Skip to main content
Erschienen in: Cancer Cell International 1/2018

Open Access 01.12.2018 | Primary research

M3 muscarinic acetylcholine receptors regulate epithelial–mesenchymal transition, perineural invasion, and migration/metastasis in cholangiocarcinoma through the AKT pathway

verfasst von: Yujie Feng, Xiao Hu, Guangwei Liu, Lianfang Lu, Wei Zhao, Fangzhen Shen, Kai Ma, Chuandong Sun, Chengzhan Zhu, Bingyuan Zhang

Erschienen in: Cancer Cell International | Ausgabe 1/2018

Abstract

Background

Cholangiocarcinoma is a highly malignant tumor type that is not sensitive to radiotherapy or chemotherapy due to aggressive perineural invasion and metastasis. Unfortunately, the mechanisms underlying these processes and the signaling factors involved are largely unknown. In this study, we analyzed the role of M3 muscarinic acetylcholine receptors (M3-mAChR) in cell migration, perineural invasion, and metastasis during cholangiocarcinoma.

Methods

We assessed 60 human cholangiocarcinoma tissue samples and 30 normal biliary tissues. Immunohistochemical staining was used to detect M3-mAChR expression and the relationship between expression and clinical prognosis was evaluated. The biological functions of M3-mAChR in cholangiocarcinoma cell migration, perineural invasion, and epithelial–mesenchymal transition (EMT) were investigated using the human cholangiocarcinoma cell lines FRH0201 and RBE in conjunction with various techniques, including agonist/antagonist treatment, RNA interference, M3-mAChR overexpression, dorsal root ganglion co-culturing, immunohistochemistry, western blotting, etc.

Results

M3-mAChR were highly expressed in cholangiocarcinoma tissue and expression was closely related to differentiation and lymphatic metastasis, affecting patient survival. Treatment with the M3-mAChR agonist pilocarpine and M3-mAChR overexpression significantly promoted migration and perineural invasion, while the M3-mAChR antagonist atropine blocked these effects. Similarly, M3-mAChR knock-down also weakened cell migration and perineural invasion. The expression of phosphatase and tensin homolog, AKT, E-cadherin, vimentin, and Snail, which are components of the phosphatidylinositol 3-kinase/AKT signaling pathway and EMT, were altered by pilocarpine, and these effects were again blocked by atropine. Notably, AKT knock-down decreased M3-mAChR expression and reversed the downstream effects of this receptor.

Conclusions

M3-mAChR are involved in tumor cell migration, perineural invasion, and EMT during cholangiocarcinoma, and these effects are modulated via the AKT signaling pathway.
Abkürzungen
M3 mAchRs
M3 muscarinic acetylcholine receptors
EMT
epithelial–mesenchymal transition
PNI
perineural invasion
PTEN
phosphatase and tensin homolog
AKT
protein kinase B
shRNA
small hairpin RNA
DRG
dorsal root ganglio
PILO
pilocarpine
ATR
atropine
PI3K
phosphatidylinositol-3-kinase

Background

Cholangiocarcinoma, which originates from biliary epithelial cells, is a type of malignant tumor that is characterized by low diagnostic rates and high fatality rates [1]. Concealed pathogenesis, lack of early diagnostic markers, and low sensitivity to conventional radiotherapy and chemotherapy treatments are the crucial reasons underlying the poor prognosis of cholangiocarcinoma patients. Currently, surgical resection is the only effective treatment method for this disease. Moreover, recent studies have found that perineural invasion is a common biological characteristic of cholangiocarcinoma and is closely related to postoperative recurrence rates and prognoses [2].
Cholangiocarcinoma undergoes perineural invasion early in development and involves tumor cells surrounding the nerve fibers, entering the perineurium, and spreading via local infiltration and metastasis. Recent studies have shown that the biliary system is rich in autonomic nerves, and biliary tract malignancy occurs near the celiac plexus; thus, tumor cells are more likely to invade the surrounding nerve plexus, causing perineural invasion [3]. Another study reconstructed the perineural invasion and metastasis processes that occur during cholangiocarcinoma by computer-assisted three-dimensional (3D) analysis and formed 3D images of cholangiocarcinoma to display existing small vessels, lymph vessels, and nerve fibers around the tumor [4]. They concluded that tumor cells survive independently in spaces around the nerves, including in the small vessels and small lymph vessels. Accordingly, perineural invasion was found to be the fifth self-existent route of metastasis following direct invasion and metastasis, implantation metastasis, lymphatic metastasis, and blood metastasis of abdominal tumors. Metastasis occurs in the nerve fibers via jumping, meaning that cholangiocarcinoma can either invade and metastasize to the liver through the perineural space or metastasize to the retroperitoneal ganglions. This is also why cholangiocarcinoma is difficult to cure.
As an important digestive organ, the biliary system regulates biliary secretion and excretion and is surrounded by vagus nerves, which can interact with the liver vascular system and inevitably affect the pathophysiology of the biliary epithelium [5]. However, while these systems interact and affect the health and function of each other, for the cancer cells to spread from the primary site, they must develop migratory and invasive properties. Epithelial-to-mesenchymal transition (EMT) is a phenomenon in which epithelial cells lose their original polarity and transform into Leydig cells. EMT is comprised of a series of intricate biological and biochemical changes that cause the cell to lose its differentiated epithelial-like state and gain a more mesenchymal-like phenotype. Moreover, the EMT is closely related to tumorigenesis, local invasion, distant metastasis, and tumor resistance [69] and appears to play a distinct role in cholangiocarcinoma [10]. The various processes involved in cholangiocarcinoma, including perineural invasion, metastasis, and EMT, likely require complex signaling pathway function as well as a host of independent factors, many of which have not been elucidated.
Muscarinic acetylcholine receptors (mAChRs) are members of the G protein-coupled receptor family and include five receptor subtypes (M1, M2, M3, M4, and M5) [11]. The M3-mAChR are mainly distributed in the digestive tract glands and vascular smooth muscles and modulate glandular secretion and smooth muscle relaxation. Various studies have also shown that M3-mAChR play an important role in the proliferation and invasion of cancer cells [1214]. However, the role of M3-mAChR in cholangiocarcinoma has not been evaluated.
In this study, we investigated the function of M3-mAChR during perineural invasion, metastasis, and EMT in cholangiocarcinoma. To our knowledge, this is the first time M3-mAChR-mediated regulation has been explored in cholangiocarcinoma.

