1
|
You HJ, Li Q, Ma LH, Wang X, Zhang HY, Wang YX, Bao ES, Zhong YJ, Kong DL, Liu XY, Kong FY, Zheng KY, Tang RX. Inhibition of GLUD1 mediated by LASP1 and SYVN1 contributes to hepatitis B virus X protein-induced hepatocarcinogenesis. J Mol Cell Biol 2024; 16:mjae014. [PMID: 38587834 PMCID: PMC11440430 DOI: 10.1093/jmcb/mjae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 01/18/2024] [Accepted: 04/06/2024] [Indexed: 04/09/2024] Open
Abstract
Glutamate dehydrogenase 1 (GLUD1) is implicated in oncogenesis. However, little is known about the relationship between GLUD1 and hepatocellular carcinoma (HCC). In the present study, we demonstrated that the expression levels of GLUD1 significantly decreased in tumors, which was relevant to the poor prognosis of HCC. Functionally, GLUD1 silencing enhanced the growth and migration of HCC cells. Mechanistically, the upregulation of interleukin-32 through AKT activation contributes to GLUD1 silencing-facilitated hepatocarcinogenesis. The interaction between GLUD1 and AKT, as well as α-ketoglutarate regulated by GLUD1, can suppress AKT activation. In addition, LIM and SH3 protein 1 (LASP1) interacts with GLUD1 and induces GLUD1 degradation via the ubiquitin-proteasome pathway, which relies on the E3 ubiquitin ligase synoviolin (SYVN1), whose interaction with GLUD1 is enhanced by LASP1. In hepatitis B virus (HBV)-related HCC, the HBV X protein (HBX) can suppress GLUD1 with the participation of LASP1 and SYVN1. Collectively, our data suggest that GLUD1 silencing is significantly associated with HCC development, and LASP1 and SYVN1 mediate the inhibition of GLUD1 in HCC, especially in HBV-related tumors.
Collapse
Affiliation(s)
- Hong-Juan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Qi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
- Laboratory Department, The People's Hospital of Funing, Yancheng 224400, China
| | - Li-Hong Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xing Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Huan-Yang Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu-Xin Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - En-Si Bao
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu-Jie Zhong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - De-Long Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiang-Ye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Fan-Yun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou 221004, China
| | - Ren-Xian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
2
|
Niimi A, Limsirichaikul S, Kano K, Mizutani Y, Takeuchi T, Sawangsri P, Tran DQ, Kawamoto Y, Suzuki M. LASP1, CERS6, and Actin Form a Ternary Complex That Promotes Cancer Cell Migration. Cancers (Basel) 2023; 15:2781. [PMID: 37345118 DOI: 10.3390/cancers15102781] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
CERS6 is associated with metastasis and poor prognosis in non-small cell lung cancer (NSCLC) patients through d18:1/C16:0 ceramide (C16 ceramide)-mediated cell migration, though the detailed mechanism has not been elucidated. In the present study, examinations including co-immunoprecipitation, liquid chromatography, and tandem mass spectrometry analysis were performed to identify a novel binding partner of CERS6. Among the examined candidates, LASP1 was a top-ranked binding partner, with the LIM domain possibly required for direct interaction. In accord with those findings, CERS6 and LASP1 were found to co-localize on lamellipodia in several lung cancer cell lines. Furthermore, silencing of CERS6 and/or LASP1 significantly suppressed cell migration and lamellipodia formation, whereas ectopic addition of C16 ceramide partially rescued those phenotypes. Both LASP1 and CERS6 showed co-immunoprecipitation with actin, with those interactions markedly reduced when the LASP1-CERS6 complex was abolished. Based on these findings, it is proposed that LASP1-CERS6 interaction promotes cancer cell migration.
Collapse
Affiliation(s)
- Atsuko Niimi
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
| | - Siripan Limsirichaikul
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
- Department of Biopharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Keiko Kano
- Institute of Transformative Bio-Molecules (WPI-ItbM), Nagoya University, Chikusa, Nagoya 464-8602, Japan
| | - Yasuyoshi Mizutani
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
| | - Toshiyuki Takeuchi
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
| | - Patinya Sawangsri
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
| | - Dat Quoc Tran
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
| | - Yoshiyuki Kawamoto
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Motoshi Suzuki
- Department of Molecular Oncology, Fujita Health University, Toyoake 470-1192, Japan
| |
Collapse
|
3
|
Ma Q, Lu Q, Lei X, Zhao J, Sun W, Wang J, Zhu Q, Huang D. UCHL3 promotes hepatocellular carcinoma cell migration by de-ubiquitinating and stabilizing Vimentin. Front Oncol 2023; 13:1088475. [PMID: 36969045 PMCID: PMC10036040 DOI: 10.3389/fonc.2023.1088475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
BackgroundHepatocellular carcinoma (HCC) is a common malignant tumor associated with a poor prognosis. Ubiquitin carboxyl-terminal hydrolase L3 (UCHL3) has been reported to promote diverse tumors, but little is known about its role in HCC.MethodsExpression levels of UCHL3 in Huh7 and Hep3B cells were measured by qRT-PCR. UCHL3, Vimentin protein levels, and ubiquitination levels were determined by Western blot assay. co-immunoprecipitation, Immunofluorescence, and IHC were used to detect the interaction and expression association between UCHL3 and Vimentin in the cells. Wound healing and Transwell assays were used to measure cell migration. Spheroid formation assay were used to assess stem-like properties.ResultsUCHL3 expression was found to be significantly elevated in HCC and associated with poor prognosis. UCHL3 promoted migration and stem-like properties of HCC cells. Vimentin was identified as a potential de-ubiquitination substrate of UCHL3 and UCHL3 interacted with and promoted the de-ubiquitination of Vimentin, enhancing its stability. Moreover, the suppression of UCHL3 by siRNA or the inhibition by TCID upregulated ubiquitinated Vimentin. Vimentin attenuated the suppression of cell migration caused by knockdown of UCHL3.ConclusionUCHL3 was highly expressed in HCC and functioned as an oncogene. Vimentin is a novel substrate of UCHL3 and its stabilization and de-ubiquitination enhanced HCC cell migration.
Collapse
Affiliation(s)
- Qiancheng Ma
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qiliang Lu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiangxiang Lei
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Wen Sun
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Wang
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qing Zhu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- *Correspondence: Qing Zhu, ; Dongsheng Huang,
| | - Dongsheng Huang
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Qing Zhu, ; Dongsheng Huang,
| |
Collapse
|
4
|
Grossi I, Schiavone M, Cannone E, Grejdan OA, Tobia C, Bonomini F, Rezzani R, Salvi A, De Petro G. Lasp1 Expression Is Implicated in Embryonic Development of Zebrafish. Genes (Basel) 2022; 14:genes14010035. [PMID: 36672776 PMCID: PMC9858601 DOI: 10.3390/genes14010035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The LIM and SH3 domain protein 1 (LASP1) was originally identified in metastatic breast cancer and mainly characterized as a cytoskeleton protein overexpressed in various cancer types. At present, little is known about LASP1 expression in physiological conditions, and its function during embryonic development has not been elucidated. Here, we focused on Lasp1 and embryonic development, choosing zebrafish as a vertebrate model. For the first time, we identified and determined the expression of Lasp1 protein at various stages of development, at 48 and 72 h post-fertilization (hpf), at 6 days pf and in different organs of zebrafish adults by Western blotting, 3D light-sheet microscopy and fluorescent immunohistochemistry. Further, we showed that specific lasp1 morpholino (MO) led to (i) abnormal morphants with alterations in several organs, (ii) effective knockdown of endogenous Lasp1 protein and (iii) an increase in lasp1 mRNA, as detected by ddPCR. The co-injection of lasp1 mRNA with lasp1 MO partially rescued morphant phenotypes, thus confirming the specificity of the MO oligonucleotide-induced defects. We also detected an increase in apoptosis following lasp1 MO treatment. Our results suggest a significant role for Lasp1 in embryonic development, highlighting zebrafish as a vertebrate model suitable for studying Lasp1 function in developmental biology and organogenesis.
