Treatment of hepatocellular carcinoma (HCC) is currently challenging. Cancer-associated fibroblasts (CAFs) promote the malignancy of HCC cells via production of cytokines. Conophylline (CnP), a vinca alkaloid obtained from Ervatamia microphylla leaves, has been reported to suppress activation of hepatic stellate cells and liver fibrosis in rats. We examined the efficacy of CnP in suppressing tumor growth in HCC. Specifically, we investigated whether CnP could inhibit CAFs, which were derived from HCC tissues in vitro and in vivo. Same as previous reports, CAFs promoted proliferative and invasive ability of HCC cells. CnP suppressed α-smooth muscle actin expression of CAFs, and inhibited their cancer-promoting effects. CnP significantly suppressed CAFs producting cytokines such as IL6, IL8, C-C motif chemokine ligand 2, angiogenin, and osteopontin (OPN). Combined therapy with sorafenib and CnP against HCC cells and CAFs in vivo showed to inhibit tumor growth the most compared with controls and single treatment with CnP or sorafenib. Transcriptome analysis revealed that GPR68 in CAFs was strongly suppressed by CnP. The cancer-promoting effects of cytokines were eliminated by knockdown of GPR68 in CAFs. CnP inhibited the HCC-promoting effects of CAFs by suppressing several HCC-promoting cytokines secreted by CAFs expressing GPR68. Combination therapy with CnP and existing anticancer agents may be a promising strategy for treating refractory HCC associated with activated CAFs.

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and is challenging to treat (1). Cancer-associated fibroblasts (CAFs) are reported to promote the malignancy of HCC cells (2, 3). Treatment of HCC has advanced with the availability of the multikinase inhibitor, sorafenib; however, CAFs increase the resistance of HCC cells to this drug via secretion of cytokines such as IL6 (4–7). Many cytokine regulation mechanisms are reported for cancer and hematopoietic cells (8), and IL6 is reported to be regulated by G protein–coupled receptor 68 (GPR68; ref. 9).

Conophylline (CnP) is a vinca alkaloid obtained from the leaves of Ervatamia microphylla (Supplementary Fig. S1A). The compound is reported to have a effect of β-cell differentiation in pancreatic precursor cells and quell liver fibrosis by suppressing the activation of hepatic stellate cells (10, 11). Recently, we reported that CnP can suppress pancreatic cancer cell growth by inhibiting CAFs (12). However, to date, no investigation into the effects and mechanisms of action of CnP on CAFs in HCC has been reported.

This study aimed to investigate whether CnP can reduce HCC aggressiveness by suppressing activated CAFs. We thus evaluated the effects of CnP on CAFs derived from HCC. In addition, we evaluated the therapeutic benefit of combining the conventional anti-HCC agent sorafenib with CnP to target activated CAFs using mouse models. Subsequently, we elucidated the fundamental mechanism of the effect of CnP-altered CAFs using cap analysis of gene expression (CAGE, RRID:SCR_007574).

Cell isolation and cell cultures

We established primary CAFs from HCC tissues, which were surgically resected as reported by Lau and colleagues (6). Specifically, HCC tissues with their surrounding capsules were minced and incubated in DMEM containg 1% penicillin–streptomycin (Thermo Fisher Scientific), 20% FBS, and 1 ng/mL basic FGF (bFGF, Wako). To allow cells to attach to the culture plates, the samples were cultured in a humidified incubator with 5% CO2 at temperature of 37°C. We replenished the medium and removed unattached cells twice a week. The cells with myofibroblast-like morphology were defined as CAFs. Expression of α-smooth muscle actin (α-SMA) of cells was evaluated using Western blotting. CAFs were used between passage numbers 4 and 8. We were approved to establish CAFs from surgically resected HCC tissues by the Clinical Ethic Committee in our University (approval number 2016–118). CAF1 and CAF2 were isolated from different patients with HCC.

The human hepatoblastoma cell line HepG2 (RRID: CVCL_0027) and the human HCC cell line PLC/PRF/5 (RRID:CVCL_0485) were also used in this study. Both were purchased from the JCRB Cell Bank and authenticated within the last 3 years by short tandem repeat DNA profiling by BEX. All experiments were performed with Mycoplasma-free cells. Cells were cultured in DMEM containg 1% penicillin–streptomycin and 10% FBS and cultured at 37°C in a humidified incubator with 5% CO2 atmosphere. We used cells between passage numbers 4 and 8.

CnP and sorafenib

The CnP used in the in vitro study was isolated from leaves of E. microphylla grown in Thailand and purified as described previously (Supplementary Fig. S1A; ref. 13). Specifically, a chloroform extract of the leaves of E. microphylla (4.97 g) was chromatographed on a silica gel column with hexane:ethylacetate (1:2) to yield 99.7 mg of the active fraction. This fraction was dissolved in MeOH, and the insoluble fraction was removed. The solution was then applied to a Toyopearl HW-40 column and eluted with methanol to yield 20.5 mg of active material. Finally, the crude material was purified by centrifugal partition chromatography and freeze-dried from t-butyl alcohol to yield 15.0 mg of white powder. The crude CnP preparation II (CCP-II) used for the in vivo study was obtained from leaves of Tabernaemontana divaricata. The method for extraction was described previously (14). Dry leaves (5 kg) were added to 500 L of 0.025 N hydrochloric acid and heated to 60°C for 30 minutes with gentle mixing. A clear supernatant was obtained by centrifugal filtration. The supernatants obtained from three serial extractions were pooled and cleaned with column chromatography using a synthetic adsorbent resin, Diaion HP20 (Mitsubishi Chemical). The column was washed with water followed by 30% (colume for volume) ethanol, and CnP was then eluted with pure ethanol. The elute was concentrated in vacuo and lyophilized. The CCP-II thus obtained contained 14 mg/g of CnP. Sorafenib was provided by Bayer HealthCare Pharmaceuticals Inc.