Methods

Patients and tissue specimens

We collected paraffin-embedded biliary pathology tissues obtained during surgical resection at the Affiliated Hospital of Qingdao University from December 2008 to December 2013. The samples included 60 cholangiocarcinoma tissues and 30 normal biliary tissues (from patients with choledochocysts and liver transplantation). Patient data was also collected. None of the patients had undergone radiotherapy or chemotherapy/drugs treatment before the operation. All procedures in this study that involved human participants were performed in accordance with the 1964 Helsinki declaration and its later amendments. The protocol was approved by the Ethics Committee of The Affiliated Hospital of Qingdao University. Informed consent was obtained from all individual participants included in the study.

Immunohistochemistry

The paraffin-embedded biopsy tissue specimens were cut into 4-μm-thick sections and deparaffinized. The sections were pretreated and incubated with primary anti-M3 mAchR antibodies (1:1000; Abcam, Cambridge, UK) overnight at 4 °C according to the manufacturer’s instructions. Then, the sections were incubated with secondary antibodies for 30 min, stained with diaminobenzidine, and finally, counterstained with hematoxylin. The stained sections were then imaged (400×; OLYMPUS, Tokyo, Japan). Positive staining in the basement membrane was manifested as a brownish-yellow color.
For statistical analysis, 10 random fields of the tumor were selected in each section, and 100 cells from each field were counted. The samples were then graded according to the proportion of positive cells to the total number of cells. When the percentage of positively stained cholangiocarcinoma cells was less than 25%, the result was considered negative. Alternatively, when the percentage was between 25 and 55% or more than 55%, the staining was described as positive and strongly positive, respectively.

Cell lines, culture conditions and reagents

The cholangiocarcinoma cell lines FRH0201 and RBE were purchased from the Shanghai Cell Bank of the Chinese Academy of Sciences. The cell lines were all authenticated. The cells were grown and cultivated in RPMI-1640 medium supplemented with 10% newborn calf serum (Thermo Scientific Hyclone, USA) in an incubator containing 5% CO2 at 37 °C. M3-mAChR agonist pilocarpine and M3-mAChR antagonist atropine were purchased from Sigma-Aldrich (Taufkirchen, Germany).

Constructing the lentivirus vector for RNAi and receptor overexpression

The most effective M3-targeting small hairpin RNA (shRNA) sequences (5′-GCAGTGACAGTTGGAACAACA-3′) were transfected into the cultured cells using Lipofectamine 2000 Reagent (Invitrogen, Carlsbad, CA, USA). M3-NC (5′-TTCTCCGAACGTGTCACGT-3′) was used to generate the negative (scrambled) control lentiviral vectors. Lentivirus was added to the cells according to the manufacturer’s recommended protocol.
The high-copy plasmid pcDNA 3 1(+) (Invitrogen, Carlsbad, CA, USA) harbouring the complete cDNA of the human mAchR3 was performed according to the manufacturer’s protocol using a commercially available kit (Qiagen, Hilden, Germany).

RNAi treatment

FRH0201-M3 cells were transfected with siAKT (5′-TTGTACGCAGAGAGAATAACT-3′), using the Lipofectamine transfection reagent (Invitrogen) for 48 h. Retrovirus packaging and transfection were conducted according to the manufacturer’s instructions.

Quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA was extracted from cultured cells using TRIzol reagent in a one-step method according to the manufacturer’s instructions. Primer design was performed using Primer5 software with sequences for human M3-mAChR obtained from GenBank. The following primers were used in this study:M3-mAChR (forward, 5-CACAATAACAGTACAACCTCGCC-3 and reverse, 5-GCCAGGATGCCCGTTAAGAAA-3). internal control GAPDH (forward, 5-GCACCGTCAAGGCTGAGAAC-3 and reverse, 5-TGGTGAAGACGCCAGTGGAT-3. M3-mAChR gene expression was quantitated using the 2−∆∆Ct method. The data are presented as the fold change compared to the specified controls using an average result of three independent experiments. Differences were considered biologically relevant when the fold change was > 2.0 or < 0.5 [15].

Western blot analysis

Whole cell lysates were prepared using RIPA lysis buffer and protease inhibitor (keyGEN, Nanjing, China), and the protein concentrations were determined using a BCA kit (Thermo Scientific™). A total of 50 μg of protein was loaded into each well of a sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel, and the proteins were separated. After the proteins were transferred to a polyvinylidene difluoride membrane, the membranes were blocked with 5% nonfat dried milk for 2 h. Next, the membranes were incubated overnight with primary antibodies targeting M3-mAChR, phosphatase and tensin homolog (PTEN), AKT, E-cadherin, vimentin, and Snail (all purchased from Santa Cruz Biotechnology, Santa Cruz, CA, USA) at 4 °C. The membranes were then incubated with secondary antibodies for 2 h at room temperature. Finally, the protein bands were visualized using enhanced chemiluminescence, and their intensity was quantified.

Migration assay

For the transwell migration assays, 105 cells were seeded in 100 μL of serum-free Dulbecco’s modified Eagle’s medium in fibronectin-coated polycarbonate membrane inserts in a transwell apparatus. The diameter of the membrane micropores was 8 μm. A total of 600 μL of RPMI-1640 culture medium with 10% serum was added to the lower wells. After culturing cells for 36 h at 37 °C, the transwell chambers were removed and washed with phosphate-buffered saline three times. Cells that had not invaded were removed using aseptic cotton buds, and the chambers were then fixed in 4% formalin solution for 20 min. Next, cells on the chambers were stained with 0.1% crystal violet for 15 min and then imaged under an optical microscope. Cells in 10 fields were counted (400× magnification). The experiment was performed in triplicate.

