Skip to main content
Erschienen in: Diagnostic Pathology 1/2016

Open Access 01.12.2016 | Research

DLC-1 is an independent prognostic marker and potential therapeutic target in hepatocellular cancer

verfasst von: L. J. Song, Q. Liu, X. R. Meng, SH. L Li, L. X. Wang, Q. X. Fan, X. Y. Xuan

Erschienen in: Diagnostic Pathology | Ausgabe 1/2016

Abstract

Background

The 5-year survival rate of patients with hepatocellular cancer (HCC) was very low because of invasion and metastasis in the early stage. Biomarkers might help predict early occurrence of invasion and metastasis. Accumulating evidence has shown that deleted in liver cancer-1 (DLC1) may be considered as a metastasis suppressor gene in numerous solid and hematological cancers. However, its prognostic role and mechanisms that regulate and coordinate these activities remain poorly understood.

Methods

With the method of immunohistochemistry, the expression of DLC-1 as well as Rho A, ROCK2, moesin had been characterized in 80 HCC tissues and adjacent noncancerous tissues. The correlation between their expression and their relationships with clinicopathological characteristics of HCC were also investigated. In addition, the prognostic value of DLC1 expression within the tumor tissues was assessed by Cox regression and Kaplan-Meier analysis.

Results

DLC1 expression was significantly lower in HCC tissues than in adjacent noncancerous tissues, and DLC-1 expression was found to be negatively correlated with tumor differentiation, TNM stage and lymph node metastasis. Furthermore, DLC-1 expression was found to inversely correlate with Rho A, ROCK2 and moesin which were all highly expressed in HCC tissues. Kaplan-Meier analysis showed that significantly longer 5-year survival rate was seen in HCC patients with higher DLC1 expression, compared to those with lower expression of DLC1. Multivariate Cox proportional hazard analyses revealed that DLC1 was an independent factor affecting the overall survival probability.

Conclusion

DLC1 could be served as a tumor suppressor gene in the progression especially in the invasion and metastasis of HCC. DLC1 perhaps played its role by regulating the expression of Rho A, ROCK2 and moesin. Evaluation of the expression of DLC-1 might be a good prognostic marker for patients with HCC.
Hinweise

Competing interests

The authors declare that there are no conflicts of interest.

Authors’ contributions

LJ S and QL are responsible for laboratory work, data analysis and paper writing. XR M is responsible for laboratory work and data analysis. SL L is responsible for sample collection and clinical data analyses. LX W and QX F are responsible for project design. XY X is responsible for project design and manuscript preparation. All authors read and approved the final manuscript.

Background

Liver cancer is one of the most common malignancies in China and the 5-year survival rate of it is very low because of invasion and metastasis in the early stage [1, 2]. Mechanisms of invasion and metastasis are diverse and not yet clear-cut in most cases. So the identification of indicators or markers that may help us to assess tumor behavior of invasion and metastasis are very important for us [3].
Deleted in liver cancer-1 (DLC-1) was first cloned by using subtractive hybridization method in human hepatocellular carcinomas and later it was identified as a breast cancer metastasis suppressor in microarray comparisons between breast cancer cell lines [4, 5]. Then, deletion or promoter methylation of DLC-1 has been described in multiple cancers. DLC-1 is a RhoGAP (GTPase-activating protein) that inhibits/inactivates Rho-dependent signal transduction [4, 6]. While Rho is key factors in cell proliferation, polarity, cytoskeletal remodeling and migration, the aberrant function of their regulators may lead to cell transformation [6, 7]. Data showed that DLC-1 mRNA expression was lost in 95 % of patient with NSCLC tumors tissues and 58 % of NSCLC cell lines, due at least in part through its function as a RhoGAP and thus negatively regulating the expression of RhoA and related RhoB, RhoC [8]. Nevertheless, a detection of DLC-1 as a therapeutic indicator or prognostic markers in liver cancers have litter been elucidated and the role and mechanism of DLC-1 in liver cancer especially in the invasion and metastasis process remains to be fully eclucidated.
There is considerable and growing evidence for the importance of the ROCK gene in actomyosin contractility, focal adhesion assembly, cytokinesis and cell proliferation [9, 10]. ROCK has also been implicated in colorectal [11], breast [12], gastric [13], and hepatocellular cancer metastatic growth [14]. One identified mechanism for ROCK activation in cancer involves the loss of function of the DLC-1, which encodes a GTPase activating protein (RhoGAP) for the RhoA and RhoC small GTPases [15].
Moesin, a member of the ERM (moesin, radixin, ezrin) family of proteins has been reported to be overexpressed in many kinds of cancers [16, 17]. Leroi et al. [18] sowed that moesin not ezrin and radixin was up-regulated in glioblastoma multiforme in comparison to non-malignant brain tissue samples. In addition, He [19] et al. reported that the extracellular small GTPase RhoA/ROCK-2 cascade mediated the increased moesin expression and phosphorylation, however, the relationship between DLC-1 and moesin in liver cancer has not been evaluated before.
In the current study, the relationship between the expression of DLC1 and various clinicopathologic parameters and immunohistochemical markers, such as RhoA, ROCK2 and Moesin were analyzed. Furthermore, the prognostic significance of DLC1 in human HCC was also explored.