Protein extraction and Western blot analysis

Protein extraction was performed using RIPA buffer (Wako), following the manufacturer's protocol. SDS-PAGE with 10% Bis–Tris gels was used for separating proteins. They were transferred to nitrocellulose sandwiches (No. 12369; Cell Signaling Technology). After blocking with 5% BSA or 5% skim milk, the membranes were incubated with primary antibodies overnight at 4°C. Anti-α-SMA mouse mAb (A2547; 1:1,000; Sigma-Aldrich, RRID:AB_476701), GPR68 rabbit antibody (No. ab133818; 1:1,000; Abcam), anti–β-actin mouse mAb (A5316; 1:1,000; Sigma-Aldrich, RRID:AB_476743) were used as primary antibodies. We treated the membranes with horseradish peroxidase–linked secondary antibodies. We detected the bands of protein on it with an Image Quant LAS 4000 instrument (GE Healthcare) and ECL Prime Western Blotting Detection System.

Conditioned medium

We cultured CAFs in DMEM containg 1 ng/mL bFGF and 20% FBS till they proliferated 90% confluency. Next, we changed the medium and the CAFs were cultured with DMEM (serum-free) for an additional 5 days. CAF-CM (conditioned medium) was then collected and centrifuged for 10 minutes at 1,500 g. The CnP effect on the CAF-CM was evaluated in CAFs cultured in DMEM (serum-free) containing CnP (0.3 μg/mL) for 5 days, and the CM was collected as CnP-treated CAF-CM.

Cell proliferation assay

The Cell Counting Kit-8 (CCK-8; Dojindo Laboratories) was used to evaluate cell proliferation. We seeded and cultured HCC cell lines and CAFs onto 96-well plates. We removeded the culture medium and change to DMEM (serum-free) with varying concentrations (from 0 to 1 μg/mL) of CnP after overnight incubation. Proliferation of HCC cell lines was also assessed by replacing HCC cell medium with CAF-CM and CnP-treated CAF-CM. We evaluated them by measuring the absorbance of each well after 48 hours using a spectrophotometerat 450 nm with a reference of 650 nm (Bio-Rad).

Cell invasion assay

We evaluated the invasion ability of cells using 24-well Corning BioCoat Matrigel Invasion Chambers (Corning). The lower chamber was filled with medium supplemented with 3% FBS containing CnP (0.3 μg/mL), CAF-CM, or CnP-treated CAF-CM, and HCC cell lines in DMEM (serum-free) were seeded into the upper chamber. After 48 hours, Diff-Quik stain (Sysmex) were used for fixity and staining the cells. We evaluated five randomly selected fields under a microscope and counted the cells that invaded and migrated through the membrane.

Cytokine array and ELISA

Cytokine Array Kit (ARY022B; R&D Systems) was used for evaluation of cytokine profiles in CAF-CM and CnP-treated CAF-CM, following the manufacturer's instructions. An Image Quant LAS 4000 imager was used for quantification of spots on the array. An ELISA was used for measuring concentrations of cytokines in CAF-CMs. ELISA kits of IL6 (ab46027; Abcam), IL8 (ab46032; Abcam), and chemokine (C-C motif) ligand 2 (CCL2; ab179886; Abcam) were used following the manufacturer's instructions.

RNA extraction and reverse transcription-quantitative PCR

We extracted total RNA using a miRNeasy Mini Kit (No. 217004, Qiagen) and quantified it with an ND-1000 (NanoDrop Technologies). Reverse transcription and quantitative (RT-q) PCR were performed using an RT Kit (Toyobo) and Power SYBR Green PCR Master Mix (Life Technologies), respectively, following the manufacturer's instructions. Quantitative real-time PCR was performed with a Step-One-Plus PCR system (Applied Biosystems) by the 2−ΔΔCt method. The following primers were used: IL6 forward, 5′-TGCAATAACCACCCCTGACC-3′; reverse, 5′-CCCAGTGGACAGGTTTCT GA-3′; IL8 forward, 5′-TCCAAACCTTTCCACCCC-3′; reverse, 5′-CACAACCCTCTGCACCCA-3′; CCL2 forward, 5′-GAAGAATCACCAGCAGCAAGT-3′; reverse, 5′-TCCTGAACCCACTTCTGCTT-3′; Angiogenin forward, 5′-TGGGCGTTTTGTTGTTGGTCTTC-3′; reverse, 5′-CGTTTCTGAACCCCGCTGTGG-3′; Osteopontin(OPN) forward, 5′-ATCTCCTAGCCCCACAGAAT-3′; reverse, 5′-GTGGGTTTCAGCACTCTGGT-3′; GPR68 forward, 5′-CCTTCCGCTTCCACCAGTT-3′; reverse, 5′-TCGTCCTCGATGACCTCCT-3′; and β-actin forward, 5′-CACCATTGGCAATGAGCGGTTC-3′; reverse, 5′-AGGTCTTTGCGGATGTCCACGT-3′. β-actin was used to normalize RNA input for all RT-qPCR analyses.