Dorsal root ganglion isolation and ex vivo culture

As described in a previous study [16], newborn Kunming mice (24 h old) were disinfected using 75% ethyl alcohol, sacrificed by cervical dislocation, and fixed in the prone position. The mediodorsal skin was cut using sterile eye scissors to expose the muscles of the back. Taper eye scissors were then used to cut an opening and expose the spinal cord, which was excised and washed with normal saline several times, until the dorsal root ganglion (DRG) could be observed. Next, the DRG was carefully removed using smooth microforceps and washed once in normal saline. The washed DRG was then transferred into a 24-well plate, and thawed Matrigel (BD Biosciences, Franklin Lake, NJ, USA) was added to completely cover it. The Matrigel-coated DRG was then incubated at 37 °C for 30 min until coagulation. Notably, all of these procedures involving the care and use of laboratory animals were performed in accordance with the principles and standards set forth in the Principles for Use of Animals (National Guide for Grants and Contracts).

DRG immunohistochemical staining

Nissl staining to observe the state of nerve cell viability

Remove the neurons from which the culture solution was discarded, and wash it with 0.01 mol/L PBS three times for 5 min each time. Then 4% paraformaldehyde was fixed at room temperature for 1 h, and washed with 0.01 mol/L PBS for 3 times for 5 min each time followed by 0.1% toluidine blue dyeing at 37 °C for 10 min. After the final 95% ethanol separation, the cells were dehydrated in 50%–100% gradient ethanol for 3–5 min each time. The xylene was transparent and the gel was sealed, and then the neuron viability was observed under a microscope.

Neurofilament protein (NF) immunocytochemical staining of DRG

Co-infiltrate and purify the cultured DRG, discard the culture solution and wash it with 0.0 l mol/L PBS for 3 times for 5 min each time; 4% paraformaldehyde was fixed at room temperature for 1 h, washed with PBS; 0.3% H2O2 methanol was added., placed for 10 min to remove endogenous catalase, washed with PBS and moved to a wet box. Add 10% sheep serum to the coverslip, add 3 drops per well, and incubate in a 37 °C oven for 30 min; after aspirating the blocking solution, l:200 rabbit anti-mouse NF antibody (Beyotime, Shanghai, China) 3 drops per tablet, at 4 °C refrigerator save. Immediately after the PBS cleaning, add goat anti-rabbit IgG (1:200 biotinylated, Beyotime, Shanghai, China) on the coverslip, 3 drops per well, incubate in a 37 °C oven, and wash with PBS after 30 min. Add the ready-to-use ABC solution onto the coverslip and incubate in a 37 °C oven for 30 min; wash with PBS, add DAB coloring solution for 8–10 min, then rinse again with PBS. Finally, dehydrated in 50% concentration gradient ethanol, about 5 min each time, xylene was transparent and the gel was sealed, and then the axon extension of each group was observed under a microscope.

Co-culturing of DRG segments and RBE cells

For co-culturing, untreated RBE cells in the logarithmic growth phase were added to each well of the 24-well plate containing a Matrigel-coated DRG at a concentration of 2.5 × 104 cells/well in 500 μL of medium (RPMI-1640 containing serum). The DRGs were treated as follows: MOCK (medium + RBE cells), ShM3 (medium + ShM3-treated RBE cells), PILO [medium + RBE cells + pilocarpine (1.0 mM)]; ATR [medium + RBE cells + atropine (0.1 mM)], and PILO + ATR [medium + RBE cells + pilocarpine (1.0 mM) + atropine (0.1 mM)]. Cell adhesion to the DRG nerve fibers was observed using an inverted microscope every 4 h. After 72 h, the number of cholangiocarcinoma cells adhering to nerve fibers was quantified.

General matrix metalloproteinase (MMP) activity assay

The general activity of MMP enzymes was determined using an assay kit (ab112146, Abcam, Cambridge, MA, USA) according to the manufacturer’s protocol. In brief, experimental cells were seeded into triplicate wells of 6-well plates and allowed to attach overnight, then starved with serum free media for another 18 h. A kinetic measurement was then performed for the MMP activity by taking medium samples at 5 min intervals over a 1 h period after starting the reaction by using a microplate reader with a filter set of Ex/Em = 490/525 nm.

Statistical analysis

Statistical analyses were performed using SPSS 17.0 software. Each value in the present study was obtained from at least three independent experiments. Data are presented as the mean ± standard error of the mean (SEM). Survival analyses and cumulative recurrence rates were assessed using the Kaplan–Meier method and the log-rank test. The difference between two groups was evaluated with the Student’s t-test and was considered statistically significant when P < 0.05.

Results

Clinicopathological features and patient demographics

All of the cases of cholangiocarcinoma (n = 60; 43 men, 17 women; median age, 56 years; range, 30–82 years) were diagnosed as adenocarcinoma. Of these, 46 cases (76.67%) had lymphatic metastasis, while 14 (23.33%) did not. A total of 18 cases (30%) of high differentiation and 42 cases (70%) of moderate or low differentiation were observed. Furthermore, after classification the tumors according to the Union International Control Cancer TNM staging system [17], 16 cases (26.67%) were found to be stage I-II, while 44 cases (73.33%) were stage III-IV. Finally, we found that 53 cases (88.33%) of them had perineural invasion and the remaining 7 cases (11.67%) without perineural invasion.

M3-mAChR is highly expressed in cholangiocarcinoma and plays an important role in metastasis and prognosis in cholangiocarcinoma patients

Using immunohistochemistry, we observed M3-mAChR to be highly expressed in histopathological sections from cholangiocarcinoma tissues, but was found at lower levels in normal biliary tissues, with positive rates of 90% (54/60) and 3.33% (1/30), respectively (P < 0.01; Fig. 1a, b). In the positive tissues, M3-mAChR expression was mainly distributed in granules or sheets located in the cytoplasm near the cell membrane. In addition, perineural invasion was observed in the cholangiocarcinoma tissues at a positive rate of 91.7% (55/60; Fig. 1c, d).
Notably, M3-mAChR expression appears to be significantly correlated with the level of differentiation (P < 0.05), with the positive rate of M3-mAChR expression in III–IV staged tumors being higher than that in I–II staged tumors (P < 0.01). Similarly, expression was also correlated with lymphatic metastasis (P < 0.05), being higher in histopathological sections of cholangiocarcinoma with lymph node metastasis (P < 0.01). We also found that M3-mAChR expression is significantly associated with perineural invasion (P < 0.01; Table 1). The survival time of cholangiocarcinoma patients with high M3-mAChR expression was also distinctly shorter than that of patients with low M3-mAChR expression. Moreover, the survival time of cholangiocarcinoma patients was shorter when perineural invasion had occurred than in patients without perineural invasion (Fig. 1e, f). There were no connections between M3-mAChR expression and patient age, sex, tumor location (P ˃ 0.05). Collectively, these results show that M3-mAChR plays an important role in the invasion and metastasis of cholangiocarcinoma and affects treatment and prognosis.
Table 1
Clinicopathological features of patients with cholangiocarcinoma and correlations with M3 expression
Factor
n
M3 expression
P value
(−)
( + )
( ++)
( +++)
Positive rate (%)
Sex
 Male
43
5
6
14
18
88.89
0.500
 Female
17
1
6
4
6
93.33
 
