDR (≤1.0%). The protein expressions in the mice of group 1 compared with those of group 3 was analyzed using Scaffold DIA software (Promega Software, Portland, OR, USA). Among the identified up-regulated and down-regulated proteins, the proteins suggested to contribute to the anticancer effects of Fx was selected based on a comprehensive literature review regarding cancer development and suppression on each molecule.
Cell viability assay by FxOH treatment. HT-29 and HCT116 cells were seeded at a density of 5×104 cells/ml into a 24-well plate in 10%FBS/RPMI (2.5×104 cells/well) and allowed to adhere to the wells for 1 day. The cells were exposed to 10%FBS/RPMI with FxOH (final concentrations, 5 and 20 μmol/l) and further incubated for 1 day. Control cells were treated with the vehicle (DMSO) alone. Cell viability was measured using the WST-1 assay. Absorbance at 450 nm was determined using an ELISA plate reader (TECAN Japan, Tokyo, Japan). To measure the expression and activation of these cells, they were seeded at a density of 5×104 cells/ml into a 10-cm dish in 10%FBS/RPMI (50×104 cells/well) and allowed to adhere for 1 day. The cells were exposed to 10%FBS/RPMI with FxOH (final concentrations, 5 and 20 μmol/l) and incubated for 1 day. Control cells were treated with the vehicle (DMSO) alone. Protein expression and activation of the cells were measured using western blot analysis as described in Section Western blot analysis of the Materials and Methods.
Western blot analysis. Whole tumor tissues sampled from mice in Groups 1 and 3 were collected and sonicated in SDS lysis buffer. The HT-29 and HCT116 cells with gene knockdown or FxOH treatment were harvested, washed twice with PBS, and sonicated in lysis buffer. Protein concentrations in the tumors or cells were determined photometrically using the Bradford assay, and the proteins then denatured at 95°C for 5 min. The tumor or cell proteins (10 μg each) were separated on a 10% gel using SDS electrophoresis. The gels were immersed in a transfer buffer, and transferred onto Hybond PVDF membranes (cat.no. 10600058) (Amersham Bioscience, Little Chalfont, UK). The PVDF membranes were washed with Tris-buffered saline containing 0.1% Tween 20 (TBS-T), incubated with a blocking buffer of TBS-T containing 1 w/v% BSA (1% BSA/TBS-T) at room temperature for 1 h, and probed with each primary antibody (diluted 1:1,000) in 1% BSA/TBS-T overnight at 4 °C. The membranes were incubated with HRP-conjugated anti-mouse or anti-rabbit secondary antibodies in TBS-T at room temperature for 1 h. Protein bands were visualized using Immobilon Western chemiluminescence HRP reagent (cat.no. WBKLS0500) (Millipore, Billerica, MA, USA). The image of each protein band was normalized to that of the loading control (β-actin) and quantified by comparing with that of control group using FIJI Image J2 software (version 2.9.0/1.53t) (NIH, http://rsb.info.nih.gov/ij/).
Cell viability assay in DCN-treated cells. HCT116 cells were seeded at a density of 5×104 cells/ml into a 96-well plate in 10%FBS/RPMI (5×103 cells/well) and allowed to adhere for 1 day. The cells were incubated in 10%FBS/RPMI with recombinant human DCN (final concentrations, 10 and 50 μg/ml) and incubated for 1 day. Control cells were treated with 10%FBS/RPMI only. Cell viability was measured using the WST-1 assay, as described in Section Cell viability assay by FxOH treatment of the Materials and Methods.
Gene knockdown. Twenty-seven mer of dicer-substrate short interfering RNAs (dsiRNAs) targeting the coding sequences of DSN1 mRNA in Homo sapiens were prepared using Integrated DNA Technologies (Coralville, IA, USA). The designed DSN1 dsiRNAs were as follows: DSN1 dsRNA duplex-1, 5′-GUA AAG GAA UGA UAC UAA UUU UCT A-3′ and duplex-2, 5′-GCA GGA GGC UAA AGA GAU AUU GUC C-3′; and negative control (NC), 5′-GUG UUC UAC ACC AUU ACU CAA UUC UUA-3′. DsiRNA/Lipofectamine RNAiMAX/Opti-MEM I complex solution was prepared according to the manufacturer’s instructions. HCT116 cells were routinely passaged in RPMI-1640 medium containing 10% heat-inactivated FBS and 1% GlutaMAX™ without antibiotics [10%FBS/RPMI (−)]. The cells were seeded into 6-well plates containing 10%FBS/RPMI (−) at a density of 10×104 cells/ml (20×104 cells/well). The cells were allowed to adhere for 1 day. The dsiRNA or NC/Lipofectamine RNAiMAX complex solution was added to the culture medium (final concentration of dsiRNA or NC: 10 nmol/l) for 1 day and boosted with the new complex solution for 1 day. Some of the DSN1 knockdown HCT116 cells were seeded into a 24-well plate at a density of 5×104 cells/mL (2.5×104 cells/well), and the plate was incubated for 1 day. Cell viability was measured using the WST-1 assay as described in Section Cell viability assay by FxOH treatment. The remaining DSN1 knockdown HCT116 cells were subjected to western blotting to analyze protein expression, as described in Section Western blot analysis.