Methods

Patient specimens

This study was approved by the Ethics Committee of the First Affiliated Hospital of Zhengzhou University (Zhengzhou, Henan, China). Written consents were obtained from all those patients included in this study. A total of 80 formalin-fixed paraffin-embedded liver cancer tissues and the corresponding normal tissues were collected from the archive of the First Affiliated Hospital of Zhengzhou University from 2009 to 2013. None of these patients received radiotherapy or chemotherapy prior to surgery. Briefly, the 80 patients consisted of 19 females and 61 males with ages ranging between 36 and 78 years old (mean age: 65 years old). According to the newer 2010 TNM classification of malignant tumours, liver cancers were staged into 54 I-II and 26 III stages. The histological grade of tumor differentiation was assigned into 61 I--II stage and 29 III-IV stage. Cancer size was calculated by measuring the largest dimension of the cancer specimen. 34 cases localized liver cancers without dissemination in liver and portal vein invasion were viewed as low invasion groups while other 46 cases with dissemination in liver and portal vein invasion were viewed as high.

Immunohistochemistry

The immunohistochemistry protocols were described previously [20]. Primary antibodies against DLC-1, Rho A, ROCK2 and Moesin were used to detect the corresponding proteins. Briefly, slides were incubated with 1.5 % H2O2 at room temperature for 20 min to eliminate the endogenous peroxidase activity. Then the slides were incubated with a solution comprising 0.2 % triton-X 100 and 5 % goat serum at room temperature for 2 h to quench immunoglobulin’s non-specific binding. After that, the primary antibodies were added to the slides individually and incubated overnight at 4 °C. Next day, secondary antibody was applied to the slides and incubated at room temperature for 2 h. At last, the immune complexs were visualized by incubating the sections with DAB. The slides were counterstained with hematoxylin and mounted. Negative controls were identically treated with the primary antibodies by PBS.

Immunohistochemistry evaluation

Immunostained sections were evaluated independently by two pathologists. Immunostaining was evaluated semi-quantitatively by estimating immunoreactivity extent and intensity. Staining extent was rated according to the percentage of positive cells. Samples with no stained tumor cells were rated as 0, those with <10 % of stained tumor cells were rated as 1, those 11–50 % of stained tumor cells were rated as 2, those with 50–75 % of stained tumor cells were rated as 3 and those with >75 % of stained tumor cells were rated as 4. Staining intensity was graded as follows: negative (0), weak (1), moderate (2) and strong (3). The overall scores were the product of staining intensity multiplied the staining extent. An overall staining score of 0–3 and >3 were regarded as negative (−) and positive (+) protein expression, respectively.

Statistical analyses

The significant difference of DLC-1, Rho A, ROCK2 and moesin gene expression between specimen groups was carried out using the unpaired Student’s t-test. The survival rates after tumor removal were calculated by the Kaplan-Meier method, and differences in survival curves were analyzed by the Log-rank tests. Multivariate survival analysis was performed on all the significant characteristics measured by univariate survival analysis through the Cox proportional hazard regression model. Differences were considered statistically significant when the P-value was <0.05.