Age
 < 59
23
2
5
7
9
91.30
0.962
 ≥ 59
37
4
7
11
15
89.19
 
Differentiation of tumors
 High
18
3
4
5
6
83.33
0.308
 Middle and poorly
42
3
8
13
18
92.86
 
Lymph node metastasis
 Yes
46
1
8
15
22
97.83
0.001
 No
14
5
4
3
2
64.29
 
TNM stage
 I–II
16
5
5
3
3
68.75
0.002
 III–IV
44
1
7
15
21
97.73
 
Tumor location
 Hilar
19
2
3
6
6
88.23
0.756
 Middle and distal
41
4
9
12
18
90.70
Perineural invasion
 Yes
53
3
10
16
24
94.34
0.007
 No
7
3
2
2
0
57.14
 
Correlations were estimated by Fisher’s exact test. Significant when p < 0.05

M3-mAChR mediate migration in cultured cholangiocarcinoma cells

Preliminary studies showed that both FRH0201 and RBE cells express M3-mAChR, although expression was higher in RBE cells than in FRH0201 cells (Fig. 2a, b). Notably, in RBE and FRH0201 cells treated with the M3-mAChR agonist pilocarpine, we observed an increase in the quantity of migrating cells compared with that of the untreated control group (Fig. 2c, d), suggesting that the M3-mAChR agonist significantly promoted the invasive characteristics of the cholangiocarcinoma cells. Moreover, while there was no difference between cells treated with the M3-mAChR antagonist atropine and control cells, when cells were treated with both pilocarpine and atropine, the quantity of migrating cells was reduced (Fig. 2b, c). These data suggest that the invasive capacity of the cholangiocarcinoma cells was decreased owing to antagonism of the pilocarpine-treated cells by atropine.
To further understand the role of M3-mAChR in cholangiocarcinoma cells, we transfected RBE cells with lentiviral vectors carrying shM3 to downregulate M3-mAChR expression (Fig. 3a). Our results show that M3-mAChR expression was downregulated in shM3-treated RBE cells, and the number of migrating cells was reduced. Furthermore, treatment with pilocarpine increased the invasive capacity of the shM3-treated RBE cells (Fig. 3b). In contrast, in FRH0201 cells, M3-mAChR expression was increased by transfecting the cells with an M3-mAChR plasmid, and this overexpression mediated an increase in cholangiocarcinoma cell invasion (Fig. 3b). Moreover, downregulation of M3-mAChR expression largely blocked invasion.

Perineural invasion is regulated by M3-mAChR

To investigate the role of M3-mAChR in perineural invasion of cholangiocarcinoma cells, we used an ex vivo DRG model. When the DRG from newborn Kunming mice were cultured alone (Fig. 4a-1–3), the Nissl bodies became granular after being dyed, showing obvious staining around the perinuclear region, but little staining around the edges of the nucleus (Fig. 4a–7). After Gomori staining and neurofilament protein immunohistochemical staining (NF IHC), neuritis was observed growing up from the DRG periphery, most of which showed branching and were generally present in a radial arrangement (Fig. 4a-6). Notably, when the DRG was co-cultured with untreated RBE cells, the cells appear to invade the DRG and the surrounding nerve fibers (Fig. 4b). Furthermore, when pilocarpine was added to the co-cultures, the quantity of RBE cells invading the DRG and surrounding nerve fibers was increased, suggesting that pilocarpine promotes perineural invasion during cholangiocarcinoma. The effects of pilocarpine were decreased by co-treatment with atropine, which decreased the number of RBE cells invading the DRG (Fig. 4b). Notably, atropine alone did not significantly affect the level of perineural invasion compared with that in the control group, which is consistent with our earlier cell culture data.
Furthermore, when the DRG were co-cultured with shM3-treated RBE cells, we found that although the shM3-treated RBE cells still invade the DRG, the quantity of invading cells was reduced compared with that in the untreated RBE group. Treating these DRG-shM3-RBE co-cultured cells with pilocarpine, the number of invading cells was increased (Fig. 4c). Conversely, when FRH0201 cells overexpressing M3-mAChR were co-cultured with DRG, the number of cells invading the DRG increased compared with that in the untreated FRH0201 group. Treatment these DRG-M3-FRH0201 co-cultured cells with pilocarpine, the number of invading cells was further increased (Fig. 4c). These results show that M3-mAChR expression in cholangiocarcinoma cells regulate perineural invasion.

M3-mAChR participate in AKT-mediated EMT in cholangiocarcinoma

Through this experiment we found that pilocarpine can induce the increase of M3-mAChR protein levels in cholangiocarcinoma, and atropine blocked these changes. So we believe that pilocarpine interacts with M3-mAChR and acts through M3-mAChR.
As EMT plays a key role in cholangiocarcinoma metastasis, we examined the effects of M3-mAChR agonist and antagonist treatment on the expression of EMT factors related to the PI3K/AKT pathway in RBE cells. Our results showed that pilocarpine treatment decreases the expression of PTEN and the epithelial cell marker E-cadherin, but increases the expression AKT and the Leydig cell marker vimentin. The expression of Snail, which induces EMT, was also increased. Notably, atropine blocked these changes (Fig. 5a, b). Similarly, shM3-mediated disruption of receptor expression in the RBE cells also resulted in increased expression of PTEN and E-cadherin and decreased expression of AKT, vimentin, and Snail. In contrast, upregulation of M3-mAChR in FRH0201 cells decreased PTEN and E-cadherin expression and increased AKT, vimentin, and Snail expression (Fig. 5c, d).
To better understand the role of M3-mAChR in AKT-mediated EMT, we knocked down AKT expression in FRH0201 cells overexpressing M3-mAChR. This disruption resulted in the downregulation of M3-mAChR, Snail, and vimentin expression, whereas the expression of PTEN and E-cadherin was upregulated (Fig. 6). Moreover, when AKT was downregulated, the levels of invasion and perineural invasion were significantly decreased. We also found that down-regulating the expression of AKT can reduce the activity of MMP in cholangiocarcinoma cells. Taken together, these results showed that the AKT pathway plays a significant role in M3-mAChR-mediated invasion and perineural invasion in cholangiocarcinoma.