Statistical analysis. Results are expressed as the mean±standard error (SE). Statistical differences in tumor incidence between Groups 1 and 3 were determined using Fisher’s exact probability test. The Shapiro-Wilk test was used to evaluate the normality of the distribution of the results. Differences between two groups with and without normal distribution were assessed using the post hoc Student’s t-test (parametric) or the Mann-Whitney U-test (nonparametric), respectively. In addition, differences among Groups 1-3 with and without normal distribution were analyzed using one-way ANOVA with post hoc Tukey test (parametric) or Kruskal-Wallis test with post hoc Dunn-Bonferroni test (nonparametric), respectively. Statistical analyses were performed using the SPSS Statistics version 25 (IBM Co. Ltd., Armonk, NY, USA). Differences were represented as statistically significant at *p<0.05, **p<0.01, and exact p-values.
Results
Characteristics of a tumor in a patient with CRC. Table II shows the general characteristics of a patient with CRC. The defined race, age, sex, and location of sample collection were Japanese, late 40 years, female, and sigmoid colon, respectively. The clinical and pathological features of the tumor of the patient with CRC were as follows: disease, primary sigmoid colon cancer without metastasis and recurrence; pathological type, adenocarcinoma; tumor-node-metastasis (TNM) status, T3N0M0; clinical stage, stage II; prior treatment history (chemotherapy, radiotherapy and immunotherapy), none. An integrated analysis of 161 genes in the Trans Generation 3 (TG3) tumor in PDX mice using the Oncomine database demonstrated that hotspot mutations in tumor protein p53 (TP53) and neuroblastoma RAS viral oncogene homolog (NRAS), two truncated mutations in AT-rich interaction domain 1A (ARID1A), and a truncated mutation in PMS1 homolog 2 (PMS2) were identified (Table III). No gene fusions or copy number variations were observed among the 161 genes.
Characteristics of a patient with colorectal cancer.
Hotspot and truncating mutations of four genes in a colorectal adenocarcinoma of a patient1.
Tumor suppressive effects of Fx-high and Fx-low doses in CRC-PDX mice. Fx-high (0.3 w/w%) and Fx-low (0.1 w/w%) doses were administered to CRC-PDX mice ad libitum for 20 days. No clinical signs of severe side-effects, such as decreased mouse movement or hair loss, were observed in groups 1-3 during the experimental period. The amounts of intake among the Fx-high (Group 1), Fx-low (Group 2), and control diet (Group 3) were almost at equal levels (no significant difference was confirmed using the Shapiro-Wilk test and one-way ANOVA test): Group 1, 158.7±3.1 g Fx-high/kg body weight (bw); Group 2, 164.0±2.5 g Fx-low/kg bw; Group 3, 162.8±2.8 g control diet/kg bw. The intake amounts of Fx in Groups 1 and 2 were calculated as 476.1 and 164.0 mg Fx/kg bw, respectively. The average liver weight ratios of Groups 1 and 3 were a similar: Group 1, 5.9±0.2%; Group 2, 5.9±0.1% (no significant difference was observed using Shapiro-Wilk test and Student’s t-test). No significant differences in body weights were observed among Groups 1-3 (Figure 3A). The estimated tumor sizes in Groups 1 and 2 were significantly decreased and/or tended to be lower, respectively, than in Group 3 at Day 20 before sacrifice: Group 1, 103.5±29.0 mm3 (0.6-fold vs. group 3); Group 2, 99.1±41.8 mm3 (0.5-fold vs. Group 3); Group 3, 185.4±35.7 mm3 (Figure 3B). However, little difference in the tumor size was observed among Groups 1-3 during most of the experimental periods (Day 0-13), except for days 17 and 20. Pathological hallmarks revealed that the well-differentiated adenocarcinoma (ADC) and moderately differentiated ADC in the tumors of Group 1 tended to increase and decrease, respectively, relative to those of Group 3. Poorly differentiated ADCs were not observed in either groups 1 or 3 (Table IV). Next, to estimate the degrees of differentiation and cell growth, we assessed malignant tissue area per whole tumor area. The malignant tissue area in the tumors of Group 1 tended to decrease relative to that of Group 3 (Figure 4A and B).