Collapse
Affiliation(s)
- Ilaria Grossi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, 25123 Brescia, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, 25123 Brescia, Italy
| | - Elena Cannone
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, 25123 Brescia, Italy
| | - Oana Andreea Grejdan
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, 25123 Brescia, Italy
| | - Chiara Tobia
- Department of Molecular and Translational Medicine, Division of Experimental Oncology and Immunology, University of Brescia, 25123 Brescia, Italy
| | - Francesca Bonomini
- Department of Clinical and Experimental Sciences, Division of Anatomy and Physiopathology, University of Brescia, 25123 Brescia, Italy
| | - Rita Rezzani
- Department of Clinical and Experimental Sciences, Division of Anatomy and Physiopathology, University of Brescia, 25123 Brescia, Italy
| | - Alessandro Salvi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, 25123 Brescia, Italy
- Correspondence:
| | - Giuseppina De Petro
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
5
|
Vanderheijden C, Vaessen T, Yakkioui Y, Riedl R, Temel Y, Hovinga K, Hoogland G. LIM and SH3 protein 1 (LASP1) differentiates malignant chordomas from less malignant chondrosarcomas. J Neurooncol 2022; 158:81-88. [PMID: 35507100 PMCID: PMC9166821 DOI: 10.1007/s11060-022-04012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/06/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Chordomas are malignant tumors that develop along the neuraxis between skull-base and sacrum. Chondrosarcomas show similarities with chordomas, yet show less malignant behavior. LIM and SH3 protein 1 (LASP1) is a cytoskeletal protein known to promote the malignant behavior of tumors. LASP1 was previously identified as a possibly overexpressed protein in a chordoma proteomics experiment. In this study we compare LASP1 expression in chordoma and chondrosarcoma tissue. METHODS Biopsies of primary tumors were collected from surgically treated chordoma (n = 6) and chondrosarcoma (n = 6) patients, flash-frozen upon collection and collectively analyzed for LASP1 RNA (real-time PCR) and protein expression (western blotting). Additionally, tissue micro array (TMA)-based immunohistochemistry was applied to an archive of 31 chordoma and 1 chondrosarcoma specimen. RESULTS In chordoma samples, LASP1 mRNA was detected in 4/6 cases and a strong 36 kDa immunoreactive protein band was observed in 4/5 cases. In contrast, 0/6 chondrosarcoma samples showed detectable levels of LASP1 mRNA and only a weak 36 kDa band was observed in 4/5 cases. Immunohistochemical analysis showed LASP1 expression in all chordoma samples, whereas chondrosarcoma specimen did not show immunoreactivity. CONCLUSION LASP1 is strongly expressed in the majority of chordoma cases and shows low expression in chondrosarcoma tissue. Since LASP1 is known to function as oncogene and regulate cell proliferation in other tumor types, this study implicates a role for LASP1 in chordoma biology. Further studies are warranted to improve understanding of LASP1's expression and functioning within chordoma, both in vitro and in vivo.
Collapse
Affiliation(s)
- Cas Vanderheijden
- Department of Neurosurgery, Maastricht University Medical Center, PO Box 5800, 6202 AZ, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Thomas Vaessen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Youssef Yakkioui
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neurosurgery, Noordwest Hospital, Alkmaar, The Netherlands
| | - Robert Riedl
- Department of Pathology, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, PO Box 5800, 6202 AZ, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Koos Hovinga
- Department of Neurosurgery, Maastricht University Medical Center, PO Box 5800, 6202 AZ, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Center, PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
6
|
LASP-1 interacts with ErbB2 in ovarian cancer cells. Biochem J 2021; 479:23-38. [PMID: 34881777 DOI: 10.1042/bcj20210173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022]
Abstract
LASP-1 was identified as a protein following mass spectrometric analysis of phosphoproteins consequent to signaling by ErbB2 in SKOV-3 cells. It has been previously identified as an oncogene and is located on chromosomal arm 17q 0.76Mb centromeric to ErbB2. It is expressed in serous ovarian cancer cell lines as a 40kDa protein. In SKOV-3 cells, it was phosphorylated and was inhibited by Lapatinib and CP7274714. LASP-1 co-immunoprecipitated with ErbB2 in SKOV-3 cells, suggesting a direct interaction. This interaction and phosphorylation were independent of the kinase activity of ErbB2. Moreover, the binding of LASP-1 to ErbB2 was independent of the tyrosine phosphorylation of LASP-1. LASP-1 was neither expressed on the surface epithelium of the normal ovary nor in the fallopian tube. It was expressed in 28% of ovarian tumours (n=101) that did not significantly correlate with other clinical factors. In tumours from patients with invasive ductal carcinoma of the breast who had ErbB2 amplification (3+), LASP-1 was expressed in 3/20 (p <0.001). Analysis of the expression of an independent dataset of ovarian and breast tumors from TCGA showed the significant co-occurrence of ErbB2 and LASP-1 (p<0.01). These results suggest that LASP-1 and ErbB2 interaction could be important in the pathogenesis of ovarian cancer.
Collapse
|
7
|
Liao Q, Ren Y, Yang Y, Zhu X, Zhi Y, Zhang Y, Chen Y, Ding Y, Zhao L. CCT8 recovers WTp53-suppressed cell cycle evolution and EMT to promote colorectal cancer progression. Oncogenesis 2021; 10:84. [PMID: 34862361 PMCID: PMC8642402 DOI: 10.1038/s41389-021-00374-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 11/09/2022] Open
Abstract
LIM and SH3 protein 1 (LASP1) is a metastasis-related protein reported to enhance tumor progression in colorectal cancer (CRC). However, the underlying mechanism is still elusive. The chaperonin protein containing TCP1 (CCT) is a cellular molecular chaperone complex, which is necessary for the correct folding of many proteins. It contains eight subunits, CCT1-8. CCT8 is overexpressed in many cancers, however, studies on CCT8 are limited and its role on CRC development and progression remains elusive. In this study, we confirmed that CCT8 and LASP1 can interact with each other and express positively in CRC cells. CCT8 could recover the ability of LASP1 to promote the invasion of CRC; CCT8 could significantly promote the proliferation, invasion, and metastasis of colorectal cells in vivo and in vitro. Mechanically, CCT8 inhibited the entry of WTp53 into the nucleus, and there was a negative correlation between the expression of CCT8 and the nuclear expression of WTp53 in clinical colorectal tissues. CCT8 promoted the cell cycle evolution and EMT progression of CRC by inhibiting the entry of WTp53 into the nucleus. Clinically, CCT8 was highly expressed in CRC. More importantly, the overall survival of CRC patients with high expression of CCT8 was worse than that of patients with low expression of CCT8. These findings indicate that as LASP1-modulated proteins, CCT8 plays a key role in promoting the progression of colorectal cancer, which provides a potential target for clinical intervention in patients with colorectal cancer.
Collapse
Affiliation(s)
- Qing Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China
| | - Yun Ren
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China.,Department of Pathology, Affiliated Tumor Hospital of Guangzhou, Medical University, Guangzhou, China
| | - Yuyi Yang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiaohui Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China
| | - Yunfei Zhi
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China
| | - Yujie Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China
| | - Yi Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China.
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong province, People's Republic of China.
| |
Collapse
|
8
|
You H, Yuan D, Bi Y, Zhang N, Li Q, Tu T, Wei X, Lian Q, Yu T, Kong D, Yang X, Liu X, Liu X, Kong F, Zheng K, Tang R. Hepatitis B virus X protein promotes vimentin expression via LIM and SH3 domain protein 1 to facilitate epithelial-mesenchymal transition and hepatocarcinogenesis. Cell Commun Signal 2021; 19:33. [PMID: 33722250 PMCID: PMC7958410 DOI: 10.1186/s12964-021-00714-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) X protein (HBX) has been reported to be responsible for the epithelial-mesenchymal transition (EMT) in HBV-related hepatocellular carcinoma (HCC). Vimentin is an EMT-related molecular marker. However, the importance of vimentin in the pathogenesis of HCC mediated by HBX has not been well determined. METHODS The expression of vimentin induced by HBX, and the role of LIM and SH3 domain protein 1 (LASP1) in HBX-induced vimentin expression in hepatoma cells were examined by western blot and immunohistochemistry analysis. Both the signal pathways involved in the expression of vimentin, the interaction of HBX with vimentin and LASP1, and the stability of vimentin mediated by LASP1 in HBX-positive cells were assessed by western blot, Co-immunoprecipitation, and GST-pull down assay. The role of vimentin in EMT, proliferation, and migration of HCC cells mediated by HBX and LASP1 were explored with western blot, CCK-8 assay, plate clone formation assay, transwell assay, and wound healing assay. RESULTS Vimentin expression was increased in both HBX-positive hepatoma cells and HBV-related HCC tissues, and the expression of vimentin was correlated with HBX in HBV-related HCC tissues. Functionally, vimentin was contributed to the EMT, proliferation, and migration of hepatoma cells mediated by HBX. The mechanistic analysis suggested that HBX was able to enhance the expression of vimentin through LASP1. On the one hand, PI3-K, ERK, and STAT3 signal pathways were involved in the upregulation of vimentin mediated by LASP1 in HBX-positive hepatoma cells. On the other hand, HBX could directly interact with vimentin and LASP1, and dependent on LASP1, HBX was capable of promoting the stability of vimentin via protecting it from ubiquitination mediated protein degradation. Besides these, vimentin was involved in the growth and migration of hepatoma cells mediated by LASP1 in HBX-positive hepatoma cells. CONCLUSION Taken together, these findings demonstrate that, dependent on LASP1, vimentin is crucial for HBX-mediated EMT and hepatocarcinogenesis, and may serve as a potential target for HBV-related HCC treatment. Video abstract.