Discussion

As an important digestive organ, the biliary system is responsible for biliary secretion and excretion and is surrounded by vagus nerves that can affect and be affected by processes occurring in the biliary cells. Notably, acetyl choline acts is the main neurotransmitter in this system, and acetyl choline receptors, including nicotinic acetylcholine receptors and mAchRs, play an important functional role during signaling. mAChRs are G protein-coupled receptors that regulate adenylate cyclase, ion channels, and phosphatidylinositol lipid turnover, thus modulating various biological reactions [18, 19].
Studies have shown that the M3-mAChR is widely expressed in digestive tract tumors and may play an important role in the proliferation, differentiation, and progression of cancer [13, 14, 2022]. Our results show that the M3-mAChR is highly expressed in cholangiocarcinoma and is closely related to the differentiation and clinical stages of the tumor as well as the presence of lymphatic metastasis. Indeed, poor cholangiocarcinoma differentiation was associated with high M3-mAChR expression. In III-IV cancers, the positive rate of M3-mAChR expression in cholangiocarcinoma was notably higher than that observed in I–II cancers, and the expression of M3-mAChR in cholangiocarcinoma patients with concomitant lymphatic metastasis was also increased. Thus, the pattern and correlation of M3-mAChR expression with survival indicate an important function for this receptor in the occurrence and development of cholangiocarcinoma along with patient prognosis.
In 1998, Wessler et al. [23] proposed the concept of the non-neuronal acetylnergic system, which is based on the non-neuronal acetylcholine system, and includes its synthetase, transporter, inactivation enzyme, and functional receptors. Functional abnormalities in this non-neuronal acetylnergic system have been related to various diseases, including inflammation, atherosclerosis, local and systemic infection, and cancer [24]. Many recent studies have also examined the interactions between neurotransmitters and tumors, particularly tumors exhibiting perineural invasion, such as cholangiocarcinoma [2527]. In fact, abnormal neurotransmitter expression and/or receptor signaling in tumors greatly affects tumor proliferation, differentiation, and metastasis, with receptor agonists increasing these characteristics and receptor antagonists blocking them [28, 29]. In a previous study, we found that the M3-mAChR agonist pilocarpine promoted the proliferation of cholangiocarcinoma cells, increased M3-mAChR expression in cholangiocarcinoma, and enhanced the effects of M3-mAChR; all of which were blocked by treatment with the antagonist atropine [12]. In the present study, we not only verified these changes, but we also found that M3-mAChR regulate the migration and metastasis of cholangiocarcinoma cells. Our data show that when M3-mAChR expression is downregulated via receptor knock-down, the migratory capacity of the affected cholangiocarcinoma cells was low. However, when treated with the M3-mAChR agonist pilocarpine, migration was increased. A similar effect was also observed when M3-mAChR was overexpressed in cholangiocarcinoma cells. However, it appears that these enhanced effects can be blocked using atropine, highlighting the potential use of this or other antagonists in clinical treatment.
Cholangiocarcinoma is one of the deadliest malignant tumors because of the high levels of local invasion and metastasis and the insensitivity of the cells to radiotherapy and chemotherapy. The early occurrence of perineural invasion and metastasis in cholangiocarcinoma has also been shown to affect mortality rate [30]. In addition to its role in migration, we also found that high M3-mAChR expression (via overexpression) promotes perineural invasion, whereas low M3-mAChR expression (via receptor knock-down) was associated with lower levels of perineural invasion. Similar results were also observed using pilocarpine and atropine. These findings suggest that decreasing M3-mAChR expression via knock-down or antagonist treatment may limit perineural invasion of cholangiocarcinoma tumors in the clinical setting.
To understand how M3-mAChR regulate migration, perineural invasion, and metastasis in cholangiocarcinoma, we evaluated the expression and function of various factors and signaling pathways. EMT is widely known to play an essential role in metastasis and confers noninvasive cells with invasive and aggressive features [7], leading to the occurrence and progression of epithelial cell tumors [17]. The phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway plays an important role in regulating cell proliferation and EMT of malignant cells [3133]. Our results show that downregulation of M3-mAChR expression in cholangiocarcinoma cells also decreases the expression of PTEN and the epithelial cell marker E-cadherin, while increasing that of AKT and the Leydig cell marker vimentin. These results demonstrate that downregulation of M3-mAChR blocks EMT, whereas upregulation of M3-mAChR transforms cholangiocarcinoma cells into Leydig cells, leading to invasion and metastasis. Additionally, the expression of Snail, an important factor promoting EMT, was also increased after M3-mAChR downregulation, which could subsequently activate other deviant signaling pathways.
The serine/threonine kinase AKT (also known as protein kinase B), which contains a panel of three isoforms (AKT1, AKT2 and AKT3) in mammals, is a key communicator of PI3 K signaling. Further, knockdown of AKT in FRH0201 cells overexpressing M3-mAChR which resulted in the downregulation of M3-mAChR, Snail, and vimentin expression, whereas the expression of PTEN and E-cadherin was upregulated. AKT may have negative feedback in the regulation of M3-mAChR. Moreover, when AKT was downregulated, the levels of invasion and perineural invasion were significantly decreased. We also found that down-regulating the expression of AKT can reduce the activity of MMP in cholangiocarcinoma cells which may be the cause of the decline in invasiveness. The AKT pathway appears to play a particularly prominent role in M3-mAChR signaling as downregulation of this factor significantly decreased migration, metastasis, and perineural invasion.