Body weight and estimated tumor size in colorectal cancer tissue-engrafted patient-derived xenograft (CRC-PDX) mice with or without fucoxanthin (Fx) administration. The mice in groups 1 (red dots), 2 (orange dots), and 3 (blue dots) were given Fx-high, Fx-low, and control diet, respectively, ad libitum for 20 days until sacrificed. Data are presented as mean±standard error (SE) (n=6-12). (A) Body weight changes in groups 1-3. After the normality distribution of the results was checked using the Shapiro-Wilk test, comparison among groups 1-3 was performed using one-way ANOVA test. No significant difference was observed among groups 1-3 during the entire experimental periods. (B) Estimated tumor size in mice. The sizes with visible tumor (about ≥1 mm of major axis) were estimated based on a formula: a (mm)×b2 (mm)/2 (a, long range; b, short range). After the normality distribution of the results was checked using Shapiro-Wilk test, the comparison among groups 1-3 was performed using Kruskal-Wallis test with post hoc Dunn-Bonferroni test. *p<0.05 (*statistically significant difference was found between groups 1 and 3 on day 20).
Incidence (%) of differentiated adenocarcinoma (ADC) in colorectal cancer tissue-transplanted patient-derived xenograft (CRC-PDX) mice with fucoxanthin (Fx) administration.
Malignant tissue areas per whole tumor tissue areas on colorectal cancer tissue-transplanted patient-derived xenograft (CRC-PDX) mice with fucoxanthin (Fx) administration. Malignant tissue area per whole tumor tissue area between Fx-high diet administered (group 1) and control (group 3) mice were evaluated by a clinical pathologist. (A) Representative histopathology of adenocarcinomas (ADCs). Upper panel, a well differentiated tubular ADC (Tub 1) on one mouse of group 1. Lower panel, a moderately differentiated tubular ADC (Tub 2) on one mouse of group 3. (B) The malignant tissue area (%) in whole tumor tissue area. Flat bars, mean±standard error (SE) (n=10-11). After the normality distribution of the results was checked using Shapiro-Wilk test, the comparison between groups 1 and 3 was performed using post hoc Student’s t-test. n.s., No significance.
Proteome profiling in the tumor tissue of CRC-PDX mice after Fx-high administration. First, comprehensive protein profiles targeting epithelial tissue in the tumor tissue of CRC-PDX mice following Fx-high administration were investigated using LC-MS/MS. As a result, 714 and 364 proteins (1,078 proteins in total) were found to be up-regulated and down-regulated, respectively, in Group 1 relative to those in Group 3. Nineteen up-regulated proteins (≥1.5-fold) were suggested to contribute to the suppression of cancer development following Fx-high administration (Table V). In particular, we observed an up-regulated protein, decorine (DCN) (1.6-fold) (the protein is displayed in bold in Table V). In addition, 65 down-regulated proteins (≤−1.5-fold) were suggested to contribute to suppression of cancer development following Fx-high administration (Table VI). In particular, we noted the 22 down-regulated proteins, such as collagen α1 (XVII) chain (COL17A1) (N.D., Not detected in Group 1 but detected in Group 3), transcription factor p65 subunit (RELA) (N.D.), Lipocalin 2 (LCN2) (N.D.), bone morphogenetic protein 1 (BMP1) (N.D.), tyrosine-protein kinase Yes (YES1) (−17.2-fold), Pleckstrin-2 (PLEK2) (−3.8-fold), dual specificity mitogen-activated protein kinase kinase 3 (MAP2K3) (−2.4-fold), elongation factor 1-α2 (EEF1A2) (−2.4-fold), integrin α3 (ITGA3) (−2.2-fold), BAG family molecular chaperone regulator 1 (BAG1) (−2.0-fold), semaphoring-4D (SEMA4D) (−2.0-fold), dermcidin (DCD) (−2.0-fold), ribose-5-phosphate isomerase (RPIA) (−2.0-fold), Disintegrin and metalloprotease domain-containing protein 17 (ADAM17) (−1.9-fold), Glycogen synthase 1 (GYS1) (−1.8-fold), Kinetochore-associated protein DSN1 homolog (DSN1) (−1.7-fold), laminin subunit α-5 (LAMA5) (−1.7-fold), leucine-rich repeat protein SHOC-2 (SHOC2) (−1.6-fold), fibroblast growth factor receptor 4 (FGFR4) (−1.6-fold), carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) (−1.6-fold), kinetochore protein Spc 25 (SPC25) (−1.5-fold), and mucin-5B (MUC5B) (−1.5-fold) as key regulators related to anticancer effects of Fx (the proteins are displayed in bold in Table VI).