Collapse
Affiliation(s)
- Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Dongchen Yuan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Yanwei Bi
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
- Clinical Laboratory, Xuzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, Jiangsu People’s Republic of China
| | - Ning Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Qi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Tao Tu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Xiao Wei
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Qi Lian
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Tong Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Delong Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Xiaomei Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| |
Collapse
|
9
|
The CXCR4-Dependent LASP1-Ago2 Interaction in Triple-Negative Breast Cancer. Cancers (Basel) 2020; 12:cancers12092455. [PMID: 32872485 PMCID: PMC7564666 DOI: 10.3390/cancers12092455] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/28/2022] Open
Abstract
The CXCR4-LASP1 axis is an emerging target in the field of breast cancer metastasis. C-X-C chemokine receptor type 4 (CXCR4) mediates directed cell migration when activated by its cognate ligand CXCL12. LIM and SH3 Protein 1 (LASP1) is a critical node in the CXCR4 signaling pathway, as its deficiency blocks CXCR4-dependent Matrigel invasion. The mechanism by which LASP1 facilitates this invasive ability of tumor cells when CXCR4 is activated is unknown. Our previous proteomics work had revealed several components of the RNA interference (RNAi) machinery as being potential LASP1 interacting proteins. Here we report that argonaute 2 (Ago2), a protein with central involvement in RNAi, associates with LASP1 in triple-negative breast cancer (TNBC) cells. We demonstrate that LASP1 co-immunoprecipitates with Ago2 endogenously in a CXCL12-dependent manner, with further confirmation of this interaction by proximity ligation assay. Furthermore, this association is specific to CXCR4 as it can be abrogated by the CXCR4 antagonist, AMD3465. By GST-pulldown approach, we identify that LASP1 directly binds to Ago2 through its LIM and SH3 domains, and that this binding is dictated by the S146 and Y171 phosphorylation sites of LASP1. Additionally, the phosphorylation status of LASP1 affected tumor suppressor microRNA (miRNA) Let-7a-guided Ago2 activity. Levels of several endogenous targets of Let-7a were found to be altered including C-C chemokine receptor type 7 (CCR7), which is another critical chemokine receptor involved in metastasis to lymph nodes. Our results suggest a novel role for the LASP1-Ago2 module in shaping the RNAi landscape, functionally impacting the invasive ability of cancer cells.
Collapse
|
10
|
LASP1 interacts with N-WASP to activate the Arp2/3 complex and facilitate colorectal cancer metastasis by increasing tumour budding and worsening the pattern of invasion. Oncogene 2020; 39:5743-5755. [PMID: 32704133 DOI: 10.1038/s41388-020-01397-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/16/2020] [Indexed: 12/26/2022]
Abstract
LIM and SH3 protein 1 (LASP1) is a metastasis-related protein reported to enhance tumour progression in colorectal cancer (CRC). However, the underlying mechanism is still elusive. As the major biological and pathological functions of LASP1 are accomplished by its LIM and SH3 domains via protein-protein interactions, a yeast two-hybrid system was employed to screen novel LASP1-interacting proteins. N-WASP, a member of the Wiskott-Aldrich syndrome protein (WASP) family, was screened and identified as a LASP1-interacting protein overexpressed in CRC tissues. N-WASP could stimulate the migration and invasion of CRC cells in vitro and increase the formation of subcutaneous tumours, mesenteric implanted tumours and hepatic metastatic tumours. N-WASP could interact with and activate the Arp2/3 complex to stimulate actin polymerization, thus changing the migratory and invasive capabilities of CRC cells. The interaction of LASP1 with N-WASP did not influence the expression of N-WASP but recovered the reduced actin polymerization induced by N-WASP silencing. High N-WASP expression was detected in most clinical colorectal samples, and it was positively correlated with the expression of LASP1 and ARP3, as well as the tumour budding and pattern of invasion, but negatively correlated with host lymphocytic response. Our study suggests a new mechanism for LASP1-mediated CRC metastasis determined by exploring LASP1-interacting proteins and identifies N-WASP as a potential therapeutic target for CRC.
Collapse
|
11
|
The Expressions and Mechanisms of Sarcomeric Proteins in Cancers. DISEASE MARKERS 2020; 2020:8885286. [PMID: 32670437 PMCID: PMC7346232 DOI: 10.1155/2020/8885286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
The sarcomeric proteins control the movement of cells in diverse species, whereas the deregulation can induce tumours in model organisms and occurs in human carcinomas. Sarcomeric proteins are recognized as oncogene and related to tumor cell metastasis. Recent insights into their expressions and functions have led to new cancer therapeutic opportunities. In this review, we appraise the evidence for the sarcomeric proteins as cancer genes and discuss cancer-relevant biological functions, potential mechanisms by which sarcomeric proteins activity is altered in cancer.
Collapse
|
12
|
Chen Q, Wu K, Qin X, Yu Y, Wang X, Wei K. LASP1 promotes proliferation, metastasis, invasion in head and neck squamous cell carcinoma and through direct interaction with HSPA1A. J Cell Mol Med 2019; 24:1626-1639. [PMID: 31793711 PMCID: PMC6991697 DOI: 10.1111/jcmm.14854] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 10/30/2019] [Accepted: 11/08/2019] [Indexed: 01/02/2023] Open
Abstract
LIM and SH3 protein 1 (LASP1) is a specific focal adhesion protein that promotes metastasis in a variety of tumours. However, its role in head and neck squamous cell carcinoma (HNSCC) has not been fully validated. The purpose of this study was to analyse the interaction of LASP1 and its binding partner in HNSCC. The expression of LASP1 and HSPA1A in HNSCC was analysed by real-time PCR and Western blot. The effects of LASP1 on the biology behaviour of HNSCC cell lines were observed in vivo and in vitro. Co-immunoprecipitation analysis was performed to confirm the interaction between LASP1 and HSPA1A. LASP1 was highly expressed in HNSCC and associated with poor prognosis for patients. LASP1 also promoted cell proliferation, colony formation, invasion and cell cycle G2/M phase transition. Heat shock protein family A member 1A (HSPA1A) is identified as a chaperone protein of LASP1 and co-localized in the cytoplasm. HSPA1A positively regulates the interaction of LASP1 with P-AKT and enhances the malignant behaviour of HNSCC cells. LASP1 and HSPA1A are both up-regulated in HNSCC, and directly binds to each other. Double inhibition of LASP1 and HSPA1A expression may be an effective method for the treatment of HNSCC.
Collapse
Affiliation(s)
- Qi Chen
- Department of Stomatology Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kun Wu
- Department of Oral and Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing Qin
- Department of Oral and Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youcheng Yu
- Department of Stomatology of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Wang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kuijie Wei
- Department of Stomatology Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Stacchiotti A, Grossi I, García-Gómez R, Patel GA, Salvi A, Lavazza A, De Petro G, Monsalve M, Rezzani R. Melatonin Effects on Non-Alcoholic Fatty Liver Disease Are Related to MicroRNA-34a-5p/Sirt1 Axis and Autophagy. Cells 2019; 8:cells8091053. [PMID: 31500354 PMCID: PMC6770964 DOI: 10.3390/cells8091053] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Melatonin, an indole produced by pineal and extrapineal tissues, but also taken with a vegetarian diet, has strong anti-oxidant, anti-inflammatory and anti-obesogenic potentials. Non-alcoholic fatty liver disease (NAFLD) is the hepatic side of the metabolic syndrome. NAFLD is a still reversible phase but may evolve into steatohepatitis (NASH), cirrhosis and carcinoma. Currently, an effective therapy for blocking NAFLD staging is lacking. Silent information regulator 1 (SIRT1), a NAD+ dependent histone deacetylase, modulates the energetic metabolism in the liver. Micro-RNA-34a-5p, a direct inhibitor of SIRT1, is an emerging indicator of NAFLD grading. Thus, here we analyzed the effects of oral melatonin against NAFLD and underlying molecular mechanisms, focusing on steatosis, ER stress, mitochondrial shape and autophagy. Male C57BL/6J (WT) and SIRT1 heterozygous (HET) mice were placed either on a high-fat diet (58.4% energy from lard) (HFD) or on a standard maintenance diet (8.4% energy from lipids) for 16 weeks, drinking melatonin (10 mg/kg) or not. Indirect calorimetry, glucose tolerance, steatosis, inflammation, ER stress, mitochondrial changes, autophagy and microRNA-34a-5p expression were estimated. Melatonin improved hepatic metabolism and steatosis, influenced ER stress and mitochondrial shape, and promoted autophagy in WT HFD mice. Conversely, melatonin was ineffective in HET HFD mice, maintaining NASH changes. Indeed, autophagy was inconsistent in HET HFD or starved mice, as indicated by LC3II/LC3I ratio, p62/SQSTM1 and autophagosomes estimation. The beneficial role of melatonin in dietary induced NAFLD/NASH in mice was related to reduced expression of microRNA-34a-5p and sterol regulatory element-binding protein (SREBP1) but only in the presence of full SIRT1 availability.