Results

The expression of DLC-1, Rho A, ROCK2 and moesin proteins in liver cancer tissues

The cellular locations of DLC-1, Rho A, ROCK2 and moesin proteins were all mainly found in the cytoplasm. As shown in Figs. 1a, b; 2a, b; 3a, b and 4a, b, positive staining rates of DLC-1, Rho A, ROCK2 and moesin in liver cancer tissue samples were 38.75, 78.75, 75.00 and 86.25 % respectively, which was significantly different from those in normal mucosa (83.75, 43.75, 40.00 and 66.25 %) (P < 0.001, Table 1).
Table 1
The expression of DLC-1, RhoA, ROCK2 and moesin in HCC tumor and adjacent tissues
  
HCC tissues (n = 80)
Adjacent liver tissues (n = 80)
p
DLC-1
High
31
67
<0.001
Low
49
13
RhoA
High
63
35
<0.001
Low
17
45
ROCK2
High
60
32
<0.001
Low
20
48
moesin
High
69
53
<0.001
Low
11
27

Clinicopathological significance of the expression of DLC-1, Rho A, ROCK2 and moesin proteins in liver cancer tissues

Clinicopathological significance of the expression of DLC-1, Rho A, ROCK2 and moesin proteins in liver cancer tissues are shown in Table 2. We found a negative correlation between DLC-1 expression and Tumor differentiation (p < 0.001) and TNM stage (p < 0.001).In addition, the expression of DLC-1 in low invasion groups was higher than those in high invasion groups (p < 0.001).
Table 2
The relationship between DLC-1, RhoA, ROCK2, moesin and clinicopathological variables in HCC tissues
Clinicopatholog-ical variables
DLC1 expression
RhoA expression
Rock2 expression
moesin expression
 
H
L
p
H
L
p
H
L
p
H
L
p
Gender
 Male
23
38
0.731
48
13
0.98
46
15
0.879
55
8
0.29
 Female
8
11
15
4
14
5
15
4
Age (years)
  < 45
9
14
0.958
17
6
0.502
17
6
0.887
19
4
0.548
  ≥ 45
22
35
46
11
43
14
50
7
Tumor number
 Singel
20
30
0.767
40
10
0.724
39
11
0.424
43
7
0.933
 Multiple
11
19
23
7
21
9
26
4
Maximal tumor size (cm)
  < 5
19
30
0.995
38
11
0.742
37
13
0.789
43
6
0.623
  ≥ 5
12
19
25
6
23
7
26
5
Tumor differentiation
 I-II
12
49
<0.001
36
15
0.018
33
18
<0.001
41
10
0.043
 III-IV
19
10
27
2
27
2
28
1
HBV infection
 No
7
11
0.989
14
4
0.909
13
5
<0.001
15
3
0.683
 Yes
24
38
49
13
47
15
54
8
TNM stage
 I-II
11
43
<0.001
39
15
0.034
37
17
0.002
44
10
0.074
 III
20
6
24
2
23
3
25
1
Portal vein invasion and dissemination
 Yes
8
38
<0.001
39
7
<0.001
40
6
0.004
43
3
0.029
 No
23
11
14
20
20
14
  
While the expression levels of Rho A, ROCK2 and moesin were positively correlated with Tumor differentiation, and TNM stage. Furthermore, Rho A, ROCK2 and moesin expression levels were significantly higher in HCC cancer patients with high invasion ability.

Association between DLC-1 and Rho A, ROCK2, moesin expression in HCC cancer tissues

Spearman’s rank correlational analysis revealed a significant negative correlation between the expression levels of DLC-1 and Rho A, ROCK2, moesin (r = −0.963, −0.669, −0.631).