Profile of up-regulated proteins identified by LC-MS/MS in whole tumor tissue of colorectal cancer tissue-transplanted patient-derived xenograft (CRC-PDX) mice treated with fucoxanthin (Fx) administration1.
Profile of down-regulated proteins identified by LC-MS/MS in whole tumor tissue of colorectal cancer tissue-transplanted patient-derived xenograft (CRC-PDX) mice treated with fucoxanthin (Fx) administration1.
Protein expression and activation in tumor tissue of CRC-PDX mice after Fx-high administration based on proteome profiles. Protein expression levels selected based on the proteome profile were investigated in whole tumors of Fx-high–administrated CRC-PDX mice using western blot analysis. In the present study, we focused on molecules that showed remarkably different profiles between Groups 1 and 3 in the median, average, and interquartile ranges (25-75%) using box plots. Consequently, administration of Fx-high diet in Group 1 altered the median expression levels of glycanated DCN (Gc-DCN) (2.7-fold), LCN2 (4.3-fold), ADAM17 (1.8-fold), GYS1 (1.5-fold), as well as average expression levels of PLEK2 (1.3-fold) and DSN1 (0.6-fold) compared with those of Group 3. In addition, the interquartile range of RELA tended to be lower in Group 1 than in Group 3. Little change or appearance was observed in the expressions of non-glycanated DCN (DCN-A), DCN-B, and other 16 proteins between the two groups (Figure 5).
Protein expression profiles in tumors in colorectal cancer tissue-transplanted patient-derived xenograft (CRC-PDX) mice with fucoxanthin (Fx) administration. Protein expression levels between Fx-high administered (group 1) and control (group 3) mice were determined using western blot analysis. Density of each protein band of tumors of CRC-PDX mice in groups 1 and 3 were normalized to the density of the β-actin band from the image using the FIJI image J2 software. Each protein level in group 1 (red box plot, n=5) was represented compared to that of group 3 (white box plot, n=5). After the normality distribution of the results was checked using the Shapiro-Wilk test, comparison between groups 1 and 3 was performed using post hoc Student’s t-test. There was no significant difference between groups 1 and 3 on all 25 proteins tested. N.D., not detected.
Next, protein expression and activation of molecules belonging to the upstream and downstream regions of Gc-DCN, RELA, and DSN1, which are thought to be involved in the anticancer effect of Fx, were examined in tumor tissue. The results showed that the average levels of phospho(p) focal adhesion kinase (pFAK)(Tyr397) (0.6-fold), pPaxillin(Tyr31) (0.5-fold), and c-MYC (0.7-fold) tended to decrease in Fx-administered Group 1 than in Group 3. Little change or appearance was observed in the expression or activation of other 22 proteins between the two groups (Figure 6).
Protein expression and activation profiles in tumors in colorectal cancer tissue-transplanted patient-derived xenograft (CRC-PDX) mice with fucoxanthin (Fx) administration, of molecules that are located upstream and downstream to decorin (DCN), transcription factor p65 subunit (RELA), and kinetochore-associated protein DSN1 homolog (DSN1). Protein expression and activation levels in Fx-high diet administered (group 1) and control (group 3) mice were determined using western blot analysis. Density of each protein band of tumors of CRC-PDX mice in groups 1 and 3 were normalized to density of β-actin–expressed band using FIJI image J2 software. Each protein level in group 1 (red box plot, n=5) was represented compared to that of group 3 (white box plot, n=5). After the normality distribution of the results was checked using the Shapiro-Wilk test, comparison between groups 1 and 3 was performed using post hoc Student’s t-test. There was no significant difference between groups 1 and 3 on all of 25 protein expressions. N.D., Not detected; PI3K/AKT, phosphatidylinositol-3 kinase/protein kinase B; NF-B, nuclear factor kappa B; TGF-β, transforming growth factor-β; MAPK, mitogen-activated protein kinase. WNT/β-catenin; Wingless/β-catenin; TME, tumor microenvironment; EMT, epithelial mesenchymal transition.