Collapse
Affiliation(s)
- Alessandra Stacchiotti
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartmental University Center of Research "Adaptation and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Ilaria Grossi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Raquel García-Gómez
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), 28029 Madrid, Spain.
| | | | - Alessandro Salvi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Antonio Lavazza
- Instituto Zooprofilattico Sperimentale della Lombardia ed Emilia-Romagna (IZSLER), 25124 Brescia, Italy.
| | - Giuseppina De Petro
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), 28029 Madrid, Spain.
| | - Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartmental University Center of Research "Adaptation and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
14
|
Zhou S, Ma X, Wang ZJ, Zhang WY, Jiang H, Li SD, Zhang TZ, Du J, Lu Z. Research on the establishment of a TPM3 monoclonal stable transfected PANC-1 cell line and the experiment of the EMT occurrence in human pancreatic cancer. Onco Targets Ther 2019; 12:5577-5587. [PMID: 31371995 PMCID: PMC6628969 DOI: 10.2147/ott.s212689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/21/2019] [Indexed: 01/16/2023] Open
Abstract
Background: Pancreatic cancer is one of the most aggressive human malignancies that is associated with early metastasis and chemoresistance. Tropomyosin (TPM) is an indispensable regulatory protein for muscle contraction, Abnormal expressions of TPM gene are closely related to the carcinogenesis and metastasis of malignant tumors. Purpose: In this experiment, a monoclonal stable transfected cell line was established by the knock-down of TMP3 expression in PANC-1 cells with the lentivirus method, and the impacts of the downregulated TPM3 gene expression on the EMT-related molecules and biological behaviors of PANC-1 cells were explored. Methods: Based on the TPM3 gene sequence, we designed the RNA interference sequence, constructed and screened out the recombinant plasmid segment TPM3-shRNA with the optimal silencing effect, and carried out lentivirus titer determination and packaging. The recombinant lentiviral interference vector LV-TPM3-shRNA was transfected into PANC-1 cells; the transfection efficiency was then evaluated to screen out the monoclonal stable transfected PANC-1 cell line with downregulated TPM3 expression. The qRT-PCR and Western blot were used to detect the changes in the gene- and protein-levels expressions of EMT-related transcription factors in the target cell line and to respectively test the variations of the invasion and proliferation capacities. Results: It is shown that the monoclonal stable transfected PANC-1 cell line with downregulated TPM3 expression was successfully established with the recombinant lentiviral vector. After knocking down the expression of TPM3 gene in PANC-1 cells, EMT occurred in the cells; the cell phenotype showed malignant transformation, and the in vitro biological behaviors of the cells (such as proliferation and invasion) were enhanced to different degrees. Conclusion: It is indicated that the TPM3 gene can be a potential target spot for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Shuo Zhou
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Xiang Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Zhen-Jie Wang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Wei-Yue Zhang
- Department of Emergency Medicine, The Second People's Hospital of Bengbu City, Bengbu 233000, Anhui, People's Republic of China
| | - Hai Jiang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - San-Dang Li
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Tai-Zhe Zhang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Jie Du
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| | - Zheng Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, People's Republic of China
| |
Collapse
|
15
|
Zhou H, Xu J, Zhang C, Wen Y. Aberrant histone deacetylase 1 expression upregulates vimentin expression via an NF-κB-dependent pathway in hepatocellular carcinoma. Oncol Lett 2019; 18:339-347. [PMID: 31289505 PMCID: PMC6540068 DOI: 10.3892/ol.2019.10309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/12/2019] [Indexed: 12/14/2022] Open
Abstract
Aberrantly elevated expression levels of histone deacetylase 1 (HDAC1) and vimentin are closely associated with disease progression in hepatocellular carcinoma (HCC). It was previously demonstrated that knocking down expression of HDAC1 resulted in a concurrent decrease in the expression levels of vimentin. However, a causal link between these two proteins has not yet been demonstrated, to the best of our knowledge. In the present study, the association between HDAC1 and vimentin was investigated using an HDAC1 overexpression platform. HDAC1 and vimentin were significantly increased in HCC cells, and HDAC1 overexpression enhanced vimentin mRNA and protein expression levels in an HDAC1 dose-dependent manner. Subsequently, truncation and mutation of a vimentin promoter demonstrated that HDAC1-induced vimentin expression was dependent on a nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) binding site in the vimentin promoter sequence. Furthermore, HDAC1 induced vimentin expression by promoting NF-κB translocation between the cytoplasm and the nucleus, as opposed to modulating the total expression level of vimentin directly. The data in the present study demonstrated that HDAC1 is overexpressed in HCC and that HDAC1 may upregulate vimentin expression through the NF-κB signaling pathway, thus demonstrating a causal link between HDAC1 and vimentin in HCC, and may provide valuable information in understanding the pathogenesis of HCC.
Collapse
Affiliation(s)
- Huancheng Zhou
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Jiwei Xu
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Caiyun Zhang
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Yuanzhang Wen
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| |
Collapse
|
16
|
Zhang Y, Zhang L. Knockdown of LASP2 inhibits the proliferation, migration, and invasion of cervical cancer cells. J Cell Biochem 2019; 120:15389-15396. [PMID: 31026088 DOI: 10.1002/jcb.28806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/25/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Abstract
LIM and SH3 protein 2 (LASP2) belongs to nebulin family. It has been proven that LASP2 is involved in several cancers; however, its role in cervical cancer is unclear. Herein, we showed that LASP2 was highly expressed in cervical cancer tissues and cell lines. To knockdown LASP2 in cervical cancer cells, small interfering RNAs (siRNAs) targeting LASP2 (si-LASP2) were used. We found that cell proliferation, migration/invasion were markedly reduced after si-LASP2 transfection. A significant increase in E-cadherin expression, and decrease in N-cadherin and vimentin expressions were observed in si-LASP2 transfected cervical cancer cells. Knockdown of LASP2 caused significant inhibitory effect on the PI3K/Akt pathway. Treatment with the activator of the PI3K/Akt pathway, 740Y-P, abolished the effects of si-LASP2 transfection on cervical cancer cells. These findings suggested that LASP2 may be an oncogene through regulating the PI3K/Akt pathway in cervical cancer.
Collapse
Affiliation(s)
- Yimeng Zhang
- Department of Gynecology and Obstetrics, Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Liya Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| |
Collapse
|
17
|
Butt E, Raman D. New Frontiers for the Cytoskeletal Protein LASP1. Front Oncol 2018; 8:391. [PMID: 30298118 PMCID: PMC6160563 DOI: 10.3389/fonc.2018.00391] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
In the recent two decades, LIM and SH3 protein 1 (LASP1) has been developed from a simple actin-binding structural protein to a tumor biomarker and subsequently to a complex, nuclear transcriptional regulator. Starting with a brief historical perspective, this review will mainly compare and contrast LASP1 and LASP2 from the angle of the newest data and importantly, examine their role in transcriptional regulation. We will summarize the current knowledge through pictorial models and tables including the roles of different microRNAs in the differential regulation of LASP1 levels and patient outcome rather than specify in detail all tumor entities. Finally, the novel functional roles of LASP1 in secretion of vesicles, expression of matrix metalloproteinases and transcriptional regulation as well as the activation of survival and proliferation pathways in different cancer types are described.
