1
|
Fu Y, Wang X, Chen X, Hong J, Qin Y, Zhou Z, Zhou X, Wang Y, Zhou J, Fang H, Liu P, Huang B. Establishment of matrix metalloproteinase 3 time-resolved immunoassay and some potential clinical applications. Anal Biochem 2023; 666:115072. [PMID: 36738873 DOI: 10.1016/j.ab.2023.115072] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/01/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
AIM To develop a highly sensitive time-resolved fluorescence immunoassay (TRFIA) for the detection of serum matrix metalloproteinase-3 (MMP-3) and to assess MMP-3's clinical value in patients with colorectal cancer (CRC).st. METHODS MMP-3 levels were established using the double antibody sandwich technique. The MMP-3 TRFIA technique was developed and optimized, and its linearity, sensitivity, accuracy, specificity, and recovery were assessed. Then, serum concentrations in healthy individuals and patients with CRC were determined by MMP-3 TRFIA. RESULTS The linear range of MMP-3 TRFIA was 0.73-500 ng/mL. MMP-3 TRFIA had an intra-batch precision range of 2.16%-7.10% percent and an inter-batch precision range of 3.99%-11.21%. MMP-3, tumor-associated trypsinogen 2, and AFP had no cross reaction.The recovery is between 90% and 110%, and had no serum interference. Patients with CRC had serum MMP-3 levels (73.95 ± 78.43 ng/mL) that were considerably higher than those of healthy individuals (21.45 ± 11.12 ng/mL), and those with metastasis had serum MMP-3 levels (95.89 ± 76.21 ng/mL) that were considerably higher than those of patients without metastasis (52.74 ± 47.25 ng/mL). CONCLUSIONS A highly sensitive MMP-3 TRFIA assay was successfully developed, and serum MMP-3 may be associated with CRC invasion and metastasis. Therefore, MMP-3 can be used in the auxiliary diagnosis of CRC.
Collapse
Affiliation(s)
- Yulin Fu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaoyan Wang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xindong Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jianfeng Hong
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zixuan Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | | | - Hongming Fang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Pengfei Liu
- The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| |
Collapse
|
2
|
Yu X, Jin J, Zheng Y, Zhu H, Xu H, Ma J, Lan Q, Zhuang Z, Chen CC, Li M. GBP5 drives malignancy of glioblastoma via the Src/ERK1/2/MMP3 pathway. Cell Death Dis 2021; 12:203. [PMID: 33608513 PMCID: PMC7896088 DOI: 10.1038/s41419-021-03492-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Guanylate binding proteins (GBPs), a family of interferon-inducible large GTPase, play a pivotal role in cell-autonomous immunity and tumor malignant transformation. Glioblastoma (GBM) is the most prevalent and lethal primary brain tumor in adults. Here we show that GBP5 was highly expressed in GBM cell lines and in clinical samples, especially in the mesenchymal subtype. The expression levels of GBP5 were negatively correlated with the prognosis of GBM patients. Overexpression of GBP5 promoted the proliferation, migration, and invasion of GBM cells in vitro and in vivo. In contrast, silencing GBP5 by RNA interference exhibited the opposite effects. Consequently, targeting GBP5 in GBM cells resulted in impaired tumor growth and prolonged survival time of mice with GBM tumors. We further identified that the Src/ERK1/2/MMP3 axis was essential for GBP5-promoted GBM aggressiveness. These findings suggest that GBP5 may represent a novel target for GBM intervention.