Conclusions

In this study, we evaluated the changes in M3-mAChR expression in human tissue samples and in cultured tumor cells in addition to investigating the effects of these changes on migration/metastasis, perineural invasion, and EMT in cholangiocarcinoma. Our results demonstrate that M3-mAChR regulate EMT through the AKT pathway and subsequently affect perineural invasion and metastasis during cholangiocarcinoma pathogenesis. While additional work, particularly the validation of these findings in animal models of the disease, is required, this study is the first to indicate a distinct role for M3-mAChR-mediated regulation in multiple aspects of cholangiocarcinoma. Furthermore, our data also highlight a theoretical basis for blocking perineural invasion and metastasis during cholangiocarcinoma using antagonists. Taken together, this study provides essential insight into the mechanisms and potential treatment options of cholangiocarcinoma.

Authors’ contributions

FYJ and ZBY designed the experiments; FYJ, HX and LGW performed the experiments; LLF and ZW collected the data; MK and FYJ analyzed the data; SFZ and SCD provided the human samples; FYJ and ZCZ wrote the manuscript. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.
All participants consent to their data being published.
All procedures in this study that involved human participants were performed in accordance with the 1964 Helsinki declaration and its later amendments. The protocol was approved by the Ethics Committee of The Affiliated Hospital of Qingdao University. Informed consent was obtained from all individual participants included in the study. Furthermore, the care and use of laboratory animals was in accordance with the principles and standards set forth in the Principles for Use of Animals (National Guide for Grants and Contracts).