Treatment with CnP and sorafenib in vivo

First, we evaluated the effect of CAFs in a xenograft model by comparing tumor volume rates of PLC alone and PLC plus CAF in vivo. We used female mice in this study, following previous study (15). Female NOD-SCID mice which were 7 weeks old (CLEA Japan) were injected 5 × 106 PLC cells and 5 × 106 PLC plus 1 × 106 CAF (CAF1) cells bilaterally and subcutaneously. Each group contained four xenografts. Moreover, we analyzed the CnP effects on PLC plus CAF xenograft in vivo. A week after injection the cells into mice, mice were divided into four groups (control, CnP, sorafenib, and CnP and sorafenib) randomly. Mice were treated with 1.0 μg/g CnP via subcutaneous injection twice weekly and 10 mg/kg sorafenib oral administration five times weekly. Each group had six xenografts. Tumor volumes and body weights of mice were measured twice weekly. The method to calculate tumor volumes was following formula: L/2 × S × S (L and S are the long and short diameters of the tumors). Protein levels of xenografts were measured immunohistochemically using tumors collected 5 days after the treatment started. We stained xenografts with hematoxylin and eosin staining and IHC and evaluated them microscopically. The serum biochemical tests of blood samples of mice were performed (Oriental Yeast Co) as evaluation of adverse events of the treatment. All in vivo experiments were conducted in compliance with the guidelines of our laboratory for animal experiments (approval number 18-024).

IHC

A paraffin-embedded block of xenografts was cut into 2-μm-thick sections and put on glass slides. Xylene was used for deparaffinization of each section and alcohol was used for dehydration. They were soaked in 0.3% H2O2/methanol for 30 minutes at room temperature to inhibit endogenous peroxidase. Next, they were soaked in heated water with 0.5% Immunosaver (Nisshin EM Co. Ltd.) at 98°C for 45 minutes. Nonspecific antigens were blocked by incubation with Protein Block Serum-Free (Dako) for 30 minutes at room temperature. The sections were then incubated with primary antibodies: anti–α-SMA mouse mAb (A2547; 1:1,000; Sigma-Aldrich, RRID:AB_476701), anti–caspase-3 antibody (No. 9662; 1:1,000; Cell Signaling Technology, RRID:AB_331439) and anti–Ki-67 antibody (M7240; 1:200; Dako, RRID:AB_2142367) for 24 hours at 4°C. After washing with PBS, they were incubated with Histofine Simple Stain MAX-PO (MULTI) kit (Nichirei Co.) for 45 minutes at room temperature. The chromogen 3, 3′-diaminobenzidine tetrahydrochloride was then applied as a 0.02% solution containing 0.005% H2O2 in 50 mmol/L ammonium acetate-citrate acid buffer (pH 6.0). Finally, the nucleus was counterstained with Mayer hematoxylin solution. A negative control was evaluated by replacing the primary antibodies with PBS in 0.1% BSA, and no staining was detected (Supplementary Fig. S1B). ImageJ (RRID:SCR_003070) version 1.51 was used for quantification of the positivity of α-SMA, Ki-67, and caspase-3. Masson Trichrome staining reagents (Muto Pure Chemical Ltd.) were used following the manufacturer's instructions.

Cap analysis of gene expression

CAFs were seeded in 10-cm dishes and cultured until they grew to their 90% confluency. The medium was changed to with or without 0.3 μg/mL CnP. After 6 hours, CAFs were collected and washed with PBS, snap frozen in nitrogen, and stored at −80°C. DNAFORM performed CAGE library preparation, sequencing, mapping, and gene expression and motif discovery analysis. Briefly, RNA quality was assessed using a Bioanalyzer (Agilent). First-strand cDNAs were transcribed to the 5′ ends of capped RNAs and attached to CAGE “bar code” tags. The sequenced CAGE tags were then mapped to mouse mm9 genomes using BWA (SCR_010910) software (v0.5.9) after discarding ribosomal or non-A/C/G/T base containing RNAs. Finally, the CAGE-tag 5′ coordinates were input for CAGEr clustering (16) using the Paraclu algorithm (17) with default parameters.

RNAi of GPR68

We used nontargeted control siRNA (ON-TARGETplus Non-targeting Pool; Dharmacon GE Healthcare) and GPR68-specific siRNA (MISSION esiRNA, EHU027261; Sigma). CAFs were suspended at a density of 5.0 × 105 cells in 100-μL Opti-MEM I Reduced Serum Media (Thermo Fisher Scientific) and then mixed with GPR68-specific siRNA or nontargeted control siRNA as a negative control. We performed transfection of siRNA with a T CUY21 EDIT II electroporator (BEX) with 175 V as a poring pulse and 10 V as atransfer pulses. The mRNA levels in CAFs with GPR68-specific siRNA transfection are expressed relative to those in CAFs with nontargeted control siRNA transfection. We checked mRNA and protein levels in CAFs 48 hours after transfection with siRNA. We collected GPR68-knockdown CAF-CM, as described above.

Statistical analysis

Data are presented as means ± SD. To compare defferences between two groups was analyzed with t tests. ANOVA with Tukey multiple comparison tests was used to evaluated to compare the differences among four groups. For the in vivo study, differences between groups were evaluated using repeated-measures ANOVA with Bonferroni post hoc tests. Results were considered statistically significant when the relevant P < 0.01 or 0.05. Data were analyzed using EZR.

Data availability

CAGE data in this study have been deposited to the Gene expression Omnibus and are available (accession number: GSE140530).

Proliferation and invasion of HCC cells were promoted by HCC-derived CAFs

To evaluated the CAFs effect on HCC progression, we performed proliferation and invasion assays with HCC cell lines incubated in CAF-CM. The proliferation and invasion of HCC cells were promoted by CAF-CM treatment (Fig. 1A and B). Furthermore, the volumes of tumors formed in vivo by HCC cells coimplanted with CAFs increased in size compared with tumors formed by HCC cells alone (Fig. 1C). Tumors with HCC cells plus CAFs had α-SMA staining and Masson Trichrome staining in the stroma (Fig. 1C).