Collapse
Affiliation(s)
- Elke Butt
- Institute for Experimental Biomedicine II, University Clinic, Wuerzburg, Germany
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
| |
Collapse
|
18
|
You H, Kong F, Zhou K, Wei X, Hu L, Hu W, Luo W, Kou Y, Liu X, Chen X, Zheng K, Tang R. HBX protein promotes LASP-1 expression through activation of c-Jun in human hepatoma cells. J Cell Physiol 2018; 233:7279-7291. [PMID: 29600594 DOI: 10.1002/jcp.26560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 02/21/2018] [Indexed: 12/15/2022]
Abstract
LIM and SH3 domain protein 1 (LASP-1) is known to participate in the progression of hepatocellular carcinoma (HCC). We previously showed that ectopic expression of hepatitis B virus (HBV) X protein (HBX) enhanced the expression of LASP-1, which promoted proliferation and migration of HCC cells. Here, we further demonstrated the molecular mechanism underlying upregulation of LASP-1, mediated by HBX, in HBV-infected HCC cells. Through a luciferase activity assay, we discovered that the LASP-1 promoter region regulated by HBX contained an AP-1 binding element in human hepatoma cells. Interestingly, c-Jun, one subunit of AP-1, was mainly responsible for activation, mediated by HBX, of the LASP-1 promoter. Furthermore, HBX was shown not only to interact with phosphorylated c-Jun in HCC cells but also to activate c-Jun by increasing the activation of PI3-K/JNK signaling. Chromatin immunoprecipitation (ChIP) assay demonstrated that HBX was capable of binding to the LASP-1 promoter with c-Jun. Further, the expression levels of HBX were shown to be significantly positively correlated with that of LASP-1 and phosphorylatedc-Jun in HBV-related HCC tissues by immunohistochemistry analysis. In addition, the N-terminus of HBX was found to be responsible for the activation of c-Jun, as well as the expression of LASP-1. Taken together, these results suggest that HBX contributes to LASP-1 expression via the activation of c-Jun to increase the promoter activity of LASP-1 in HBV-related HCC cells.
Collapse
Affiliation(s)
- Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kai Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao Wei
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenya Luo
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanbo Kou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaomei Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xi Chen
- Bio-pharmaceuticals (Collaboration Articulation Program), School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
19
|
Gao Q, Tang L, Wu L, Li K, Wang H, Li W, Wu J, Li M, Wang S, Zhao L. LASP1 promotes nasopharyngeal carcinoma progression through negatively regulation of the tumor suppressor PTEN. Cell Death Dis 2018. [PMID: 29531214 PMCID: PMC5847534 DOI: 10.1038/s41419-018-0443-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
LIM and SH3 protein 1 (LASP1) enhances tumor growth and metastasis in various cancers, but its role in nasopharyngeal carcinoma (NPC) remains unclear. Herein, we investigated the role of LASP1 in NPC and explored the underlying mechanisms in NPC. Clinically, overexpression of LASP1 is associated with tumor metastasis and poor prognosis of NPC patients. Gain-of-function and loss-of-function assays showed that LASP1 promoted NPC cell proliferation, metastasis, and invasion in vitro and in vivo. Mechanistically, we observed clear co-localization between LASP1 and PTEN in NPC cells. LASP1 interacted with PTEN and decreased the expression of PTEN in NPC. The ubiquitination assay indicated that LASP1 overexpression increased PTEN ubiquitination. PTEN was known as a tumor suppressor by negatively regulating phosphoinositide 3-kinase/AKT signaling pathway. Rescue experiments showed that PTEN weakened LASP1-mediated cell proliferation, migration, and invasive abilities and decreased the phosphorylation of AKT in NPC cells. Our findings suggest that LASP1 has a crucial role in NPC progression via LASP1/PTEN/AKT axis, highlighting LASP1 as a therapeutic target for NPC.
Collapse
Affiliation(s)
- Qingzu Gao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Lihua Tang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ling Wu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Kaitao Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Wang
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weidong Li
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Juan Wu
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingyi Li
- Radiotherapy Department, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Zhao H, Liu B, Li J. LIM and SH3 protein 1 knockdown suppresses proliferation and metastasis of colorectal carcinoma cells via inhibition of the mitogen-activated protein kinase signaling pathway. Oncol Lett 2018; 15:6839-6844. [PMID: 29731863 PMCID: PMC5920965 DOI: 10.3892/ol.2018.8222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/13/2017] [Indexed: 12/15/2022] Open
Abstract
LIM and SH3 protein 1 (Lasp-1), a focal adhesion protein, serves a critical role in the regulation of cell proliferation and migration. The role of Lasp-1, as well as the intracellular signaling pathways involved in metastasis and progression of colorectal carcinoma, remains unclear. In the present study, the regulatory effect of Lasp-1 and the underlying molecular mechanism involved in migration and invasion of colorectal carcinoma were investigated. RNA interference and overexpression in SW480 cells were performed to elucidate the role of Lasp-1. Reverse transcription-quantitative polymerase chain reaction, western blotting and immunofluorescence were conducted to determine the mRNA and protein levels of Lasp-1 and extracellular-signal-regulated kinase 1/2 (ERK1/2) in SW480 cells as well as tumor and adjacent normal tissues obtained from 20 patients with colorectal carcinoma. Furthermore, a cell proliferation assay, flow cytometric analysis, and cell migration and invasion assays were performed to examine the effect of Lasp-1 on cell growth. The results of the present study demonstrated that Lasp-1 and ERK1/2 were upregulated in tumor tissue compared with adjacent normal colorectal mucosa. Cell-based in vitro assays demonstrated that Lasp-1 knockdown suppressed the expression and activation of ERK1/2, whereas Lasp-1 overexpression resulted in ERK1/2 upregulation. Additionally, Lasp-1 knockdown inhibited cell proliferation, migration, and invasion and induced cellular apoptosis and G0/G1 cell-cycle arrest. The results of the present study indicate that Lasp-1 serves a critical role in the progression of colorectal carcinoma regulating the activation of the mitogen-activated protein kinase signaling pathway.
Collapse
Affiliation(s)
- Hongpeng Zhao
- Department of Gastrointestinal Surgery, Shandong Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Bo Liu
- Department of Gastrointestinal Surgery, Shandong Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Jie Li
- Department of Hepatobiliary Surgery, Shandong Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
21
|
Epigenetically regulated miR-145 suppresses colon cancer invasion and metastasis by targeting LASP1. Oncotarget 2018; 7:68674-68687. [PMID: 27626692 PMCID: PMC5356582 DOI: 10.18632/oncotarget.11919] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/25/2016] [Indexed: 11/25/2022] Open
Abstract
MiR-145 is a tumor-suppressive microRNA that participates in the malignant progression of colorectal cancer (CRC). Although miR-145 has been reported to inhibit proliferation and to induce apoptosis of CRC cells, the reports about its role in invasion and metastasis are controversial. The regulation of miR-145 its own expression also requires further elucidation. In this study, we firstly found that miR-145 is markedly downregulated in the metastatic tumors of CRC patients. Then through gain- and loss-of function studies, we demonstrated that miR-145 suppresses the invasion and metastasis of CRC cells. We also provided experimental evidences which include direct binding assays and verifications on tissue specimens to confirm that LIM and SH3 protein 1 (LASP1) is a direct target of miR-145. Furthermore, we identified the core promoter regions of miR-145 and observed the cooperation between histone methylation and transcription factors through binding to these core promoter regions to regulate the expression of miR-145 in CRC cells. Our study provides an insight into the regulatory network in CRC cells, thus offering new targets for treating CRC patients.