Protein expression and activation in human colorectal cancer cells with Fucoxanthinol (FxOH) treatment. FxOH is the main metabolite in blood and is derived from Fx intake in humans (4, 5). We examined the suppressive effects of FxOH on human CRC cells by treating them with FxOH instead of Fx. Protein expression and activation were investigated in HT-29 and HCT116 cells treated with FxOH using western blot analysis. We investigated the growth inhibitory effects of FxOH on HT-29 and HCT116 cells. Addition of FxOH (5 and 20 μmol/l) significantly attenuated cell growth in both the cell types in a dose-dependent manner than in control cells: 5 μmol/l FxOH in HT-29 cells, 70.3±1.9%; 20 μmol/l FxOH in HT-29 cells, 59.4±1.2%; 5 μmol/l FxOH in HCT116 cells, 89.7±6.0%; 20 μmol/l FxOH in HCT116 cells, 52.4±2.8% (Figure 7A). Next, protein expression and activation levels selected based on altered protein levels in the whole tumor of Fx-high administrated CRC-PDX mice were investigated in the two cell lines using western blot analysis. The results suggested that FxOH treatment (5 and/or 20 μmol/l) up-regulated the expression levels (≥1.5-fold change) of DCN-A, pro-caspase-3 and active forms of caspase-3, and down-regulated the expression levels (≤0.6-fold change) of integrin α5, pFAK(Tyr397), pPaxillin(Tyr31), and c-MYC in HT-29 cells. In addition, FxOH treatment (5 and/or 20 μmol/l) up-regulated the expression levels (≥1.5-fold change) of DCN-A, and active forms of caspase-3, and down-regulated the expression levels (≤0.6-fold change) of DSN1, Cyclin D1, integrin α5, pFAK(Tyr397), and c-MYC in HCT116 cells (Figure 7B).
Effects of fucoxanthinol (FxOH) on growth and protein alterations in human colorectal cancer cells. HT-29 and HCT116 cells were treated with 5 and 20 μmol/l of FxOH for one day. Control cells received vehicle only. (A) Growth of HT-29 and HCT116 cells for the two tests was determined using the WST-1 assay. Data for cell growth were represented as mean±standard error (SE) (n=6). After the normality distribution of the results was checked using Shapiro-Wilk test, comparison among cells treated with 5 and 20 μmol/l FxOH, and control cells was performed using one-way ANOVA with post hoc Tukey test for HT-29 cells and Kruskal-Wallis test with post hoc Dunn-Bonferroni test for HCT116 cells. *p<0.05 and **p<0.01. (B) Protein expression and activation levels in the HT-29 and HCT116 cells with or without FxOH treatment were determined using western blot analysis. Density of each protein band was normalized to that of β-actin, a loading control, from the image using FIJI image J2 software. Each protein level in cells treated with 5 and 20 μmol/l FxOH was represented by comparing with the average value (1.0-fold) of control cells. aβ-Actin as a loading control of a membrane analyzing c-MYC, cyclin B1, integrin α5, pro-caspase-3, and active form of caspase-3. bβ-Actin as a loading control of a membrane analyzing pFAK(Tyr397), pPaxillin(Tyr31), DSN1, integrin β1, and pERK1/2(Thr202/Tyr204). cβ-Actin as a loading control of a membrane analyzing p-p38(Thr180/Tyr182), pJNK(Thr183/Tyr185), Glycanated DCN (Gc-DCN), non-glycanated DCN (DCN-A), and DCN-B.