Funding

This work was supported by a Grant from the National Natural Science Foundation (81272362).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Razumilava N, Gores GJ. Cholangiocarcinoma. Lancet. 2014;383(9935):2168–79.CrossRef Razumilava N, Gores GJ. Cholangiocarcinoma. Lancet. 2014;383(9935):2168–79.CrossRef
2.
Zurück zum Zitat Nakagohri T, Asano T, Kinoshita H, Kenmochi T, Urashima T, Miura F, Ochiai T. Aggressive surgical resection for hilar-invasive and peripheral intrahepatic cholangiocarcinoma. World J Surg. 2003;27(3):289–93.CrossRef Nakagohri T, Asano T, Kinoshita H, Kenmochi T, Urashima T, Miura F, Ochiai T. Aggressive surgical resection for hilar-invasive and peripheral intrahepatic cholangiocarcinoma. World J Surg. 2003;27(3):289–93.CrossRef
3.
Zurück zum Zitat Murakawa K, Tada M, Takada M, Tamoto E, Shindoh G, Teramoto K, Matsunaga A, Komuro K, Kanai M, Kawakami A, Fujiwara Y, Kobayashi N, Shirata K, Nishimura N, Okushiba S, Kondo S, Hamada J, Katoh H, Yoshiki T, Moriuchi T. Prediction of lymph node metastasis and perineural invasion of biliary tract cancer by selected features from cDNA array data. J Surg Res. 2004;122(2):184–94.CrossRef Murakawa K, Tada M, Takada M, Tamoto E, Shindoh G, Teramoto K, Matsunaga A, Komuro K, Kanai M, Kawakami A, Fujiwara Y, Kobayashi N, Shirata K, Nishimura N, Okushiba S, Kondo S, Hamada J, Katoh H, Yoshiki T, Moriuchi T. Prediction of lymph node metastasis and perineural invasion of biliary tract cancer by selected features from cDNA array data. J Surg Res. 2004;122(2):184–94.CrossRef
4.
Zurück zum Zitat Maxwell P, Hamilton PW, Sloan JM. Three-dimensional reconstruction of perineural invasion in carcinoma of the extrahepatic bile ducts. J Pathol. 1996;180(2):142–5.CrossRef Maxwell P, Hamilton PW, Sloan JM. Three-dimensional reconstruction of perineural invasion in carcinoma of the extrahepatic bile ducts. J Pathol. 1996;180(2):142–5.CrossRef
5.
Zurück zum Zitat Natsis K, Paraskevas G, Papaziogas B, Agiabasis A. “Pes anserinus” of the right phrenic nerve innervating the serous membrane of the liver: a case report (anatomical study). Morphologie. 2004;88(283):203–5.CrossRef Natsis K, Paraskevas G, Papaziogas B, Agiabasis A. “Pes anserinus” of the right phrenic nerve innervating the serous membrane of the liver: a case report (anatomical study). Morphologie. 2004;88(283):203–5.CrossRef
6.
Zurück zum Zitat Yazumi S, Ko K, Watanabe N, Shinohara H, Yoshikawa K, Chiba T, Takahashi R. Disrupted transforming growth factor-beta signaling and deregulated growth in human biliary tract cancer cells. Int J Cancer. 2000;86(6):782–9.CrossRef Yazumi S, Ko K, Watanabe N, Shinohara H, Yoshikawa K, Chiba T, Takahashi R. Disrupted transforming growth factor-beta signaling and deregulated growth in human biliary tract cancer cells. Int J Cancer. 2000;86(6):782–9.CrossRef
7.
Zurück zum Zitat Zhang KJ, Wang DS, Zhang SY, Jiao XL, Li CW, Wang XS, Yu QC, Cui HN. The E-cadherin repressor slug and progression of human extrahepatic hilar cholangiocarcinoma. J Exp Clin Cancer Res. 2010;29:88.CrossRef Zhang KJ, Wang DS, Zhang SY, Jiao XL, Li CW, Wang XS, Yu QC, Cui HN. The E-cadherin repressor slug and progression of human extrahepatic hilar cholangiocarcinoma. J Exp Clin Cancer Res. 2010;29:88.CrossRef
8.
Zurück zum Zitat Yoshida GJ. Emerging role of epithelial–mesenchymal transition in hepatic cancer. J Exp Clin Cancer Res. 2016;35(1):141.CrossRef Yoshida GJ. Emerging role of epithelial–mesenchymal transition in hepatic cancer. J Exp Clin Cancer Res. 2016;35(1):141.CrossRef
9.
Zurück zum Zitat Du R, Wu S, Lv X, Fang H, Wu S, Kang J. Overexpression of brachyury contributes to tumor metastasis by inducing epithelial–mesenchymal transition in hepatocellular carcinoma. J Exp Clin Cancer Res. 2014;33:105.CrossRef Du R, Wu S, Lv X, Fang H, Wu S, Kang J. Overexpression of brachyury contributes to tumor metastasis by inducing epithelial–mesenchymal transition in hepatocellular carcinoma. J Exp Clin Cancer Res. 2014;33:105.CrossRef
10.
Zurück zum Zitat Vaquero J, Guedj N, Clapéron A, Nguyen Ho-Bouldoires TH, Paradis V, Fouassier L. Epithelial–mesenchymal transition in cholangiocarcinoma: from clinical evidence to regulatory networks. J Hepatol. 2017;66(2):424–41.CrossRef Vaquero J, Guedj N, Clapéron A, Nguyen Ho-Bouldoires TH, Paradis V, Fouassier L. Epithelial–mesenchymal transition in cholangiocarcinoma: from clinical evidence to regulatory networks. J Hepatol. 2017;66(2):424–41.CrossRef
11.
Zurück zum Zitat Caulfield MP, Birdsall NJ. International union of pharmacology. XVII. Classification of muscarinic acetylcholine receptors. Pharmacol Rev. 1998;50(2):279–90.PubMed Caulfield MP, Birdsall NJ. International union of pharmacology. XVII. Classification of muscarinic acetylcholine receptors. Pharmacol Rev. 1998;50(2):279–90.PubMed
12.
Zurück zum Zitat Feng YJ, Zhang BY, Yao RY, Lu Y. Muscarinic acetylcholine receptor M3 in proliferation and perineural invasion of cholangiocarcinoma cells. Hepatobiliary Pancreat Dis Int. 2012;11(4):418–23.CrossRef Feng YJ, Zhang BY, Yao RY, Lu Y. Muscarinic acetylcholine receptor M3 in proliferation and perineural invasion of cholangiocarcinoma cells. Hepatobiliary Pancreat Dis Int. 2012;11(4):418–23.CrossRef
13.
Zurück zum Zitat Raufman JP, Samimi R, Shah N, Khurana S, Shant J, Drachenberg C, Xie G, Wess J, Cheng K. Genetic ablation of M3 muscarinic receptors attenuates murine colon epithelial cell proliferation and neoplasia. Cancer Res. 2008;68(10):3573–8.CrossRef Raufman JP, Samimi R, Shah N, Khurana S, Shant J, Drachenberg C, Xie G, Wess J, Cheng K. Genetic ablation of M3 muscarinic receptors attenuates murine colon epithelial cell proliferation and neoplasia. Cancer Res. 2008;68(10):3573–8.CrossRef
14.
Zurück zum Zitat Ukegawa JI, Takeuchi Y, Kusayanagi S, Mitamura K. Growth-promoting effect of muscarinic acetylcholine receptors in colon cancer cells. J Cancer Res Clin Oncol. 2003;129(5):272–8.PubMed Ukegawa JI, Takeuchi Y, Kusayanagi S, Mitamura K. Growth-promoting effect of muscarinic acetylcholine receptors in colon cancer cells. J Cancer Res Clin Oncol. 2003;129(5):272–8.PubMed
15.
Zurück zum Zitat Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25(4):402–8.CrossRef Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25(4):402–8.CrossRef
16.
Zurück zum Zitat Na’ara S, Gil Z, Amit M. In vitro modeling of cancerous neural invasion: the dorsal root ganglion model. J Vis Exp. 2016;110:e52990. Na’ara S, Gil Z, Amit M. In vitro modeling of cancerous neural invasion: the dorsal root ganglion model. J Vis Exp. 2016;110:e52990.
17.
Zurück zum Zitat Hwang S, Lee YJ, Song GW, Park KM, Kim KH, Ahn CS, Moon DB, Lee SG. Prognostic impact of tumor growth type on 7th AJCC staging system for intrahepatic cholangiocarcinoma: a single-center experience of 659 cases. J Gastrointest Surg. 2015;19(7):1291–304.CrossRef Hwang S, Lee YJ, Song GW, Park KM, Kim KH, Ahn CS, Moon DB, Lee SG. Prognostic impact of tumor growth type on 7th AJCC staging system for intrahepatic cholangiocarcinoma: a single-center experience of 659 cases. J Gastrointest Surg. 2015;19(7):1291–304.CrossRef