Collapse
|
22
|
Endres M, Kneitz S, Orth MF, Perera RK, Zernecke A, Butt E. Regulation of matrix metalloproteinases (MMPs) expression and secretion in MDA-MB-231 breast cancer cells by LIM and SH3 protein 1 (LASP1). Oncotarget 2018; 7:64244-64259. [PMID: 27588391 PMCID: PMC5325439 DOI: 10.18632/oncotarget.11720] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 08/09/2016] [Indexed: 01/27/2023] Open
Abstract
The process of tumor invasion requires degradation of extracellular matrix by proteolytic enzymes. Cancer cells form protrusive invadopodia, which produce and release matrix metalloproteinases (MMPs) to degrade the basement membrane thereby enabling metastasis. We investigated the effect of LASP1, a newly identified protein in invadopodia, on expression, secretion and activation of MMPs in invasive breast tumor cell lines. By analyzing microarray data of in-house generated control and LASP1-depleted MDA-MB-231 breast cancer cells, we observed downregulation of MMP1, -3 and -9 upon LASP1 depletion. This was confirmed by Western blot analysis. Conversely, rescue experiments restored in part MMP expression and secretion. The regulatory effect of LASP1 on MMP expression was also observed in BT-20 breast cancer cells as well as in prostate and bladder cancer cell lines. In line with bioinformatic FunRich analysis of our data, which mapped a high regulation of transcription factors by LASP1, public microarray data analysis detected a correlation between high LASP1 expression and enhanced c-Fos levels, a protein that is part of the transcription factor AP-1 and known to regulate MMP expression. Compatibly, in luciferase reporter assays, AP-1 showed a decreased transcriptional activity after LASP1 knockdown. Zymography assays and Western blot analysis revealed an additional promotion of MMP secretion into the extracellular matrix by LASP1, thus, most likely, altering the microenvironment during cancer progression. The newly identified role of LASP1 in regulating matrix degradation by affecting MMP transcription and secretion elucidated the migratory potential of LASP1 overexpressing aggressive tumor cells in earlier studies.
Collapse
Affiliation(s)
- Marcel Endres
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Susanne Kneitz
- Physiological Chemistry, Biozentrum, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Martin F Orth
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Ruwan K Perera
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Elke Butt
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
23
|
Hu S, Ran Y, Chen W, Zhang Y, Xu Y. MicroRNA-326 inhibits cell proliferation and invasion, activating apoptosis in hepatocellular carcinoma by directly targeting LIM and SH3 protein 1. Oncol Rep 2017; 38:1569-1578. [PMID: 28713953 DOI: 10.3892/or.2017.5810] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/26/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth-most common cancer and third leading cause of cancer-related deaths worldwide. Increasing evidence indicates that dysregulation of microRNAs is often observed in HCC, and has been extensively investigated in terms of cancer formation, progression, diagnosis, therapy, and prognosis. Recently, microRNA-326 (miR-326) has been demonstrated to play important roles in multiple types of human cancer. However, the expression pattern, clinical significance, roles and regulatory mechanisms of miR-326 in HCC have yet to be elucidated. In this study, miR-326 was frequently downregulated in HCC tissues and cell lines. Low miR-326 expression was significantly associated with the TNM stage, differentiation and lymph node metastasis of HCC patients. Further functional assays demonstrated that the recovered miR-326 expression inhibited HCC cell proliferation and invasion and activated cell apoptosis in vitro. In addition, LIM and SH3 protein 1 (LASP1) was identified as a direct target gene of miR-326 in HCC. Furthermore, LASP1 was upregulated in HCC tissues and cell lines. The expression level of LASP1 mRNA was inversely correlated with that of miR-326 in HCC tissues. Moreover, LASP1 silencing elicited effects similar to miR-326 overexpression on HCC cells, and LASP1 upregulation markedly reversed the effects of miR-326 overexpression on HCC cells. These results revealed that miR-326 suppressed the progression of HCC by directly targeting LASP1. Therefore, miR-326 may be used as a potential therapeutic target for the treatment of patients with HCC.
Collapse
Affiliation(s)
- Shiping Hu
- Department of Hepatology, Longgang Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518172, P.R. China
| | - Yun Ran
- Department of Hepatology, Longgang Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518172, P.R. China
| | - Wenlin Chen
- Department of Hepatology, Longgang Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518172, P.R. China
| | - Yuncheng Zhang
- Department of Hepatology, Longgang Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518172, P.R. China
| | - Yongjian Xu
- Department of Hepatology, Longgang Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
24
|
Zhang X, Liu Y, Fan C, Wang L, Li A, Zhou H, Cai L, Miao Y, Li Q, Qiu X, Wang E. Lasp1 promotes malignant phenotype of non-small-cell lung cancer via inducing phosphorylation of FAK-AKT pathway. Oncotarget 2017; 8:75102-75113. [PMID: 29088849 PMCID: PMC5650404 DOI: 10.18632/oncotarget.20527] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 07/28/2017] [Indexed: 12/12/2022] Open
Abstract
Lasp1 (LIM and SH3 domain protein 1) promotes tumor proliferation and invasion in multiple cancer entities including non-small cell lung cancer (NSCLC). However, the molecular mechanism is uncertain to date. In the present study, using immunohistochemistry, we found that Lasp1 expression was significantly correlated with tumor size (P=0.005), advanced TNM stage (P=0.042), positive regional lymph node metastasis (P=0.034) and poor overall survival (P<0.001). Similar results were seen in patients with squamous cell lung carcinoma (P=0.003 for larger tumor size, P=0.017 for advanced TNM stage, P=0.003 for positive lymph node metastasis and P<0.001 for poor overall survival) but not in patients with lung adenocarcinoma (P>0.05). Proliferation and invasion assay showed that Lasp1 dramatically promoted the ability of proliferation and invasion of NSCLC cells. Subsequent western blot results revealed that Lasp1 promoted the expression of Cyclin A2, CyclinB1, and Snail, and inhibited the expression of E-cadherin. Lasp1 directly interacted with FAK and facilitated the expression of phosphorylated FAK (Tyr397) and AKT (Ser473). Incorporation of both FAK inhibitor and AKT inhibitor counteracted the upregulating expression of Cyclin A2, CyclinB1, and Snail, and downregulating expression of E-cadherin expression induced by Lasp1 overexpression. Interestingly, inhibition of FAK signaling pathway attenuated the phosphorylation of AKT, but inhibition of AKT signaling pathway did not affect the phosphorylation of FAK. In conclusion, Lasp1 facilitated tumor proliferation and invasion of NSCLC through directly binding to FAK and enhancing the phosphorylation of FAK (Tyr397) and AKT (Ser473). Lasp1 may be a novel therapeutic target in the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Xiupeng Zhang
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Chuifeng Fan
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Liang Wang
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Ailin Li
- Department of Radiotherapy, First Hospital of China Medical University, Shenyang, China
| | - Haijing Zhou
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Lin Cai
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Yuan Miao
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Qingchang Li
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Xueshan Qiu
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| | - Enhua Wang
- Department of Pathology, Basic Medicine Science and First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Wu Q, Yan H, Tao SQ, Wang XN, Mou L, Chen P, Cheng XW, Wu WY, Wu ZS. XIAP 3'-untranslated region as a ceRNA promotes FSCN1 function in inducing the progression of breast cancer by binding endogenous miR-29a-5p. Oncotarget 2017; 8:16784-16800. [PMID: 28186968 PMCID: PMC5370001 DOI: 10.18632/oncotarget.15159] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
The non-coding 3′-untranslated region (UTR) of genes play an important role in the regulation of microRNA (miRNA) functions, since it can bind and inactivate multiple miRNAs. Herein, we report that ectopic expression of XIAP 3′UTR increased human breast cancer cells proliferation, colony formation, migration, invasion and xenograft tumor growth and suppressed tumor cell death. To investigate this process, we further correlated the genome-wide transcriptional profiling with the gene expression alterations after transfecting XIAP 3′UTR in MCF-7 cells. We identified a robust, genome-wide mechanism of cell migration, motility and epithelial to mesenchymal transition by which mediated by a previously described cellular component movement factor FSCN1. Expression of XIAP and FSCN1 were up-regulated synergistically after transfecting XIAP 3′UTR in vitro and in vivo. Interactions between XIAP and FSCN1 appear to be a key determinant of these processes. Co-transfection with Dicer siRNA reversed the XIAP 3′UTR-mediated oncogenicity, suggesting the miRNAs might be involved in that process. Furthermore, we demonstrated that one miRNA, miR-29a-5p, can bind to both the XIAP and FSCN1 3′UTRs and play an important role in that interactions. We showed that the 3′UTR of XIAP was able to antagonize miR-29a-5p, and resulted in the increased translation of XIAP and FSCN1. Thus, our findings reveal important new insights into how XIAP 3′UTR works, suggesting that the non-coding XIAP 3′UTR serves as a competitor for miRNA binding and subsequently inactivates miRNA functions, by which XIAP 3′UTR frees the target mRNAs from being repressed.