Effects on cell growth of non-glycanated DCN (DCN-A) treatment and DSN1 gene silencing in human colorectal cancer HCT116 cells. To confirm whether DCN is an important suppressor of the growth of human colorectal cancer HCT116 cells, recombinant DCN-A protein was added to HCT116 cells. The growth of HCT116 cells significantly decreased in a dose-dependent manner compared to that of control cells: 10 μg DCN/ml in HCT116 cells, 94.7±0.5%; 50 μg DCN/ml in HCT116 cells, 86.4±0.6% (Figure 8A). To confirm whether DSN1 is an important regulator of HCT116 cell growth, its expression was silenced using dsiRNA gene knockdown. The DSN1 protein expressions of both DSN1 dsiRNA-1 and −2–transfected HCT116 cells were significantly lower than that of NC-transfected cells: DSN1 dsiRNA-1, 0.2-fold; DSN1 dsRNA-2, 0.1-fold (Figure 8B). The growth of DSN1 dsiRNA-1 and −2–transfected HCT116 cells were significantly attenuated compared with that of the NC cells: DSN1 dsRNA-1, 91.8±1.1%; DSN1 dsRNA-2, 48.7±0.3% (Figure 8C).
Effects of non-glycanated decorin (DCN-A) treatment and Kinetochore-associated protein DSN1 homolog (DSN1) knockdown on cell growth in human colorectal cancer cells. (A) HCT116 cells were treated with recombinant human DCN-A (final concentrations, 10 and 50 μg/ml in 10%FBS/RPMI medium) for one day. Control cells were cultured with 10%FBS/DMEM. The cell growth of HCT116 cells was determined using a WST-1 assay (n=4). After the normality distribution of the results was checked using the Shapiro-Wilk test, comparison among cells treated with 10 and 50 μg/ml DCN-A, and control cells was performed using one-way ANOVA with post hoc Tukey test. *p<0.05 and **p<0.01. (B) HCT116 cells were transfected with dicer-substrate short-interfering RNA (dsiRNA) targeting DSN1 for 2 days. The dsiRNA-1 and -2 represented dsiRNA duplex-1 and -2 sequences described in section 5.9 of Materials and Methods. Control cells were transfected with negative control (NC) dsiRNA. Protein expression of DSN1 was determined using western blot analysis. Density of the protein bands was normalized to that of the β-actin, a loading control, using FIJI image J2 software. Each protein level in DSN1 dsiRNA-1 and -2–treated cells was represented by comparing with the average value (1.0-fold) of the NC cells. (C) DSN1 dsiRNA-1 and -2–transfected HCT116 cells were incubated with 10%FBS/DMEM for 1 day. Cell growth of HCT116 cells was determined using a WST-1 assay (n=4). After the normality distribution of the results was checked using Shapiro-Wilk test, comparison among cells treated with DSN1 dsiRNA-1 and -2, and control cells was performed using one-way ANOVA with post hoc Tukey test. **p<0.01.
Discussion
This study demonstrated that Fx administration significantly decreased tumor growth in sigmoid CRC-PDX mice with the alterations of certain molecules. This is a novel finding suggesting the preclinical therapeutic effects of Fx on CRC.
The tumors derived from a patient with CRC were primary sigmoid colon adenocarcinomas (T3N0M0 and stage II), which did not display metastasis and recurrence. In addition, five mutations in four genes (TP53, ARID1A, NRAS, and PMS2) were identified in the tumors using the Oncomine Comprehensive Assay v3 (Table II and Table III). TP53 and ARID1A are tumor suppressor genes. NRAS and PMS2 are an oncogene and mismatch repair gene, respectively. In general, it is regarded that CRC progresses through a multistep pathogenesis from dysplastic crypts to adenocarcinomas. Onsets of stepwise gene mutations of adenomatous polyposis coli (APC), Kirsten-ras (KRAS), deleted in colorectal cancer (DCC), mothers against decapentaplegic homologue (SMAD) 2 (SMAD2), SMAD4, and TP53 are strongly associated with the CRC development (38-41). We did not know whether mutations in APC, DCC, and SMAD2 existed in PDX tumors because the Oncomine Comprehensive Assay v3 did not cover the detections of the 3 mutations. Increasing evidence suggests that the gene mutations in TP53, ARID1A, NRAS, and PMS2 are frequent in patients with CRC (42, 43). Therefore, we speculated that the TP53, ARID1A, NRAS, and PMS2-associated signals, such as EGFR, PI3K/AKT, MAPK, NF-kB, EMT, p53, WNT/β-catenin, DNA mismatch repair, angiogenesis, apoptosis, and others, might be key regulators for tumor development in the CRC-PDX mice (42, 44-46).