Figure 1.

CAFs effects on HCC cell lines. A, CAF1- and CAF2-CM enhance both HepG2 and PLC cells proliferation in culture (n = 5). *, P < 0.01 (vs. control). B, CAF1- and CAF2-CM enhance both HepG2 and PLC cells invasion (n = 5). *, P < 0.01 (vs. control). (CAF1 and CAF2 were established from different patients with HCC. CAFs were cultured for 120 hours in DMEM, and CAF-CM was collected and used experiment without concentration. Cell proliferation and invasion were evaluated after 48 hours.) C, The tumor growth volume at 28 days after injecting PLC or PLC plus CAF cells. Tumor volume produced by implantation of PLC plus CAF was much greater than produced by PLC alone (n = 4). *, P < 0.05. Comparison typical photomicrographs of histologic examination between the PLC and PLC plus CAF cells. (HE, α-SMA, and Masson Trichrome staining, Original magnification ×200). Tumors from PLC plus CAF had α-SMA staining and Masson Trichrome staining in the stroma. CAF1 was used in this experiment. HE, hematoxylin and eosin.

Figure 1.

CAFs effects on HCC cell lines. A, CAF1- and CAF2-CM enhance both HepG2 and PLC cells proliferation in culture (n = 5). *, P < 0.01 (vs. control). B, CAF1- and CAF2-CM enhance both HepG2 and PLC cells invasion (n = 5). *, P < 0.01 (vs. control). (CAF1 and CAF2 were established from different patients with HCC. CAFs were cultured for 120 hours in DMEM, and CAF-CM was collected and used experiment without concentration. Cell proliferation and invasion were evaluated after 48 hours.) C, The tumor growth volume at 28 days after injecting PLC or PLC plus CAF cells. Tumor volume produced by implantation of PLC plus CAF was much greater than produced by PLC alone (n = 4). *, P < 0.05. Comparison typical photomicrographs of histologic examination between the PLC and PLC plus CAF cells. (HE, α-SMA, and Masson Trichrome staining, Original magnification ×200). Tumors from PLC plus CAF had α-SMA staining and Masson Trichrome staining in the stroma. CAF1 was used in this experiment. HE, hematoxylin and eosin.

Close modal

The proliferation and activation of CAFs was suppressed by CnP

The effects of CnP were examined by evaluating the proliferation of CAFs and HCC cells. Proliferation of CAFs were significantly suppressed in a concentration-dependent manner of CnP treatment (Fig. 2A). However, HCC cell lines proliferation were suppressed by only high concentration of CnP (1 μg/mL; Fig. 2B). The expression of α-SMA, a marker of CAF activation, in CAF1 and CAF2 cells were suppressed by CnP concentration dependently (Fig. 2C).

Figure 2.

CnP effects on CAFs and HCC cell lines. A, CnP suppressed CAFs growth in a concentration-dependent manner (n = 5). *, P < 0.01 (vs. control). B, CnP could not suppress the proliferation of PLC. And high concentrations alone (1 μg/mL) of CnP suppressed that of HepG2 (n = 5). *, P < 0.01 (vs. control). C, The CnP effects on α-SMA expression in CAFs were evaluated by Western blot analysis. CnP suppressed the α-SMA expression in CAFs. (Cell proliferation and α-SMA expression in CAFs were evaluated after 48 hours).

Figure 2.

CnP effects on CAFs and HCC cell lines. A, CnP suppressed CAFs growth in a concentration-dependent manner (n = 5). *, P < 0.01 (vs. control). B, CnP could not suppress the proliferation of PLC. And high concentrations alone (1 μg/mL) of CnP suppressed that of HepG2 (n = 5). *, P < 0.01 (vs. control). C, The CnP effects on α-SMA expression in CAFs were evaluated by Western blot analysis. CnP suppressed the α-SMA expression in CAFs. (Cell proliferation and α-SMA expression in CAFs were evaluated after 48 hours).

Close modal

The CAF-induced progression abilities of HCC cells were eliminated by CnP

To clalify the CnP effects on the malignant behavior of HCC cells promoted by CAFs, we performed proliferation assay and invasion assays of HCC cells treated with CnP-treated CAF-CMs. As mentioned above, direct treatment of low (0.3 μg/mL) CnP concentrations did not have significant effect on those of HCC cells (Fig. 3A and B), incubation in CAF-CM alone promoted both effects. Interestingly, CnP treatment eliminated the proliferation-promoting effects of CAF-CM (Fig. 3A), and CnP decreased the CAF-CM–enhanced invasive ability of HCC cells (Fig. 3B). The same result was obtained with Hep3B cells (Supplementary Fig. S2).

Figure 3.

The HCC-promoting effects of CAFs were inhibited by CnP. A, CAF-CM promoted both HepG2 and PLC proliferation. However, CnP-treated CAF-CM did not promote them (n = 5). *, P < 0.01. B, CAF-CM promoted both HCC cell lines invasion, but CnP-treated CAF-CM suppressed them (n = 5). *, P < 0.01. Conditioned media were collected from CAF1 cells (CAF-CM) or CAF1 cells with 0.3 μg/mL CnP (CnP-treated CAF-CM) cultured in DMEM for 120 hours. Cell proliferation and invasion were evaluated after 48 hours.

Figure 3.