Collapse
Affiliation(s)
- Xiaoting Yu
- Central Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jing Jin
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanwen Zheng
- Central Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Hua Zhu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Hui Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jun Ma
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| | - Ming Li
- Central Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Fu X, Yao M, Ye C, Fang T, Wu R. Osteopontin Regulates Endometrial Stromal Cell Migration in Endometriosis through the PI3K Pathway : Osteopontin Regulates Endometrial Cell Migration in Endometriosis. Reprod Sci 2020; 28:435-446. [PMID: 32909189 PMCID: PMC7808973 DOI: 10.1007/s43032-020-00301-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/17/2020] [Indexed: 01/29/2023]
Abstract
Endometriosis is generally characterized as a tumor-like disease because of its potential for distant metastasis and local tissue invasion, while whether osteopontin (OPN) plays a role in the pathogenesis of endometriosis has not been thoroughly investigated. We investigated the expression of OPN, urokinase plasminogen activator (uPA), phosphatidylinositol 3 kinase (PI3K), and phospho-PI3 kinase (p-PI3K) in endometrial stromal cells (ESCs). The serum concentration of OPN was determined by enzyme-linked immunosorbent assays (ELISA). OPN was downregulated to explore the corresponding change of uPA, p-PI3K, F-actin, and α-tubulin. The expression of OPN, uPA, PI3K, and p-PI3K was evaluated by western blot and quantitative real-time PCR (RT-qPCR) and the expression of F-actin and α-tubulin was confirmed by immunofluorescence assay. The proliferation and migration abilities of ESCs were investigated by CCK8, transwell, and wound scratch assays. Endometrial OPN, p-PI3K, and uPA expressions and serum OPN levels were increased in patients with endometriosis compared with the control. The expressions of p-PI3K, uPA, and α-tubulin were decreased by siRNA-OPN interference in ectopic ESCs. Activation and inhibition of the PI3K pathway apparently upregulate and downregulate uPA expression. Knockdown of OPN and inhibition of the PI3K pathway remarkably inhibited cell migration in ectopic ESCs. Meanwhile, activation of the PI3K pathway promoted the migration ability of ectopic ESCs. OPN may regulate the expression of uPA through the PI3K signal pathway to affect the migration ability of ESCs, indicating that OPN, uPA, and the PI3K pathway may be potential targets for interrupting development of endometriosis.
Collapse
Affiliation(s)
- Xiaoxia Fu
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Mengyun Yao
- Institute of Burn Research, South-West Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University, Chongqing, China
| | - Chaoshuang Ye
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Tao Fang
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Ruijin Wu
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China.
| |
Collapse
|
4
|
Korshunov DA, Kondakova IV, Shashova EE. Modern Perspective on Metabolic Reprogramming in Malignant Neoplasms. BIOCHEMISTRY (MOSCOW) 2019; 84:1129-1142. [PMID: 31694509 DOI: 10.1134/s000629791910002x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolic reprogramming is one of the central features of transformed cells. Elucidation of interactions between oncogenic signaling and cell metabolic processes has become the basis for extensive studies of metabolism reprogramming in tumor tissue. The review summarizes the key results of studies on the catabolic and anabolic rearrangements in tumor cells with special emphasis on carbohydrate, lipid, amino acid, and acetate metabolism determining the cancer phenotype of cells.
Collapse
Affiliation(s)
- D A Korshunov
- Tomsk National Research Medical Center, Tomsk, 634009, Russia.
| | - I V Kondakova
- Tomsk National Research Medical Center, Tomsk, 634009, Russia
| | - E E Shashova
- Tomsk National Research Medical Center, Tomsk, 634009, Russia
| |
Collapse
|
5
|
Zou X, Wu Z, Huang J, Liu P, Qin X, Chen L, Zhu W, Zhao Y, Li P, Song J, Yang GY, Mao Y. The Role of Matrix Metalloproteinase-3 in the Doxycycline Attenuation of Intracranial Venous Hypertension-Induced Angiogenesis. Neurosurgery 2019; 83:1317-1327. [PMID: 29462373 DOI: 10.1093/neuros/nyx633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 12/22/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The molecular mechanism of brain arteriovenous malformation (BAVM) is largely unknown. Intracranial venous hypertension (VH) may enhance focal angiogenesis and promote BAVM development and progression. A rat VH model effectively simulates the hemodynamic microenvironment of this disease. OBJECTIVE To explore the effect of doxycycline in VH-related angiogenesis, as well as the role of matrix metalloproteinase-3 (MMP-3) and other molecular factors. METHODS A rat VH model was generated by common carotid artery and distal external jugular vein anastomosis. Microvessel density (MVD) in the perisinus area and expression of MMP-3/2/9, VEGF, TIMP-1, TGF-β, and HIF-1α were examined, with and without daily doxycycline treatment for 4 wk. The effects of doxycycline were verified in Vitro using human brain microvascular endothelial cells (HBMECs). MMP-3 overexpression or knockdown in HBMECs was used to confirm the role of MMP-3 in cell functions. RESULTS MVD in the perisinus cortex was greatly increased after VH. Doxycycline decreased MVD, suppressed MMP-3 overexpression, and reduced VEGF, TGF-β, and TIMP-1 levels compared with the controls (P < .05). In Vitro, doxycycline decreased HBMEC migration, tube formation, and the mRNA, protein, and enzymatic activity levels of MMP-3. MMP-3 overexpression in HBMECs promoted migration, while knockdown of MMP-3 significantly attenuated proliferation, migration, and tube formation (P < .05). CONCLUSION Our findings indicate that MMP-3 plays an important role in VH-related angiogenesis and the promotion of vascular remodeling. Suppression of MMP-3 overexpression by doxycycline may provide a potential strategy for inhibiting BAVM development.