Survival analysis

HCC patients with low DLC1 expression had lower disease-free and 5-year survival rates than those with high DLC1 expression as determined using the Kaplan-Meier method (Fig. 5). Univariate Cox regression analysis also identified that clinical variables including gender, tumor number, maximal tumor size, HBV infection, tumor differentiation, TNM stage and portal vein invasion and dissemination were all positively correlated with overall survival. Furthermore, to evaluate the potential of DLC1 expression as an independent prognostic marker for overall survival of HCC, multivariate Cox regression analyses were performed. While the others failed to demonstrate independence, DLC1 expression (HR: 2.234, 95 % CI: 0.937–4.148, p < 0.001), tumor differentiation (HR: 3.769, 95 % CI: 1.925–6.989, P = 0.003), and portal vein invasion and dissemination (HR: 1.341,95 % CI: 0.843–2.631, p < 0.001) may play a role in predicting the overall survival in HCC (p < 0.05).

Discussion

HCC is one of the most common malignancies worldwide. Despite advances in the detection and treatment of HCC, the mortality rate remains high because of early occurrence of invasion and metastasis [21]. Hence to improve early detection of invasion and metastasis and their molecular characterization in HCC, to develop interventions that are targeted at blocking or reversing the invasion and metastasis process are very important for us.
DLC-1 has been known as tumor suppressor gene in many kinds of cancers including liver, lung, breast, brain, prostate and colon cancers [22, 23]. And re-expression of DLC-1 in cancer cells regulates the structure of actin cytoskeleton and focal adhesions and significantly inhibits cell growth, colony formation and invasion capacity, supporting its role as a tumor suppressor [24]. Thus, DLC-1 has the potential to be a key therapeutic indicator or markers for cancer gene therapy. In the present study, DLC-1 was found to be lower expressed in HCC tissues. DLC-1 positive expression was observed in 31 out of 80 HCC patients. And the lower expression of DLC-1 was correlated negatively with tumor differentiation and TNM stage. More importantly, the lower expression of DLC-1 found to be negatively linked with aggressive clinical behavior. HCC tissues with dissemination in liver and portal vein invasion showed lower DLC-1 expression than those in localized tissues without dissemination in liver and portal vein invasion.
Regarding DLC1 expression, recent reports have shown a growing interest in its prognostic impact in terms of survival. In this study, the prognosis of HCC patients with a low expression of DLC1 was poor, and Cox regression analysis indicated that low expression level of DLC1 was a significant prognostic factor for a poor survival rate of HCC patients. These findings raised the possibility that lower expression of DLC1 could be a factor of worst disease-specific survival, independently of any other variables.
DLC-1 was also named ARHGAP7 because it contains a RhoGTPase activating protein (RhoGAP) domain and these domains can convert the active GTP-bound Rho proteins to the inactive GDP-bound state and negatively regulate Rho GTPases [25]. RhoA is one of the best-known members of RhoGAP. ROCKs were originally isolated as downstream targets of Rho A [26]. RhoA binding with the C-terminal domain of ROCK, forming the Rho A/ROCK pathway, and controls a wide variety of cellular processes dependent on the re-arrangement of the actin cytoskeleton and changes of cell contractility. Yet, ROCK does not directly act on cytoskeletal molecules and ERM (radixin/ezrin/moesin) proteins are emerged as the candidates that likely linking the activation of ROCK and the cytoskeleton reorganization [27, 28]. Moesin is one of the most important and it has been reported that Rho A/ROCK is a typical upstream pathway for the phosphorylation of moesin [29, 30]. In the present study, Rho A and ROCK2 expression were found to negatively correlated with DLC-1 expression. Rho A and ROCK2 expression were identified in 63 and 60 of the HCC cases. Expression of Rho A and ROCK2 were all correlated positively tumor differentiation and TNM stage dissemination in liver and portal vein invasion. Furthermore, positive expression of Rho A and ROCK2 were also linked with dissemination in liver and portal vein invasion of HCC tissues. These data indicated that DLC-1, Rho A and ROCK2 perhaps together played an important role in the invasion and metastasis of HCC, and that DLC-1 perhaps played as a tumor suppressor by inhibiting the activation of Rho A/ROCK2 pathway . Furthermore, we found that the expression of phosphorylation of moesin was negatively correlated with the expression of DLC-1. Positive expression of phosphorylation of moesin was all correlated positively with tumor differentiation, TNM stage and dissemination in liver and portal vein invasion. So DLC-1 perhaps exerted its effects in invasion in HCC by regulating the expression of moesin through the RhoA/ROCK pathway.
Regarding DLC1 expression, recent reports have shown a growing interest in its prognostic impact in terms of survival. In this study, the prognosis of HCC patients with a low expression of DLC1 was poor, and Cox regression analysis indicated that low expression level of DLC1 was a significant prognostic factor for a poor survival rate of HCC patients. These findings raised the possibility that lower expression of DLC1 could be a factor of worst disease-specific survival, independently of any other variables.