The HCC-promoting effects of CAFs were inhibited by CnP. A, CAF-CM promoted both HepG2 and PLC proliferation. However, CnP-treated CAF-CM did not promote them (n = 5). *, P < 0.01. B, CAF-CM promoted both HCC cell lines invasion, but CnP-treated CAF-CM suppressed them (n = 5). *, P < 0.01. Conditioned media were collected from CAF1 cells (CAF-CM) or CAF1 cells with 0.3 μg/mL CnP (CnP-treated CAF-CM) cultured in DMEM for 120 hours. Cell proliferation and invasion were evaluated after 48 hours.

Close modal

CnP-suppressed cytokine production from CAFs

To clarify the differences between control CAF- and CnP-treated CAF-CM, we performed cytokine array analysis. IL6, IL8, CCL2, angiogenin, and OPN levels were markedly decreased in CnP-treated CAF-CM compared with those in CAF-CM (Fig. 4A). These cytokines were confirmed to be also suppressed at the mRNA and protein levels using qRT-PCR and ELISA-based analysis (Fig. 4B; Supplementary Fig. S3).

Figure 4.

The cytokines production of CAFs were suppressed by CnP. A, Evaluation of cytokines of CAF-CMs (CAF- and CnP-treated CAF-CM) were performed with cytokine array. IL6, IL8, CCL2, angiogenin, and OPN molecules were reduced in cells cultured in CnP-treated CAF-CM. The density ratio comparison of dots was shown on right. Conditioned media were collected from CAF1 cells (CAF-CM) or CAF1 cells with CnP (0.3 μg/mL; CnP-treated CAF-CM) cultured in DMEM for 120 hours. B, mRNA rate of the cytokines in CAF (control) and CnP-treated CAF (CnP). (n = 3). *, P < 0.01 (vs. control).

Figure 4.

The cytokines production of CAFs were suppressed by CnP. A, Evaluation of cytokines of CAF-CMs (CAF- and CnP-treated CAF-CM) were performed with cytokine array. IL6, IL8, CCL2, angiogenin, and OPN molecules were reduced in cells cultured in CnP-treated CAF-CM. The density ratio comparison of dots was shown on right. Conditioned media were collected from CAF1 cells (CAF-CM) or CAF1 cells with CnP (0.3 μg/mL; CnP-treated CAF-CM) cultured in DMEM for 120 hours. B, mRNA rate of the cytokines in CAF (control) and CnP-treated CAF (CnP). (n = 3). *, P < 0.01 (vs. control).

Close modal

CnP-suppressed GPR68, cytokines regulator protein, and expression of CAFs

Genes of CAF that are strongly upregulated and downregulated by CnP were identified (Supplementary Fig. S4). Twenty cancer-promoting genes in CAFs strongly enhance the viability of cocultured cancer cells (18). However, whether CnP treatment can regulate these genes in HCC CAFs is not known. Therefore, we evaluated the expression of these 20 genes in CAFs using CAGE analysis. Notably, the expression of GPR68 was suppressed the most in CnP-treated CAFs relative to that in control CAFs (Fig. 5A). In addition, we validated that CnP treatment suppressed the expression of GPR68 in CAFs (Fig. 5B) and investigated the proliferative capabilities of HCC cells using CM derived from GPR68-suppressed CAFs (siGPR68 CAF-CM; Fig. 5C). Enhanced proliferation induced by CAF-CM was eliminated by GPR68 knockdown in CAFs (Fig. 5D). Furthermore, we confirmed using RT-PCR that the mRNA levels of IL6, IL8, angiogenin, and OPN were decreased but that CCL2 was not (Fig. 5E). The significant IL6 and IL8 suppression in GPR68-knockdown CAF-CM were validated with ELISA (Supplementary Fig. S5). Furthermore, we evaluated GPR68 expression levels of HCC cells and CAFs. GPR68 expression levels of HCC cells were higher than those in CAFs (Supplementary Fig. S6A). CnP had no effect on the expression of GPR68 in HCC cells (Supplementary Fig. S6B).

Figure 5.

CnP inhibits cytokines production via suppression of GPR68 in CAFs. A, The fold change of mRNA of 20 cancer-promoting genes in CAF treated by CnP (0.3 μg/mL) was analyzed by CAGE analysis. CAF1 was used in this experiment. B, GPR68 expression in CAFs treated by CnP (0.3 μg/mL) was evaluated by Western blotting. C, mRNA and protein levels of GPR68 in CAFs and CAFs by transfection with GPR68-specific siRNA. D, CAF-CM promoted the proliferation of HepG2 and PLC cells in culture. However, knockdown of GPR68 in CAF significantly weakened this promotion. (n = 5). *, P < 0.01. (CAF-CM, CAF1 cells transfected with nontargeted control siRNA, and siGPR68 CAF-CM, CAF1 cells transfected with GPR68-specific siRNA were cultured in DMEM for 120 hours and conditioned media were collected. Proliferation of cells was evaluated after 48 hours.) E, Expression of mRNA of the cytokines in CAF (negative) and GPR68-knockdown CAF (siGPR68; n = 3). *, P < 0.01.

Figure 5.

CnP inhibits cytokines production via suppression of GPR68 in CAFs. A, The fold change of mRNA of 20 cancer-promoting genes in CAF treated by CnP (0.3 μg/mL) was analyzed by CAGE analysis. CAF1 was used in this experiment. B, GPR68 expression in CAFs treated by CnP (0.3 μg/mL) was evaluated by Western blotting. C, mRNA and protein levels of GPR68 in CAFs and CAFs by transfection with GPR68-specific siRNA. D, CAF-CM promoted the proliferation of HepG2 and PLC cells in culture. However, knockdown of GPR68 in CAF significantly weakened this promotion. (n = 5). *, P < 0.01. (CAF-CM, CAF1 cells transfected with nontargeted control siRNA, and siGPR68 CAF-CM, CAF1 cells transfected with GPR68-specific siRNA were cultured in DMEM for 120 hours and conditioned media were collected. Proliferation of cells was evaluated after 48 hours.) E, Expression of mRNA of the cytokines in CAF (negative) and GPR68-knockdown CAF (siGPR68; n = 3). *, P < 0.01.