In the present study, a Fx-high diet (0.3% Fx) was administered ad libitum to CRC-PDX mice for 20 days. The estimated average Fx intake was 476.1 mg Fx/kg bw. Human equivalent dose relative to the human body surface area was calculated as 2,322.4 mg Fx/m2/human 60 kg bw/day (476.1/12.3×60), according to previous reports (47, 48). No representative abnormalities, such as mouse movement, hair loss, decreased body weight, or decreased liver weight were observed during the experimental periods. In a previous study, a safe dose of the oral Fx administration was confirmed up to 2,000 mg Fx/kg mouse bw, the human equivalent dose was 9,756.1 mg Fx/m2/human 60 kg bw/day (7). In the present study, this dose (2,322.4 mg Fx/m2/human 60 kg bw/day) of Fx was within the dose range (400-3,000 mg/m2) of 5-fluorouracil, a representative high-dose CRC therapeutic drug administered to patients with cancers, and therefore this dose might not be far from the realistic dosage for prospective clinical applications as an anticancer agent (49). However, there is a need to determine the optimal dose of this compound for clinical application in patients with CRC.
Oral administration of the Fx-high diet significantly retarded the tumor growth (0.6-fold) and tended to induce differentiation in Group 1 compared to Group 3 (Figure 3 and Table IV). In addition, the Fx-high diet altered the median expression values of Gc-DCN (2.7-fold), LCN2 (4.3-fold), ADAM17 (1.8-fold), and GYS1 (1.5-fold), as well as the average expressions of PLEK2 (1.3-fold) and DSN1 (0.6-fold) (Figure 5). Alterations in the molecules belonging to the upstream and downstream of Gc-DCN, RELA, and DSN1 were examined in the tumor tissue. The results showed that Fx administration in Group 1 tended to decrease pFAK(Tyr397), pPaxillin(Tyr31), and c-MYC levels than in mice in Group 3. Since p50 expression was unchanged, it seems unlikely that the contribution of the NF-B signal contributes to anticancer effects of Fx in the mice (Figure 6). Although the increase of LCN2, ADAM17, GYS1, and PLEK2, which are regulators of cancer development, was not well understood, these molecular enhancements may be due to compensatory reactions (cell resistance or survival) against anticancer agent. In vitro experiments demonstrated that DSN1, DCN, pFAK(Tyr397), pPaxillin(Tyr31), and c-MYC may be key molecules related to inhibition of cell growth and/or induction of apoptosis in human CRC cells treated with FxOH (Figure 7A and B, and Figure 8A-C). Therefore, we noted DCN, DSN1, pFAK(Tyr397), pPaxillin(Tyr31), and c-MYC expressions, and the related signals (growth, adhesion, and cell cycle) as key regulations for the anticancer effects of Fx in the mice.
DSN1, one of the proteins in the kinetochore protein assembly, contributes to promotion of cell cycle and is associated with the malignant progression in patients with CRC. DSN1 plays an important role not only in the cell cycle but also in cell growth, migration and invasion in human CRC cancer cells (50, 51). DCN is an ECM-embedded proteoglycan widely expressed in mammalian cells. Several types of DCNs exist: core protein (DCN-A, about 40 kDa), core proteins binding one or more glycosaminoglycans side chains (Gc-DCN, approximately 80-90 kDa), and small molecular weight of DCNs (e.g., DCN-B, approximately 25 kDa). DCN-A and/or Gc-DCN are multifunctional tumor suppressors that mediate attenuation of the cell cycle, tumorigenesis, EMT, certain signals (e.g., TGF-β, WNT/β-catenin, MAPK, PI3K/AKT/mTOR), and certain molecules (e.g., c-MYC, HIF-1α, and Rho A), as well as enhancement of apoptosis, autophagy, and of certain molecules (e.g., caspase-3, p21, LC3, and Beclin 1) (52, 53). On the other hands, it has been suggested that DCN-B accelerates cell proliferation, migration, and apoptosis, in contrast to the tumor suppressive effects of DCN-A (54). DCN is negligibly detected in cancer tissues of patient with CRC and in human CRC cancer cells, although it is well expressed in stromal cells (55). DCN secreted via paracrine effect in stromal cells positively regulates TME formation (56). DCN inhibits cell growth and colony formation in HCT116 cells (55, 57). As described above, DCN in tumor tissues originates from both epithelial and stromal cells. In the present study, αSMA, a marker protein for CAFs that is the major stromal cells in cancer tissues, was not significantly different between Groups 1 and 3 (Figure 