Conclusion

DLC-1 appeared to also play as a tumor suppressor gene in HCC. And DLC-1 might work its way by negatively regulating the expression of RhoA/ROCK2/ moesin. DLC-1 itself could be a direct indicator or marker for the invasion and prognosis of HCC.

Acknowledgements

This work was supported by grants from the Technique Foundation of Henan (152102310264).
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.

Competing interests

The authors declare that there are no conflicts of interest.

Authors’ contributions

LJ S and QL are responsible for laboratory work, data analysis and paper writing. XR M is responsible for laboratory work and data analysis. SL L is responsible for sample collection and clinical data analyses. LX W and QX F are responsible for project design. XY X is responsible for project design and manuscript preparation. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Cai ZQ, Si SB, Chen C, Zhao Y, Ma YY, Wang L, et al. Analysis of prognostic factors for survival after hepatectomy for hepatocellular carcinoma based on a bayesian network. PLoS One. 2015;10(3):e0120805.PubMedCentralCrossRefPubMed Cai ZQ, Si SB, Chen C, Zhao Y, Ma YY, Wang L, et al. Analysis of prognostic factors for survival after hepatectomy for hepatocellular carcinoma based on a bayesian network. PLoS One. 2015;10(3):e0120805.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Wang W, Li GY, Zhu JY, Huang DB, Zhou HC, Zhong W, et al. Overexpression of AGGF1 is correlated with angiogenesis and poor prognosis of hepatocellular carcinoma. Med Oncol. 2015;32(4):131.CrossRefPubMed Wang W, Li GY, Zhu JY, Huang DB, Zhou HC, Zhong W, et al. Overexpression of AGGF1 is correlated with angiogenesis and poor prognosis of hepatocellular carcinoma. Med Oncol. 2015;32(4):131.CrossRefPubMed
3.
Zurück zum Zitat Będkowska GE, Ławicki S, Gacuta E, Pawlowski P, Szmitkowski M. M-CSF in a new biomarker panel with HE4 and CA 125 in the diagnostics of epithelial ovarian cancerpatients. J Ovarian Res. 2015;8(1):27.PubMedCentralCrossRefPubMed Będkowska GE, Ławicki S, Gacuta E, Pawlowski P, Szmitkowski M. M-CSF in a new biomarker panel with HE4 and CA 125 in the diagnostics of epithelial ovarian cancerpatients. J Ovarian Res. 2015;8(1):27.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Holeiter G, Heering J, Erlmann P, Schmid S, Jähne R, Olayioye MA. Deleted in liver cancer 1 controls cell migration through a Dia1-dependent signaling pathway. Cancer Res. 2008;68(21):8743–51.CrossRefPubMed Holeiter G, Heering J, Erlmann P, Schmid S, Jähne R, Olayioye MA. Deleted in liver cancer 1 controls cell migration through a Dia1-dependent signaling pathway. Cancer Res. 2008;68(21):8743–51.CrossRefPubMed
5.
Zurück zum Zitat Ullmannova V, Popescu NC. Inhibition of cell proliferation, induction of apoptosis, reactivation of DLC1, and modulation of other gene expression by dietary flavone in breast cancer cell lines. Cancer Detect Prev. 2007;31(2):110–8.PubMedCentralCrossRefPubMed Ullmannova V, Popescu NC. Inhibition of cell proliferation, induction of apoptosis, reactivation of DLC1, and modulation of other gene expression by dietary flavone in breast cancer cell lines. Cancer Detect Prev. 2007;31(2):110–8.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Braun AC, Olayioye MA. Rho regulation: DLC proteins in space and time. Cell Signal. 2015;27(8):1643-51. Braun AC, Olayioye MA. Rho regulation: DLC proteins in space and time. Cell Signal. 2015;27(8):1643-51.