Collapse
Affiliation(s)
- Xiang Zou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zehan Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Huang
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.,The Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuanfeng Qin
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peiliang Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guo-Yuan Yang
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Ji Y, Li J, Li P, Wang L, Yang H, Jiang G. C/EBPβ Promotion of MMP3-Dependent Tumor Cell Invasion and Association with Metastasis in Colorectal Cancer. Genet Test Mol Biomarkers 2017; 22:5-10. [PMID: 29172775 DOI: 10.1089/gtmb.2017.0113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AIMS Tumor metastasis is a significant obstacle to curing colorectal cancer (CRC). C/EBPβ is thought to play an important role in CRC invasion and metastasis. In this study, we assessed whether C/EBPβ-mediated tumor invasion was dependent on MMP3, the expression of which is upregulated by C/EBPβ. We then determined whether C/EBPβ upregulation was associated with MMP3 levels and metastatic status in human CRC patients. MATERIALS AND METHODS A total of 102 patients were recruited for this study. mRNA and protein levels of C/EBPβ and MMP3 in CRC cell lines and patient specimens were determined by reverse transcription-polymerase chain reaction and western blot, respectively. Tumor cell invasion was analyzed using an in vitro Matrigel Invasion Assay. The correlation between C/EBPβ and MMP3 expression was determined by Pearson's correlation analysis. RESULTS Both mRNA and protein levels of MMP3 were upregulated by C/EBPβ overexpression and downregulated by C/EBPβ siRNA in HCT116 CRC cell cultures. C/EBPβ-enhanced tumor cell invasion was inhibited by MMP3 siRNA. In human CRC patients, C/EBPβ levels were correlated with MMP3 levels and metastatic status. CONCLUSIONS C/EBPβ upregulation promoted tumor cell invasion in an MMP3-dependent manner in vitro and was associated with metastatic status in CRC.
Collapse
Affiliation(s)
- Yuanyuan Ji
- 1 Department of Oncology, Anyang Tumor Hospital , Anyang, China
| | - Junkuo Li
- 2 Department of Pathology, Anyang Tumor Hospital , Anyang, China
| | - Pan Li
- 3 Department of Pathology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| | - Li Wang
- 3 Department of Pathology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| | - Haijun Yang
- 2 Department of Pathology, Anyang Tumor Hospital , Anyang, China
| | - Guozhong Jiang
- 3 Department of Pathology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| |
Collapse
|
7
|
Zhao LC, Li J, Liao K, Luo N, Shi QQ, Feng ZQ, Chen DL. Evodiamine Induces Apoptosis and Inhibits Migration of HCT-116 Human Colorectal Cancer Cells. Int J Mol Sci 2015; 16:27411-21. [PMID: 26580615 PMCID: PMC4661889 DOI: 10.3390/ijms161126031] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/08/2015] [Accepted: 10/08/2015] [Indexed: 01/21/2023] Open
Abstract
Evodiamine (EVO) exhibits strong anti-cancer effects. However, the effect of EVO on the human colorectal cancer cell line HCT-116 has not been explored in detail, and its underlying molecular mechanisms remain unknown. In the present study, cell viability was assessed by Cell Counting Kit-8 (CCK-8). Cell cycle and apoptosis were measured by flow cytometry, and morphological changes in the nucleus were examined by fluorescence microscopy and Hoechst staining. Cell motility was detected by Transwell assay. ELISA was used to assess the protein levels of autocrine motility factor (AMF) in the cell supernatant, and protein expression was determined by Western blotting. Our results showed that EVO inhibited the proliferation of HCT-116 cells, caused accumulation of cells in S and G2/M phases, and reduced the levels of the secreted form of AMF. The protein levels of tumor suppressor protein (p53), Bcl-2 Associated X protein (Bax), B cell CLL/lymphoma-2 (Bcl-2), phosphoglucose isomerase (PGI), phosphorylated signal transducers and activators of transcription 3 (p-STAT3) and matrix metalloproteinase 3 (MMP3) were altered in cells treated with EVO. Taken together, our results suggest that EVO modulates the activity of the p53 signaling pathway to induce apoptosis and downregulate MMP3 expression by inactivating the JAK2/STAT3 pathway through the downregulation of PGI to inhibit migration of HCT-116 human colorectal cancer cells.