Close modal

CnP inhibited tumor proliferation and combined therapy with sorafenib markedly inhibited tumor proliferation in vivo

We investigated the combination therapy with CnP and the standard HCC treatment drug, sorafenib in vivo. Single treatment with CnP or sorafenib inhibited tumor growth, but treatment with CnP and sorafenib combined inhibited tumor growth to a greater extent (Fig. 6A). The tumors treated with this combination showed a reduction in Ki-67–positive HCC cells and α-SMA staining areas, along with an increase in caspase-3–positive HCC cells (Fig. 6B). We evaluated body weight and organ toxicity related to the treatment in this study, but there were no differences between the treatment groups and controls (Supplementary Fig. S7A and S7B).

Figure 6.

Combination therapy with CnP plus sorafenib reduces tumor volume induced by injection of PLC plus CAF cells, markedly. A, Combination therapy with CnP plus sorafenib suppresses tumor growth of PLC plus CAFs the most, as indicated by tumor growth curves. Photograph showed tumors from the four groups [control; CnP, 1.0 μg/body weight (g) CnP via subcutaneous injection; sorafenib, 10 mg/kg sorafenib orally; CnP + sorafenib. (n = 6). *, P < 0.01. CAF1 was used in this experiment.] B, Protein levels in xenograft were assayed with tumor tissues collected 5 days after treatment started. Typical photomicrographs of histology comparing four groups (HE, α-SMA, Ki-67, and caspase-3; Original magnification ×200). CnP plus sorafenib treatment induced a reduction in α-SMA staining in the stroma and the rate of Ki-67–positive HCC cells, and an increase of caspase-3–positive HCC cells. (n = 5). *, P < 0.01.

Figure 6.

Combination therapy with CnP plus sorafenib reduces tumor volume induced by injection of PLC plus CAF cells, markedly. A, Combination therapy with CnP plus sorafenib suppresses tumor growth of PLC plus CAFs the most, as indicated by tumor growth curves. Photograph showed tumors from the four groups [control; CnP, 1.0 μg/body weight (g) CnP via subcutaneous injection; sorafenib, 10 mg/kg sorafenib orally; CnP + sorafenib. (n = 6). *, P < 0.01. CAF1 was used in this experiment.] B, Protein levels in xenograft were assayed with tumor tissues collected 5 days after treatment started. Typical photomicrographs of histology comparing four groups (HE, α-SMA, Ki-67, and caspase-3; Original magnification ×200). CnP plus sorafenib treatment induced a reduction in α-SMA staining in the stroma and the rate of Ki-67–positive HCC cells, and an increase of caspase-3–positive HCC cells. (n = 5). *, P < 0.01.

Close modal

We showed that CAFs promote HCC malignancy and CnP suppressed this interaction. We also demonstrated that CnP treatment strongly suppressed various cytokines such as IL6, IL8, CCL2, angiogenin, and OPN producted from CAFs. In vivo experiments showed that combined therapy with CnP and sorafenib inhibited tumor growth to a greater extent than single-agent treatment. In addition, our CAGE analysis and in vitro data indicated that CnP suppressed the expression of GPR68, which functions to promote cytokine production in CAFs. This study is the first report to show the significance of this new therapeutic compound for targeting CAF involvement in HCC.

A low concentration (0.3 μg/mL) of CnP did not suppress proliferation of HCC cells (Fig. 2B). It was suggested that CnP was more effective against CAFs than cancer cells because at low concentrations, it could specifically suppress CAFs in in vitro experiments (Figs. 2C, 4A, and B). However, sorafenib is effective against HCC and is widely used in clinical practice (19). In our in vivo study, the concurrent administration of CnP and sorafenib for targeting CAFs and HCC cells, respectively, strongly inhibited both stromal α-SMA staining in CAFs and the proliferation ability of HCC cells in xenograft tumors (Fig. 6A and B). No significant difference between the control and CnP-treated group was observed; however, xenografts in the CnP-treated group were a little smaller than those in the control group (Fig. 6A). Moreover, the expression of α-SMA, a marker of activated CAFs, in CnP-treated animals was suppressed. Therefore, we surmised that CnP suppressed the cancer-promoting effect via regulation of CAF activity. Combination therapy with CnP and existing anticancer agents, such as sorafenib, may be effective for HCC in patients with activated CAFs. The significance of combined therapy is anticipated to be verified in clinical trials. No apparent side effects were observed in the experimental mice used (Supplementary Fig. S7).

We demonstrated that CnP suppressed the production of multiple cytokines, particularly IL6, IL8, CCL2, angiogenin, and OPN. They were reported to play key roles in tumor progression. For example, IL6 regulates JAK-STAT3 signaling and the mammalian target of rapamycin signaling. These pathways promote the proliferation, invasion, and immunosuppression and invasion of tumor cells (3, 20). Furthermore, Zhao and colleagues showed that activated CAF effects on cancer cells were suppressed by an IL6-neutralizing antibody (3). IL8 is thought to promote epithelial-to-mesenchymal transition (EMT) of HCC cells (21). CCL2 is reported to activate Hedgehog signaling in HCC cells and suppress T-cell antitumor immune responses (22, 23). Angiogenin is believed to promote neovascularization of HCC and EMT of HCC cells (24, 25). OPN may promote migration and metastasis of HCC cells (26, 27). CnP might inhibit the HCC-promoting effects of CAFs via suppression of cytokine production and release.