7.
Zurück zum Zitat Ravi A, Kaushik S, Ravichandran A, Pan CQ, Low BC . Epidermal growth factor activates the Rho GTPase-activating protein (GAP) Deleted in Liver Cancer 1 via focal adhesion kinase and protein phosphatase 2A. J Biol Chem. 2015;290(7):4149–62.CrossRefPubMed Ravi A, Kaushik S, Ravichandran A, Pan CQ, Low BC . Epidermal growth factor activates the Rho GTPase-activating protein (GAP) Deleted in Liver Cancer 1 via focal adhesion kinase and protein phosphatase 2A. J Biol Chem. 2015;290(7):4149–62.CrossRefPubMed
8.
Zurück zum Zitat Yuan BZ, Jefferson AM, Baldwin KT, Thorgeirsson SS, Popescu NC, Reynolds SH. DLC-1 operates as a tumor suppressor gene in human non-small cell lung carcinomas. Oncogene. 2004;23(7):1405–11.CrossRefPubMed Yuan BZ, Jefferson AM, Baldwin KT, Thorgeirsson SS, Popescu NC, Reynolds SH. DLC-1 operates as a tumor suppressor gene in human non-small cell lung carcinomas. Oncogene. 2004;23(7):1405–11.CrossRefPubMed
9.
Zurück zum Zitat Zhang J, He X, Ma Y, Liu Y, Shi H, Guo W, et al. Overexpression of ROCK1 and ROCK2 inhibits human laryngeal squamous cell carcinoma. Int J Clin Exp Pathol. 2015;8(1):244–51.PubMedCentralPubMed Zhang J, He X, Ma Y, Liu Y, Shi H, Guo W, et al. Overexpression of ROCK1 and ROCK2 inhibits human laryngeal squamous cell carcinoma. Int J Clin Exp Pathol. 2015;8(1):244–51.PubMedCentralPubMed
10.
11.
Zurück zum Zitat Voorneveld PW, Kodach LL, Jacobs RJ, Liv N, Zonnevylle AC, Hoogenboom JP, et al. Loss of SMAD4 alters BMP signaling to promote colorectal cancer cell metastasis via activation of Rho and ROCK. Gastroenterol. 2014;147(1):196–208. e13.CrossRef Voorneveld PW, Kodach LL, Jacobs RJ, Liv N, Zonnevylle AC, Hoogenboom JP, et al. Loss of SMAD4 alters BMP signaling to promote colorectal cancer cell metastasis via activation of Rho and ROCK. Gastroenterol. 2014;147(1):196–208. e13.CrossRef
12.
Zurück zum Zitat Bhandary L, Whipple RA, Vitolo MI, Charpentier MS, Boggs AE, Chakrabarti KR, Thompson KN, Martin SS. ROCK inhibition promotes microtentacles that enhance reattachment of breast cancer cells. Oncotarget. 2015;6(8):6251–66.PubMedCentralCrossRefPubMed Bhandary L, Whipple RA, Vitolo MI, Charpentier MS, Boggs AE, Chakrabarti KR, Thompson KN, Martin SS. ROCK inhibition promotes microtentacles that enhance reattachment of breast cancer cells. Oncotarget. 2015;6(8):6251–66.PubMedCentralCrossRefPubMed
13.
14.
Zurück zum Zitat Lin L, Yang XM, Li J, Zhang YL, Qin W, Zhang ZG. Microfilament regulatory protein MENA increases activity of RhoA and promotes metastasis ofhepatocellular carcinoma. Exp Cell Res. 2014;327(1):113–22.CrossRefPubMed Lin L, Yang XM, Li J, Zhang YL, Qin W, Zhang ZG. Microfilament regulatory protein MENA increases activity of RhoA and promotes metastasis ofhepatocellular carcinoma. Exp Cell Res. 2014;327(1):113–22.CrossRefPubMed