18.
Zurück zum Zitat Gilman AG. G proteins and dual control of adenylate cyclase. Cell. 1984;36(3):577–9.CrossRef Gilman AG. G proteins and dual control of adenylate cyclase. Cell. 1984;36(3):577–9.CrossRef
19.
Zurück zum Zitat Malbon CC. G proteins in development. Nat Rev Mol Cell Biol. 2005;6(9):689–701.CrossRef Malbon CC. G proteins in development. Nat Rev Mol Cell Biol. 2005;6(9):689–701.CrossRef
20.
Zurück zum Zitat Park YS, Cho NJ. Enhanced proliferation of SNU-407 human colon cancer cells by muscarinic acetylcholine receptors. BMB Rep. 2008;41(11):803–7.CrossRef Park YS, Cho NJ. Enhanced proliferation of SNU-407 human colon cancer cells by muscarinic acetylcholine receptors. BMB Rep. 2008;41(11):803–7.CrossRef
21.
Zurück zum Zitat Frucht H, Jensen RT, Dexter D, Yang WL, Xiao Y. Human colon cancer cell proliferation mediated by the M3 muscarinic cholinergic receptor. Clin Cancer Res. 1999;5(9):2532–9.PubMed Frucht H, Jensen RT, Dexter D, Yang WL, Xiao Y. Human colon cancer cell proliferation mediated by the M3 muscarinic cholinergic receptor. Clin Cancer Res. 1999;5(9):2532–9.PubMed
22.
Zurück zum Zitat Wegener C, Hamasaka Y, Nässel DR. Acetylcholine increases intracellular Ca2+ via nicotinic receptors in cultured PDF-containing clock neurons of Drosophila. J Neurophysiol. 2004;91(2):912–23.CrossRef Wegener C, Hamasaka Y, Nässel DR. Acetylcholine increases intracellular Ca2+ via nicotinic receptors in cultured PDF-containing clock neurons of Drosophila. J Neurophysiol. 2004;91(2):912–23.CrossRef
23.
Zurück zum Zitat Wessler I, Kirkpatrick CJ, Racké K. Non-neuronal acetylcholine, a locally acting molecule, widely distributed in biological systems: expression and function in humans. Pharmacol Ther. 1998;77(1):59–79.CrossRef Wessler I, Kirkpatrick CJ, Racké K. Non-neuronal acetylcholine, a locally acting molecule, widely distributed in biological systems: expression and function in humans. Pharmacol Ther. 1998;77(1):59–79.CrossRef
24.
Zurück zum Zitat Grando SA, Kawashima K, Kirkpatrick CJ, Kummer W, Wessler I. Recent progress in revealing the biological and medical significance of the non-neuronal cholinergic system. Int Immunopharmacol. 2015;29(1):1–7.CrossRef Grando SA, Kawashima K, Kirkpatrick CJ, Kummer W, Wessler I. Recent progress in revealing the biological and medical significance of the non-neuronal cholinergic system. Int Immunopharmacol. 2015;29(1):1–7.CrossRef
25.
Zurück zum Zitat Schuller HM, Porter B, Riechert A. Beta-adrenergic modulation of NNK-induced lung carcinogenesis in hamsters. J Cancer Res Clin Oncol. 2000;126(11):624–30.CrossRef Schuller HM, Porter B, Riechert A. Beta-adrenergic modulation of NNK-induced lung carcinogenesis in hamsters. J Cancer Res Clin Oncol. 2000;126(11):624–30.CrossRef
26.
Zurück zum Zitat Drell TL 4th, Joseph J, Lang K, Niggemann B, Zaenker KS, Entschladen F. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res Treat. 2003;80(1):63–70.CrossRef Drell TL 4th, Joseph J, Lang K, Niggemann B, Zaenker KS, Entschladen F. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res Treat. 2003;80(1):63–70.CrossRef
27.
Zurück zum Zitat Masur K, Niggemann B, Zanker KS, Entschladen F. Norepinephrine-induced migration of SW480 colon carcinoma cells is inhibited by beta-blockers. Cancer Res. 2001;61(7):2866–9.PubMed Masur K, Niggemann B, Zanker KS, Entschladen F. Norepinephrine-induced migration of SW480 colon carcinoma cells is inhibited by beta-blockers. Cancer Res. 2001;61(7):2866–9.PubMed
28.
Zurück zum Zitat Radu A, Pichon C, Camparo P, Antoine M, Allory Y, Couvelard A, Fromont G, Hai MT, Ghinea N. Expression of follicle-stimulating hormone receptor in tumor blood vessels. N Engl J Med. 2010;363(17):1621–30.CrossRef Radu A, Pichon C, Camparo P, Antoine M, Allory Y, Couvelard A, Fromont G, Hai MT, Ghinea N. Expression of follicle-stimulating hormone receptor in tumor blood vessels. N Engl J Med. 2010;363(17):1621–30.CrossRef
29.
Zurück zum Zitat Leu FP, Nandi M, Niu C. The effect of transforming growth factor beta on human neuroendocrine tumor BON cell proliferation and differentiation is mediated through somatostatin signaling. Mol Cancer Res. 2008;6(6):1029–42.CrossRef Leu FP, Nandi M, Niu C. The effect of transforming growth factor beta on human neuroendocrine tumor BON cell proliferation and differentiation is mediated through somatostatin signaling. Mol Cancer Res. 2008;6(6):1029–42.CrossRef
30.
Zurück zum Zitat Shen FZ, Zhang BY, Feng YJ, Jia ZX, An B, Liu CC, Deng XY, Kulkarni AD, Lu Y. Current research in perineural invasion of cholangiocarcinoma. J Exp Clin Cancer Res. 2010;29:24.CrossRef Shen FZ, Zhang BY, Feng YJ, Jia ZX, An B, Liu CC, Deng XY, Kulkarni AD, Lu Y. Current research in perineural invasion of cholangiocarcinoma. J Exp Clin Cancer Res. 2010;29:24.CrossRef
31.
Zurück zum Zitat Wang C, Mao ZP, Wang L, Wu GH, Zhang FH, Wang DY, Shi JL. Long non-coding RNA MALAT1 promotes cholangiocarcinoma cell proliferation and invasion by activating PI3 K/Akt pathway. Neoplasma. 2017;64(5):725–31.CrossRef Wang C, Mao ZP, Wang L, Wu GH, Zhang FH, Wang DY, Shi JL. Long non-coding RNA MALAT1 promotes cholangiocarcinoma cell proliferation and invasion by activating PI3 K/Akt pathway. Neoplasma. 2017;64(5):725–31.CrossRef
32.
Zurück zum Zitat Choi Y, Ko YS, Park J, Choi Y, Kim Y, Pyo JS, Jang BG, Hwang DH, Kim WH, Lee BL. HER2-induced metastasis is mediated by AKT/JNK/EMT signaling pathway in gastric cancer. World J Gastroenterol. 2016;22(41):9141–53.CrossRef Choi Y, Ko YS, Park J, Choi Y, Kim Y, Pyo JS, Jang BG, Hwang DH, Kim WH, Lee BL. HER2-induced metastasis is mediated by AKT/JNK/EMT signaling pathway in gastric cancer. World J Gastroenterol. 2016;22(41):9141–53.CrossRef
33.
Zurück zum Zitat Xu W, Yang Z, Lu N. A new role for the PI3K/Akt signaling pathway in the epithelial–mesenchymal transition. Cell Adh Migr. 2015;9(4):317–24.CrossRef Xu W, Yang Z, Lu N. A new role for the PI3K/Akt signaling pathway in the epithelial–mesenchymal transition. Cell Adh Migr. 2015;9(4):317–24.CrossRef
Metadaten
Titel
M3 muscarinic acetylcholine receptors regulate epithelial–mesenchymal transition, perineural invasion, and migration/metastasis in cholangiocarcinoma through the AKT pathway
verfasst von
Yujie Feng
Xiao Hu
Guangwei Liu
Lianfang Lu
Wei Zhao
Fangzhen Shen
Kai Ma
Chuandong Sun
Chengzhan Zhu
Bingyuan Zhang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2018
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0667-z

Weitere Artikel der Ausgabe 1/2018

Cancer Cell International 1/2018 Zur Ausgabe

Update Onkologie

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