In this study, we showed that CnP regulates IL6, IL8, CCL-2, angiogenin, and OPN produced by CAFs. These cytokines, except for angiogenin, were found to be regulated by GPR68 expression in CAFs. Previously, GPR68 was reported to regulate IL6 expression in CAFs in pancreatic cancer cells and IL8 in β cells in the pancreas (9, 28). Production of multiple cytokines, including IL6, IL8, angiogenin, and OPN, in our study, was weakened by GPR68 knockdown in CAFs. CnP downregulated CCL-2, but this factor is not regulated by GPR68. We hypothesized that CnP might regulate several cytokines via GPR68 and also CCL-2, which are not dependent on GPR68. However, the cancer-promoting activity of CAFs was weakened by the knockdown of GPR68 by siRNA. GPR68 is important in regulating the production of cytokines in CAFs. CAFs mainly promote HCC by secretion of IL6, IL8, angiogenin, and OPN, as induced by GPR68.

GPR68 expression is induced by activation of TNFα/NFκB signaling (9). We evaluated NFκB signaling after CnP treatment in CAF cells. However, the NFκB signaling was not altered by CnP treatment (Supplementary Fig. S8A). Moreover, GPR68 did not regulate α-SMA of CAF (Supplementary Fig. S8B). Further studies are necessary to clarify the effects of CnP on GPR68 regulation.

In conclusion, CnP was demonstrated to inhibit the HCC-promoting effects of CAFs through suppressing activation and cancer-promoting cytokines production of CAFs. The combination of a multikinase inhibitor, sorafenib, and CnP against HCC and CAFs, respectively, completely suppressed tumor growth in our in vivo analysis. Therefore, combined therapy with CnP and existing anticancer agents may be a promising therapeutic strategy in overcoming refractory HCC with activated CAFs. Moreover, we showed that GPR68 in CAFs is important in regulating the secretion of cytokines from CAFs. Therefore, CnP-based alteration of GPR68 in CAFs may be one of the fundamental mechanisms of preventing HCC promotion.

No disclosures were reported.

T. Yamanaka: Formal analysis, validation, investigation, visualization, writing–original draft, project administration, writing–review and editing. N. Harimoto: Project administration. T. Yokobori: Writing–original draft. R. Muranushi: Validation, investigation. K. Hoshino: Validation, investigation. K. Hagiwara: Validation, investigation. D. Gantumur: Validation, investigation. T. Handa: Validation, investigation. N. Ishii: Supervision. M. Tsukagoshi: Supervision. T. Igarashi: Supervision. A. Watanabe: Supervision. N. Kubo: Supervision. K. Araki: Supervision. K. Umezawa: Resources, investigation. K. Shirabe: Supervision, funding acquisition, project administration.

The authors would like to thank Bayer HealthCare Pharmaceuticals Inc. for providing sorafenib. The authors would like to thank Enago (www.enago.com) for the English language review.