15.
Zurück zum Zitat Wong CC, Wong CM, Ko FC, Chan LK, Ching YP, Yam JW, et al. Deleted in liver cancer 1 (DLC1) negatively regulates Rho/ROCK/MLC pathway in hepatocellularcarcinoma. PLoS One. 2008;3(7):e2779.PubMedCentralCrossRefPubMed Wong CC, Wong CM, Ko FC, Chan LK, Ching YP, Yam JW, et al. Deleted in liver cancer 1 (DLC1) negatively regulates Rho/ROCK/MLC pathway in hepatocellularcarcinoma. PLoS One. 2008;3(7):e2779.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Deming PB, Campbell SL, Stone JB, Rivard RL, Mercier AL, Howe AK. Anchoring of protein kinase A by ERM (ezrin-radixin-moesin) proteins is required for proper netrin signaling through DCC (deleted in colorectal cancer). J Biol Chem. 2015;290(9):5783–96.CrossRefPubMed Deming PB, Campbell SL, Stone JB, Rivard RL, Mercier AL, Howe AK. Anchoring of protein kinase A by ERM (ezrin-radixin-moesin) proteins is required for proper netrin signaling through DCC (deleted in colorectal cancer). J Biol Chem. 2015;290(9):5783–96.CrossRefPubMed
17.
Zurück zum Zitat Abiatari I, Esposito I, De Oliveira T, Felix K, Xin H, Penzel R et al. Moesin-dependent cytoskeleton remodelling is associated with an anaplastic phenotype of pancreatic cancer. J Cell Mol Med. 2010;14(5):1166–79.PubMedCentralPubMed Abiatari I, Esposito I, De Oliveira T, Felix K, Xin H, Penzel R et al. Moesin-dependent cytoskeleton remodelling is associated with an anaplastic phenotype of pancreatic cancer. J Cell Mol Med. 2010;14(5):1166–79.PubMedCentralPubMed
18.
Zurück zum Zitat DeSouza LV, Matta A, Karim Z, Mukherjee J, Wang XS, Krakovska O, et al. Role of moesin in hyaluronan induced cell migration in glioblastoma multiforme. Mol Cancer. 2013;12:74.PubMedCentralCrossRefPubMed DeSouza LV, Matta A, Karim Z, Mukherjee J, Wang XS, Krakovska O, et al. Role of moesin in hyaluronan induced cell migration in glioblastoma multiforme. Mol Cancer. 2013;12:74.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Mian H, Yang C, Wen L, Liu Q, Liu J, Huang J, et al. Vascular endothelial growth factor C promotes cervical cancer metastasis via up-regulation and activation of RhoA/ROCK-2/moesin cascade. BMC Cancer. 2010;10:170.CrossRef Mian H, Yang C, Wen L, Liu Q, Liu J, Huang J, et al. Vascular endothelial growth factor C promotes cervical cancer metastasis via up-regulation and activation of RhoA/ROCK-2/moesin cascade. BMC Cancer. 2010;10:170.CrossRef
20.
Zurück zum Zitat Smith SC, Baras AS, Owens CR, Dancik G, Theodorescu D. Transcriptional signatures of Ral GTPase are associated with aggressive clinicopathologic characteristics in human cancer. Cancer Res. 2012;72(14). doi: 10.1158/0008-5472. Smith SC, Baras AS, Owens CR, Dancik G, Theodorescu D. Transcriptional signatures of Ral GTPase are associated with aggressive clinicopathologic characteristics in human cancer. Cancer Res. 2012;72(14). doi: 10.​1158/​0008-5472.
21.
Zurück zum Zitat Chen L, Li M, Li Q, Wang CJ, Xie SQ. DKK1 promotes hepatocellular carcinoma cell migration and invasion through β-catenin/MMP7 signaling pathway. Mol Cancer. 2013;12:157.PubMedCentralCrossRefPubMed Chen L, Li M, Li Q, Wang CJ, Xie SQ. DKK1 promotes hepatocellular carcinoma cell migration and invasion through β-catenin/MMP7 signaling pathway. Mol Cancer. 2013;12:157.PubMedCentralCrossRefPubMed