Collapse
Affiliation(s)
- Lv-Cui Zhao
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
- Drug Engineering Research Center of Chongqing Medical University, Chongqing 400016, China.
| | - Jing Li
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Ke Liao
- Department of Respiration, Cheng Du Tumor Hospital, Chengdu 610041, China.
| | - Nian Luo
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Qing-Qiang Shi
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Zi-Qiang Feng
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Di-Long Chen
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
8
|
Lin HY, Liu JH, Cheng KL, Lin JY, Liu NR, Meng M. A novel binding of GTP stabilizes the structure and modulates the activities of human phosphoglucose isomerase/autocrine motility factor. Biochem Biophys Rep 2015; 2:14-22. [PMID: 29124141 PMCID: PMC5668625 DOI: 10.1016/j.bbrep.2015.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/13/2015] [Accepted: 04/13/2015] [Indexed: 11/29/2022] Open
Abstract
Phosphoglucose isomerase (PGI) catalyzes the interconversion between glucose 6-phosphate and fructose 6-phosphate in the glycolysis pathway. In mammals, the enzyme is also identical to the extracellular proteins neuroleukin, tumor-secreted autocrine motility factor (AMF) and differentiation and maturation mediator for myeloid leukemia. Hereditary deficiency of the enzyme causes non-spherocytic hemolytic anemia in human. In the present study, a novel interaction between GTP and human PGI was corroborated by UV-induced crosslinking, affinity purification and kinetic study. GTP not only inhibits the isomerization activity but also compromises the AMF function of the enzyme. Kinetic studies, including the Yonetani-Theorell method, suggest that GTP is a competitive inhibitor with a Ki value of 63 μM and the GTP-binding site partially overlaps with the catalytic site. In addition, GTP stabilizes the structure of human PGI against heat- and detergent-induced denaturation. Molecular modelling and dynamic simulation suggest that GTP is bound in a syn-conformation with the γ-phosphate group located near the phosphate-binding loop and the ribose moiety positioned away from the active-site residues.
Collapse
Affiliation(s)
- Hua-Yang Lin
- Graduate Institute of Biotechnology, National Chung Hsing University (NCHU), 250 Kuo-Kuang Road, Taichung, Taiwan 40227
| | - Jyung-Hurng Liu
- Graduate Institute of Genomics and Bioinformatics, NCHU, Taichung, Taiwan 40227.,Agricultural Biotechnology Center (ABC), NCHU, Taichung, Taiwan 40227.,Rong Hsing Research Center for Translational Medicine, NCHU, Taichung, Taiwan 40227
| | - Ka-Lik Cheng
- Graduate Institute of Biotechnology, National Chung Hsing University (NCHU), 250 Kuo-Kuang Road, Taichung, Taiwan 40227
| | - Jia-Yun Lin
- Graduate Institute of Biotechnology, National Chung Hsing University (NCHU), 250 Kuo-Kuang Road, Taichung, Taiwan 40227
| | - Ni-Rung Liu
- Graduate Institute of Biotechnology, National Chung Hsing University (NCHU), 250 Kuo-Kuang Road, Taichung, Taiwan 40227
| | - Menghsiao Meng
- Graduate Institute of Biotechnology, National Chung Hsing University (NCHU), 250 Kuo-Kuang Road, Taichung, Taiwan 40227
| |
Collapse
|
9
|
Wang X, Wang H, Yang H, Li J, Cai Q, Shapiro IM, Risbud MV. Tumor necrosis factor-α- and interleukin-1β-dependent matrix metalloproteinase-3 expression in nucleus pulposus cells requires cooperative signaling via syndecan 4 and mitogen-activated protein kinase-NF-κB axis: implications in inflammatory disc disease. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2560-72. [PMID: 25063530 DOI: 10.1016/j.ajpath.2014.06.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/28/2014] [Accepted: 06/04/2014] [Indexed: 12/19/2022]
Abstract
Matrix metalloproteinase-3 (MMP-3) plays an important role in intervertebral disc degeneration, a ubiquitous condition closely linked to low back pain and disability. Elevated expression of syndecan 4, a cell surface heparan sulfate proteoglycan, actively controls disc matrix catabolism. However, the relationship between MMP-3 expression and syndecan 4 in the context of inflammatory disc disease has not been clearly defined. We investigated the mechanisms by which cytokines control MMP-3 expression in rat and human nucleus pulposus cells. Cytokine treatment increased MMP-3 expression and promoter activity. Stable silencing of syndecan 4 blocked cytokine-mediated MMP-3 expression; more important, syndecan 4 did not mediate its effects through NF-κB or mitogen-activated protein kinase (MAPK) pathways. However, treatment with MAPK and NF-κB inhibitors resulted in partial blocking of the inductive effect of cytokines on MMP-3 expression. Loss-of-function