Collapse
Affiliation(s)
- Qiang Wu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Hong Yan
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Pathology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Si-Qi Tao
- Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Nan Wang
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Lang Mou
- Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Ping Chen
- Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Xing-Wang Cheng
- Department of Emergency, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Wen-Yong Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zheng-Sheng Wu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
26
|
Ruggieri V, Agriesti F, Tataranni T, Perris R, Mangieri D. Paving the path for invasion: The polyedric role of LASP1 in cancer. Tumour Biol 2017. [PMID: 28621232 DOI: 10.1177/1010428317705757] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Although usually referred to as a structural actin-binding protein, LIM and SH3 domain-containing protein may actually be dynamically involved in the control of a wide spectrum of cellular processes, by virtue of its interaction with several molecular partners. Alongside being ubiquitously expressed in physiological conditions, LIM and SH3 domain-containing protein is overexpressed in a growing number of human cancers, in which it may actively contribute to their aggressiveness by promoting cell proliferation and migration. In view of the recent findings, implicating the protein in cancer progression, we discuss here the most relevant discoveries highlighting the role of this versatile protein in various human tumors. The correlation between LIM and SH3 domain-containing protein expression levels in cancer and the poor outcome and metastatic behavior of tumors denotes the clinical significance of this protein and hints its potential value as a new cancer prognostic or even diagnostic biomarker. This may be decisive not only to optimize existing pharmacological regimes but also to delineate novel, more efficacious therapeutic and/or preventive approaches.
Collapse
Affiliation(s)
- Vitalba Ruggieri
- 1 Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| | - Francesca Agriesti
- 1 Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| | - Tiziana Tataranni
- 1 Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| | - Roberto Perris
- 2 Center for Molecular and Translational Oncology, University of Parma, Parma, Italy
| | - Domenica Mangieri
- 3 Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
27
|
Xu Q, Liu X, Liu Z, Zhou Z, Wang Y, Tu J, Li L, Bao H, Yang L, Tu K. MicroRNA-1296 inhibits metastasis and epithelial-mesenchymal transition of hepatocellular carcinoma by targeting SRPK1-mediated PI3K/AKT pathway. Mol Cancer 2017; 16:103. [PMID: 28606154 PMCID: PMC5469159 DOI: 10.1186/s12943-017-0675-y] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/06/2017] [Indexed: 12/18/2022] Open
Abstract
Background Increasing evidences demonstrate that miRNAs contribute to development and progression of hepatocellular carcinoma (HCC). Underexpression of miR-1296 is recently reported to promote growth and metastasis of human cancers. However, the expression and role of miR-1296 in HCC remain unknown. Methods The levels of miR-1296 in HCC tissues and cells were detected by qRT-PCR. Immunoblotting and immunofluorescence were used for detection of epithelial-to-mesenchymal transition (EMT) progression in HCC cells. Transwell assays were performed to determine migration and invasion of HCC cells. A lung metastasis mouse model was used to evaluated metastasis of HCC in vivo. The putative targets of miR-1296 were disclosed by public databases and a dual-luciferase reporter assay. Results We found that the expression of miR-1296 was reduced in HCC tissues and cell lines, and it was associated with metastasis and recurrence of HCC. Notably, miR-1296 overexpression inhibited migration, invasion and EMT progress of HCCLM3 cells, while miR-1296 loss facilitated these biological behaviors of Hep3B cells in vitro and in vivo. In addition, miR-1296 inversely regulated SRPK1 abundance by directly binding to its 3′-UTR, which subsequently resulted in suppression of p-AKT. Either SRPK1 re-expression or PI3K/AKT pathway activation, at least partially, abolished the effects of miR-1296 on migration, invasion and EMT progress of HCC cells. Furthermore, miR-1296 and SRPK1 expression were markedly correlated with adverse clinical features and poor prognosis of HCC patients. We showed that hypoxia was responsible for the underexpression of miR-1296 in HCC. And the promoting effects of hypoxia on metastasis and EMT of HCC cells were reversed by miR-1296. Conclusions Underexpression of miR-1296 potentially serves as a prognostic biomarker in HCC. Hypoxia-induced miR-1296 loss promotes metastasis and EMT of HCC cells probably by targeting SRPK1/AKT pathway. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0675-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiuran Xu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, 310014, China
| | - Xin Liu
- Department of Neurosurgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, 310014, China
| | - Zhikui Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Zhenyu Zhou
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, 510120, China
| | - Yufeng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Jianfeng Tu
- Department of Emergency, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, 310014, China
| | - Lijie Li
- Department of Gynecology, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, 310014, China
| | - Hangxing Bao
- Zhejiang Hospital of Traditional Chinese Medical, Hangzhou, Zhejiang Province, 310006, China
| | - Liu Yang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, 310014, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
| |
Collapse
|
28
|
Santiago L, Daniels G, Wang D, Deng FM, Lee P. Wnt signaling pathway protein LEF1 in cancer, as a biomarker for prognosis and a target for treatment. Am J Cancer Res 2017; 7:1389-1406. [PMID: 28670499 PMCID: PMC5489786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/08/2017] [Indexed: 06/07/2023] Open
Abstract
Transcription factors are regulatory proteins that either activate or repress the transcription of genes via binding to DNA regulatory sequences and regulating recruitment of transcriptional complexes. Lymphoid enhancer-binding factor 1 (LEF1), a member of the T-cell Factor (TCF)/LEF1 family of high-mobility group transcription factors, is a downstream mediator of the Wnt/β-catenin signaling pathway, but can also modulate gene transcription independently. LEF1 is essential in stem cell maintenance and organ development, especially in its role in epithelial-mesenchymal transition (EMT) by activating the transcription of hallmark EMT effectors including N-Cadherin, Vimentin, and Snail. Aberrant expression of LEF1 is implicated in tumorigenesis and cancer cell proliferation, migration, and invasion. LEF1's activity in particular cancer cell types, such as chronic lymphocytic leukemia (CLL), Burkitt lymphoma (BL), acute lymphoblastic leukemia (ALL), oral squamous cell carcinoma (OSCC), and colorectal cancer (CRC), makes it a valuable biomarker in predicting patient prognosis. Additionally, due to aberrant LEF1 activity resulting in cancer progression, knockdown and inhibition treatments designed to target LEF1 have proven effective in alleviating cancer growth, migration, and invasion in CLL, CRC, glioblastoma multiforme (GBM), and renal cell carcinoma (RCC). In prostate cancer cells, LEF1 promotes androgen receptor expression and activity in an androgen-independent manner, ultimately increasing prostate cancer growth regardless of androgen ablation therapy. In this review, we review LEF1 regulation, its role in tumorigenesis in several cancer types, and its clinical value as a biomarker for predicting prognoses and as a target for treatment.
Collapse
Affiliation(s)
- Larion Santiago
- Department of Pathology, School of Medicine, New York UniversityNew York, American
| | - Garrett Daniels
- Department of Pathology, School of Medicine, New York UniversityNew York, American
| | - Dongwen Wang
- Department of Urology, First Hospital of Shanxi Medical UniversityTaiyuan, Shanxi, China
| | - Fang-Ming Deng
- Department of Pathology, School of Medicine, New York UniversityNew York, American
- Association of Chinese American PhysiciansFlushing, New York, American
| | - Peng Lee
- Department of Pathology, School of Medicine, New York UniversityNew York, American
- Department of Urology, School of Medicine, New York UniversityNew York, American
- Department of NYU Cancer Institute, School of Medicine, New York UniversityNew York, American
- Department of New York Harbor Healthcare System, School of Medicine, New York UniversityNew York, American
- Association of Chinese American PhysiciansFlushing, New York, American
| |
Collapse
|
29
|
Holmila R, Sklias A, Muller DC, Degli Esposti D, Guilloreau P, Mckay J, Sangrajrang S, Srivatanakul P, Hainaut P, Merle P, Herceg Z, Nogueira da Costa A. Targeted deep sequencing of plasma circulating cell-free DNA reveals Vimentin and Fibulin 1 as potential epigenetic biomarkers for hepatocellular carcinoma. PLoS One 2017; 12:e0174265. [PMID: 28333958 PMCID: PMC5363871 DOI: 10.1371/journal.pone.0174265] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/06/2017] [Indexed: 12/29/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most common cause of cancer death worldwide, but is still lacking sensitive and specific biomarkers for early diagnosis and prognosis. In this study, we applied targeted massively parallel semiconductor sequencing to assess methylation on a panel of genes (FBLN1, HINT2, LAMC1, LTBP1, LTBP2, PSMA2, PSMA7, PXDN, TGFB1, UBE2L3, VIM and YWHAZ) in plasma circulating cell-free DNA (cfDNA) and to evaluate the potential of these genes as HCC biomarkers in two different series, one from France (42 HCC cases and 42 controls) and one from Thailand (42 HCC cases, 26 chronic liver disease cases and 42 controls). We also analyzed a set of HCC and adjacent tissues and liver cell lines to further compare with 'The Cancer Genome Atlas' (TCGA) data. The methylation in cfDNA was detected for FBLN1, PSMA7, PXDN and VIM, with differences in methylation patterns between cases and controls for FBLN1 and VIM. The average methylation level across analyzed CpG-sites was associated with higher odds of HCC for VIM (1.48 [1.02, 2.16] for French cases and 2.18 [1.28, 3.72] for Thai cases), and lower odds of HCC for FBLN1 (0.89 [0.76, 1.03] for French cases and 0.75 [0.63, 0.88] for Thai cases). In conclusion, our study provides evidence that changes in VIM and FBLN1 methylation levels in cfDNA are associated with HCC and could represent useful plasma-based biomarkers. Also, the potential to investigate methylation patterns in cfDNA could bring new strategies for HCC detection and monitoring high-risk groups and response to treatment.