6). In addition, DCN-B was not detected in the tumor tissues of both Groups 1 and 3 (Figure 5). Therefore, DCN from stromal cells and DCN-B were thought to have little effect on the antitumor effects of Fx in the CRC-PDX mice. To date, little information is available regarding DSN1 or DCN expression in carcinogenic animals and cancer cell models treated with Fx. Integrins, which are representative cell-ECM receptors, are key regulators of adhesion, cell growth, TME formation, and tumorigenesis. Integrins activates FAK, stimulate downstream signaling, and then regulate cellular migration, TME formation, and tumorigenesis. Both integrins and FAK are both overexpressed in many types of cancer and contribute to the malignancies (58-60). Paxillin is an intracellular scaffold protein that mediates protein-protein interactions, directly binds to FAK, p210BCR/ABL, and vinculin, and subsequently promotes adhesion, migration, invasion, survival, EMT, and tumorigenesis (61). Recently, we revealed that Fx administration could induce anchor-dependent anoikis in both colorectal mucosal crypts and adenocarcinoma in an AOM/DSS CRC murine model via down-regulation of integrin β1, pFAK(Tyr397), and pPaxillin(Tyr31) (25). Therefore, attenuation of the adhesion signal was suggested to be one of the key control mechanisms in the Fx-administered CRC-PDX model. c-MYC, an oncoprotein, is regulated by signals, such as WNT/β-catenin and Shh, and mainly facilitates cell growth, cell cycle, adhesion, cancer stemness, and metastasis (42, 62). Addition of Fx inhibited cell growth in human ovarian cancer cells by attenuating STAT3/c-MYC signal activation (63). Down-regulation of c-MYC may also play an important role in regulating the anticancer effects of Fx in CRC-PDX mice.
In the present study, dysregulation of signals by TP53, ARID1A, NRAS, and PMS2 mutations might have been mitigated because Fx administration effectively suppressed tumor growth in CRC-PDX mice. In addition, the alterations in DSN1, Gc-DCN, pFAK(Tyr397), pPaxillin(Tyr31), and c-MYC, as well as related signals (growth, adhesion, and cell cycle) may have controlled the mechanisms triggered by mutated TP53, ARID1A, NRAS, and PMS2. Further investigation is required to elucidate the anticancer effects of Fx in CRC-PDX mice.
Conclusion
The characteristics of CRC tissue obtained from a Japanese patient were a primary adenocarcinoma with T3N0M0, stage II, and mutations of genes, such as TP53, ARID1A, NRAS, and PMS2. Fx administration significantly suppressed tumor development, with a tendency toward differentiation. DSN1, Gc-DCN, c-MYC, pFAK(Tyr397), and pPaxillin(Tyr31) are suggested to be key regulators of anticancer effects of Fx in the mice. Therefore, we confirmed the importance of these molecules for growth of human CRC cells using gene knockdown, recombinant protein treatment, and FxOH addition assays. Our findings suggest that Fx may be an attractive candidate for cancer therapy in patients with CRC.
Acknowledgements
Fx powder was kindly donated by PATH Co., Ltd. (Tokyo, Japan), ALNUR Co., Ltd. (Tokyo, Japan), and Kampoikagakukenkyujyo Co., Ltd. (Osaka, Japan). We would like to thank Wataru, Murase, Ayaka Yasuda, Momoka Wagatsuma, Takumi Ouchi, Yoshiro Kikuchi, Yuichi Shimazaki, Teppei Fujiwara, Sally Suzuki, Marin Nakagawa, Yuki Shimizu, Yuhei Shoji, and Atsuhito Kubota of School of Pharmaceutical Sciences, and the staffs of the Center for Experimental Animals at the Health Sciences University of Hokkaido for their support of animal treatment.
Footnotes
Conflicts of Interest
None declared.
Authors’ Contributions
M.Terasaki conceived and designed the study. M.Terasaki, K.T., T.T., H.M., M.S., Y.K., S.S., Y.S., and A.H. performed the experiments. M.Terasaki wrote the paper. K.M. and M.Takahashi reviewed and edited the manuscript. All Authors have read and agreed to the published version of the manuscript.
Funding
This work was supported, in part, by the Japan Society for the Promotion of Science KAKENHI (Grant Number 20K05879). In addition, The National Cancer Center J-PDX library was partly supported by the AMED (Grant no. 17pc0101011h0001) and the National Cancer Center Research and Development Fund, Japan.
- Received September 8, 2023.
- Revision received October 21, 2023.
- Accepted October 23, 2023.
- Copyright © 2023, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved
This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).