This study was supported by Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (JP18H02877 and JP18K16299) and Research Program on Hepatitis from Japan Agency for Medical Research and development, AMED.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Bray
F
,
Ferlay
J
,
Soerjomataram
I
,
Siegel
RL
,
Torre
LA
,
Jemal
A
. 
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2018
;
68
:
394
424
.
2.
Zheng
X
,
Xu
M
,
Yao
B
,
Wang
C
,
Jia
Y
,
Liu
Q
. 
IL-6/STAT3 axis initiated CAFs via up-regulating TIMP-1 which was attenuated by acetylation of STAT3 induced by PCAF in HCC microenvironment
.
Cell Signal
2016
;
28
:
1314
24
.
3.
Zhao
Z
,
Xiong
S
,
Wang
R
,
Li
Y
,
Wang
X
,
Wang
Y
, et al
Peri-tumor fibroblasts promote tumorigenesis and metastasis of hepatocellular carcinoma via Interleukin6/STAT3 signaling pathway
.
Cancer Manag Res
2019
;
11
:
2889
901
.
4.
Zhu
YJ
,
Zheng
B
,
Wang
HY
,
Chen
L
. 
New knowledge of the mechanisms of sorafenib resistance in liver cancer
.
Acta Pharmacol Sin
2017
;
38
:
614
22
.
5.
Kubo
N
,
Araki
K
,
Kuwano
H
,
Shirabe
K
. 
Cancer-associated fibroblasts in hepatocellular carcinoma
.
World J Gastroenterol
2016
;
22
:
6841
50
.
6.
Lau
EY
,
Lo
J
,
Cheng
BY
,
Ma
MK
,
Lee
JM
,
Ng
JK
, et al
Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-Met/FRA1/HEY1 signaling
.
Cell Rep
2016
;
15
:
1175
89
.
7.
Khawar
IA
,
Park
JK
,
Jung
ES
,
Lee
MA
,
Chang
S
,
Kuh
HJ
. 
Three dimensional mixed-cell spheroids mimic stroma-mediated chemoresistance and invasive migration in hepatocellular carcinoma
.
Neoplasia
2018
;
20
:
800
12
.
8.
Stanley
AC
,
Lacy
P
. 
Pathways for cytokine secretion
.
Physiology
2010
;
25
:
218
29
.
9.
Wiley
SZ
,
Sriram
K
,
Liang
W
,
Chang
SE
,
French
R
,
McCann
T
, et al
GPR68, a proton-sensing GPCR, mediates interaction of cancer-associated fibroblasts and cancer cells
.
FASEB J
2018
;
32
:
1170
83
.
10.
Saito
R
,
Yamada
S
,
Yamamoto
Y
,
Kodera
T
,
Hara
A
,
Tanaka
Y
, et al
Conophylline suppresses pancreatic stellate cells and improves islet fibrosis in Goto-Kakizaki rats
.
Endocrinology
2012
;
153
:
621
30
.
11.
Kubo
N
,
Saito
R
,
Hamano
K
,
Nagasawa
M
,
Aoki
F
,
Takei
I
, et al
Conophylline suppresses hepatic stellate cells and attenuates thioacetamide-induced liver fibrosis in rats
.
Liver Int
2014
;
34
:
1057
67
.
12.
Ishii
N
,
Araki
K
,
Yokobori
T
,
Hagiwara
K
,
Gantumur
D
,
Yamanaka
T
, et al
Conophylline suppresses pancreatic cancer desmoplasia and cancer-promoting cytokines produced by cancer-associated fibroblasts
.
Cancer Sci
2019
;
110
:
334
44
.
13.
Umezawa
K
,
Ohse
T
,
Yamamoto
T
,
Koyano
T
,
Takahashi
Y
. 
Isolation of a new vinca alkaloid from the leaves of Ervatamia microphylla as an inhibitor of ras functions
.
Anticancer Res
1994
;
14
:
2413
7
.
14.
Fujii
M
,
Takei
I
,
Umezawa
K
. 
Antidiabetic effect of orally administered conophylline-containing plant extract on streptozotocin-treated and Goto-Kakizaki rats
.
Biomed Pharmacother
2009
;
63
:
710
6
.
15.
Matsuki
M
,
Hoshi
T
,
Yamamoto
Y
,
Ikemori-Kawada
M
,
Minoshima
Y
,
Funahashi
Y
, et al
Lenvatinib inhibits angiogenesis and tumor fibroblast growth factor signaling pathways in human hepatocellular carcinoma models
.
Cancer Med
2018
;
7
:
2641
53
.
16.
Haberle
V
,
Forrest
AR
,
Hayashizaki
Y
,
Carninci
P
,
Lenhard
B
. 
CAGEr: precise TSS data retrieval and high-resolution promoterome mining for integrative analyses
.
Nucleic Acids Res
2015
;
43
:
e51
.
17.
Frith
MC
,
Saunders
NF
,
Kobe
B
,
Bailey
TL
. 
Discovering sequence motifs with arbitrary insertions and deletions
.
PLoS Comput Biol
2008
;
4
:
e1000071
.
18.
Horman
SR
,
To
J
,
Lamb
J
,
Zoll
JH
,
Leonetti
N
,
Tu
B
, et al
Functional profiling of microtumors to identify cancer associated fibroblast-derived drug targets
.
Oncotarget
2017
;
8
:
99913
30
.
19.
Llovet
JM
,
Ricci
S
,
Mazzaferro
V
,
Hilgard
P
,
Gane
E
,
Blanc
JF
, et al
Sorafenib in advanced hepatocellular carcinoma
.
N Engl J Med
2008
;
359
:
378
90
.
20.
Liu
F
,
Zhang
W
,
Yang
F
,
Feng
T
,
Zhou
M
,
Yu
Y
, et al
Interleukin-6-stimulated progranulin expression contributes to the malignancy of hepatocellular carcinoma cells by activating mTOR signaling
.
Sci Rep
2016
;
6
:
21260
.
21.
Fu
XT
,
Dai
Z
,
Song
K
,
Zhang
ZJ
,
Zhou
ZJ
,
Zhou
SL
, et al
Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway
.
Int J Oncol
2015
;
46
:
587
96
.
22.
Zhuang
H
,
Cao
G
,
Kou
C
,
Liu
T
. 
CCL2/CCR2 axis induces hepatocellular carcinoma invasion and epithelial-mesenchymal transition in vitro through activation of the Hedgehog pathway
.
Oncol Rep
2018
;
39
:
21
30
.
23.
Li
X
,
Yao
W
,
Yuan
Y
,
Chen
P
,
Li
B
,
Li
J
, et al
Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma
.
Gut
2017
;
66
:
157
67
.
24.
Hisai
H
,
Kato
J
,
Kobune
M
,
Murakami
T
,
Miyanishi
K
,
Takahashi
M
, et al
Increased expression of angiogenin in hepatocellular carcinoma in correlation with tumor vascularity
.
Clin Cancer Res
2003
;
9
:
4852
9
.
25.
Hengjuan
LV
,
Liu
G
,
Kun
LI
,
Mingqiu
LI
,
Zhang
D
. 
Angiogenin regulates epithelial-mesenchymal transition of hepatocellular carcinoma through upregulation of HMGA2
.
Pharmazie
2019
;
74
:
301
4
.
26.
Song
J
,
Ge
Z
,
Yang
X
,
Luo
Q
,
Wang
C
,
You
H
, et al
Hepatic stellate cells activated by acidic tumor microenvironment promote the metastasis of hepatocellular carcinoma via osteopontin
.
Cancer Lett
2015
;
356
:
713
20
.
27.
Yu
X
,
Zheng
Y
,
Zhu
X
,
Gao
X
,
Wang
C
,
Sheng
Y
, et al
Osteopontin promotes hepatocellular carcinoma progression via the PI3K/AKT/Twist signaling pathway
.
Oncol Lett
2018
;
16
:
5299
308
.
28.
Chandra
V
,
Karamitri
A
,
Richards
P
,
Cormier
F
,
Ramond
C
,
Jockers
R
, et al
Extracellular acidification stimulates GPR68 mediated IL-8 production in human pancreatic beta cells
.
Sci Rep
2016
;
6
:
25765
.