Collapse
Affiliation(s)
- Reetta Holmila
- Epigenetics group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Athena Sklias
- Epigenetics group, International Agency for Research on Cancer (IARC), Lyon, France
| | - David C. Muller
- Genetic Epidemiology group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Davide Degli Esposti
- Epigenetics group, International Agency for Research on Cancer (IARC), Lyon, France
| | | | - James Mckay
- Genetic Cancer Susceptibility group, International Agency for Research on Cancer (IARC), Lyon, France
| | | | | | - Pierre Hainaut
- Institut Albert Bonniot, INSERM Unité 823, La Tronche, France
| | - Philippe Merle
- Croix-Rousse Hospital, Lyon, France
- UMR INSERM 1052, CRCL, Lyon, France
| | - Zdenko Herceg
- Epigenetics group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Andre Nogueira da Costa
- Molecular mechanisms and biomarkers group, International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
30
|
Kong FY, Zhu T, Li N, Cai YF, Zhou K, Wei X, Kou YB, You HJ, Zheng KY, Tang RX. Bioinformatics analysis of the proteins interacting with LASP-1 and their association with HBV-related hepatocellular carcinoma. Sci Rep 2017; 7:44017. [PMID: 28266596 PMCID: PMC5339786 DOI: 10.1038/srep44017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/02/2017] [Indexed: 12/11/2022] Open
Abstract
LIM and SH3 domain protein (LASP-1) is responsible for the development of several types of human cancers via the interaction with other proteins; however, the precise biological functions of proteins interacting with LASP-1 are not fully clarified. Although the role of LASP-1 in hepatocarcinogenesis has been reported, the implication of LASP-1 interactors in HBV-related hepatocellular carcinoma (HCC) is not clearly evaluated. We obtained information regarding LASP-1 interactors from public databases and published studies. Via bioinformatics analysis, we found that LASP-1 interactors were related to distinct molecular functions and associated with various biological processes. Through an integrated network analysis of the interaction and pathways of LASP-1 interactors, cross-talk between different proteins and associated pathways was found. In addition, LASP-1 and several its interactors are significantly altered in HBV-related HCC through microarray analysis and could form a complex co-expression network. In the disease, LASP-1 and its interactors were further predicted to be regulated by a complex interaction network composed of different transcription factors. Besides, numerous LASP-1 interactors were associated with various clinical factors and related to the survival and recurrence of HBV-related HCC. Taken together, these results could help enrich our understanding of LASP-1 interactors and their relationships with HBV-related HCC.
Collapse
Affiliation(s)
- Fan-Yun Kong
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ting Zhu
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Nan Li
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yun-Fei Cai
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kai Zhou
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao Wei
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan-Bo Kou
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong-Juan You
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kui-Yang Zheng
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ren-Xian Tang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
31
|
Li Z, Chen Y, Wang X, Zhang H, Zhang Y, Gao Y, Weng M, Wang L, Liang H, Li M, Zhang F, Zhao S, Liu S, Cao Y, Shu Y, Bao R, Zhou J, Liu X, Yan Y, Zhen L, Dong Q, Liu Y. LASP-1 induces proliferation, metastasis and cell cycle arrest at the G2/M phase in gallbladder cancer by down-regulating S100P via the PI3K/AKT pathway. Cancer Lett 2016; 372:239-50. [PMID: 26797416 DOI: 10.1016/j.canlet.2016.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/26/2015] [Accepted: 01/06/2016] [Indexed: 12/21/2022]
Abstract
LASP-1 is an actin-binding protein that regulates cytoskeletal dynamics and cell migration. LASP-1 was previously identified in a cDNA library from metastatic breast cancer samples. This protein has since been detected in multiple human cancers, including liver cancer, gastric cancer and pancreatic cancer. S100P is a small calcium-binding protein in the S100 protein family that regulates cellular, physiological and pathological processes in various cancers. However, the clinical significance of LASP-1 and S100P expression in gallbladder cancer (GBC) is not yet clear. In our study, we focused on the clinical significance, biological function and mechanism of LASP-1 in gallbladder cancer and detected LASP-1 and S100P overexpression in GBC tissues. The expression of LASP-1 was significantly correlated with poor prognosis in GBC patients (P < 0.05). Furthermore, down-regulation of LASP-1 expression resulted in the obvious inhibition of proliferation and migration and caused cell cycle arrest by down-regulating S100P via the PI3K/AKT pathway; in mice, tumor volume was significantly decreased. In conclusion, LASP-1 may act as an oncogene to regulate the expression of S100P to influence cellular functions in GBC. LASP-1 could serve as a genetic treatment target in GBC patients.
Collapse
Affiliation(s)
- ZhiZhen Li
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - YuanYuan Chen
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - XuAn Wang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - HongChen Zhang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Yijian Zhang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - YaoHui Gao
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Mingzhe Weng
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Lei Wang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - HaiBin Liang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - MaoLan Li
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Fei Zhang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Shuai Zhao
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Shibo Liu
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Yang Cao
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Yijun Shu
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Runfa Bao
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Jian Zhou
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Xiyong Liu
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Yun Yan
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Lei Zhen
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Qian Dong
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China.
| | - Yingbin Liu
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China; Institute of Biliary Tract Disease Research, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China.
| |
Collapse
|
32
|
Zhang H, Li Z, Chu B, Zhang F, Zhang Y, Ke F, Chen Y, Xu Y, Liu S, Zhao S, Liang H, Weng M, Wu X, Li M, Wu W, Quan Z, Liu Y, Zhang Y, Gong W. Upregulated LASP-1 correlates with a malignant phenotype and its potential therapeutic role in human cholangiocarcinoma. Tumour Biol 2016; 37:8305-15. [PMID: 26729195 DOI: 10.1007/s13277-015-4704-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 12/20/2015] [Indexed: 02/08/2023] Open
Abstract
LIM and SH3 protein 1 (LASP-1) is demonstrated to play a key role in occurrence and development of tumors. However, the expression and function of LASP-1 in cholangiocarcinoma (CCA) remain largely unexplored. This study aimed to investigate the effect of regulated LASP-1 expression on migration, invasion, proliferation, and apoptosis of CCA cells and on tumorigenesis in vivo, and to examine clinico-oncological correlates of LASP-1 expression. Expression of LASP-1 by immunohistochemistry was evaluated in CCA tissue samples. HCCC-9810 and RBE cells were transfected with the LASP-1 small interfering RNA (siRNA), and the effect of knocking down LASP-1 gene expression on cell migration, invasion, proliferation, and apoptosis were examined by wound healing, transwell assays, CCK-8 assays, colony formation, and flow cytometry assays, respectively. Xenograft tumor model was used to validate the effect of downregulated LASP-1 in vivo. Our results demonstrated that LASP-1 was over-expressed in CCA tissues, positively correlating with larger tumors, poor histological differentiation, lymph node metastasis, advanced TNM stage, and poor prognosis in CCA patients (P < 0.05). Downregulation of LASP-1 in HCCC-9810 and RBE cell lines significantly increased cell apoptosis and suppressed cell migration, invasion, and proliferation in vitro and tumorigenesis in vivo. Our results indicate that LASP-1 may essentially involve in the metastasis and growth of CCA and clinical significance of LASP-1 may reside in function as a biomarker to predict prognosis and as a promising therapeutic strategy for CCA patients by the inhibition of LASP-1 expression.
Collapse
Affiliation(s)
- Hongchen Zhang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Zhizhen Li
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Bingfeng Chu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fei Zhang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yijian Zhang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fayong Ke
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuanyuan Chen
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yi Xu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shibo Liu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shuai Zhao
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Haibin Liang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Mingzhe Weng
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiangsong Wu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Maolan Li
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Wenguang Wu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Zhiwei Quan
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yong Zhang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Wei Gong
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China. .,The Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|