22.
23.
Zurück zum Zitat Ko FC, Ping Yam JW. Regulation of deleted in liver cancer 1 tumor suppressor by protein-protein interactions and phosphorylation. Int J Cancer. 2014;135(2):264–9.CrossRefPubMed Ko FC, Ping Yam JW. Regulation of deleted in liver cancer 1 tumor suppressor by protein-protein interactions and phosphorylation. Int J Cancer. 2014;135(2):264–9.CrossRefPubMed
24.
Zurück zum Zitat Feng X, Li C, Liu W, Chen H, Zhou W, Wang L, et al. DLC-1, a candidate tumor suppressor gene, inhibits the proliferation, migration and tumorigenicity of human nasopharyngeal carcinoma cells. Int J Oncol. 2013;42(6):1973–84.PubMed Feng X, Li C, Liu W, Chen H, Zhou W, Wang L, et al. DLC-1, a candidate tumor suppressor gene, inhibits the proliferation, migration and tumorigenicity of human nasopharyngeal carcinoma cells. Int J Oncol. 2013;42(6):1973–84.PubMed
25.
26.
Zurück zum Zitat Goggs R, Williams CM, Mellor H, Poole AW. Platelet Rho GTPases-a focus on novel players, roles and relationships. Biochem J. 2015;466(3):431–442.PubMedCentralCrossRefPubMed Goggs R, Williams CM, Mellor H, Poole AW. Platelet Rho GTPases-a focus on novel players, roles and relationships. Biochem J. 2015;466(3):431–442.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Wang Y, Lei R, Zhuang X, Zhang N, Pan H, Li G, et al. DLC1-dependent parathyroid hormone-like hormone inhibition suppresses breast cancer bone metastasis. J Clin Invest. 2014;124(4):1646–59.PubMedCentralCrossRefPubMed Wang Y, Lei R, Zhuang X, Zhang N, Pan H, Li G, et al. DLC1-dependent parathyroid hormone-like hormone inhibition suppresses breast cancer bone metastasis. J Clin Invest. 2014;124(4):1646–59.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Liao W, Huang W, Guo Y, Xin M, Fu X. Testosterone promotes vascular endothelial cell migration via upregulation of ROCK-2/moesincascade. Mol Biol Rep. 2013;40(12):6729–635.CrossRefPubMed Liao W, Huang W, Guo Y, Xin M, Fu X. Testosterone promotes vascular endothelial cell migration via upregulation of ROCK-2/moesincascade. Mol Biol Rep. 2013;40(12):6729–635.CrossRefPubMed
30.
Zurück zum Zitat Wang J, Liu H, Chen B, Li Q, Huang X, Wang L, et al. RhoA/ROCK-dependent moesin phosphorylation regulates AGE-induced endothelial cellular response. Cardiovasc Diabetol. 2012;11:7.PubMedCentralCrossRefPubMed Wang J, Liu H, Chen B, Li Q, Huang X, Wang L, et al. RhoA/ROCK-dependent moesin phosphorylation regulates AGE-induced endothelial cellular response. Cardiovasc Diabetol. 2012;11:7.PubMedCentralCrossRefPubMed
Metadaten
Titel
DLC-1 is an independent prognostic marker and potential therapeutic target in hepatocellular cancer
verfasst von
L. J. Song
Q. Liu
X. R. Meng
SH. L Li
L. X. Wang
Q. X. Fan
X. Y. Xuan
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Diagnostic Pathology / Ausgabe 1/2016
Elektronische ISSN: 1746-1596
DOI
https://doi.org/10.1186/s13000-016-0470-x

Weitere Artikel der Ausgabe 1/2016

Diagnostic Pathology 1/2016 Zur Ausgabe

Neu im Fachgebiet Pathologie