studies confirmed that NF-κB, p38α/β2/γ/δ, and extracellular signal-regulated kinase (ERK) 2, but not ERK1, contributed to cytokine-dependent induction of MMP3 promoter activity. Similarly, inhibitor treatments, lentiviral short hairpin-p65, and short hairpin-IκB kinase β significantly decreased cytokine-dependent up-regulation in MMP-3 expression. Finally, we show that transforming growth factor-β can block the up-regulation of MMP-3 induced by tumor necrosis factor (TNF)-α by counteracting the NF-κB pathway and syndecan 4 expression. Taken together, our results suggest that cooperative signaling through syndecan 4 and the TNF receptor 1-MAPK-NF-κB axis is required for TNF-α-dependent expression of MMP-3 in nucleus pulposus cells. Controlling these pathways may slow the progression of intervertebral disc degeneration and matrix catabolism.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopedic Surgery and the Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Bone and Soft Tissue, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Hua Wang
- Department of Orthopedic Surgery and the Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Orthopedics, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hao Yang
- Department of Orthopedic Surgery and the Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Orthopedics, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Li
- Department of Orthopedic Surgery and the Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Qiqing Cai
- Department of Bone and Soft Tissue, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Irving M Shapiro
- Department of Orthopedic Surgery and the Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Makarand V Risbud
- Department of Orthopedic Surgery and the Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
10
|
Shih WL, Yu FL, Chang CD, Liao MH, Wu HY, Lin PY. Suppression of AMF/PGI-mediated tumorigenic activities by ursolic acid in cultured hepatoma cells and in a mouse model. Mol Carcinog 2013; 52:800-12. [PMID: 22549898 DOI: 10.1002/mc.21919] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/27/2012] [Accepted: 04/06/2012] [Indexed: 01/11/2023]
Abstract
Our previous studies demonstrated that autocrine motility factor/phosphoglucose isomerase (AMF/PGI) possesses tumorigenic activities through the modulation of intracellular signaling. We then investigated the effects of ursolic acid (UA), oleanolic acid (OA), tangeretin, and nobiletin against AMF/PGI-mediated oncogenesis in cultured stable Huh7 and Hep3B cells expressing wild-type or mutated AMF/PGI and in a mouse model in this study. The working concentrations of the tested compounds were lower than their IC10 , which was determined by Brdu incorporation and colony formation assay. Only UA efficiently suppressed the AMF/PGI-induced Huh7 cell migration and MMP-3 secretion. Additionally, UA inhibited the AMF/PGI-mediated protection against TGF-β-induced apoptosis in Hep3B cells, whereas OA, tangeretin, and nobiletin had no effect. In Huh7 cells and tumor tissues, UA disrupted the Src/RhoA/PI 3-kinase signaling and complex formation induced by AMF/PGI. In the Hep3B system, UA dramatically suppressed AMF/PGI-induced anti-apoptotic signaling transmission, including Akt, p85, Bad, and Stat3 phosphorylation. AMF/PGI enhances tumor growth, angiogenesis, and pulmonary metastasis in mice, which is correlated with its enzymatic activity, and critically, UA intraperitoneal injection reduces the tumorigenesis in vivo, enhances apoptosis in tumor tissues and also prolongs mouse survival. Combination of sub-optimal dose of UA and cisplatin, a synergistic tumor cell-killing effects was found. Thus, UA modulates intracellular signaling and might serve as a functional natural compound for preventing or alleviating hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wen-Ling Shih
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | | | | | | | | | | |
Collapse
|
11
|
Suppression of apoptosis by pseudorabies virus Us3 protein kinase through the activation of PI3-K/Akt and NF-κB pathways. Res Vet Sci 2013; 95:764-74. [PMID: 23835241 DOI: 10.1016/j.rvsc.2013.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/29/2013] [Accepted: 06/02/2013] [Indexed: 12/22/2022]
Abstract
The pseudorabies virus (PRV) is a major viral disease that causes huge economic loss in the pig industry globally. Most viruses have been found to generate anti-apoptotic factors that facilitate cell survival in the early stages of infection. This study aimed to investigate the anti-apoptotic effects of PRV and study the underlying mechanisms in the early stage of infection. We investigated and compared whether the two PRV Us3 isoforms, Us3a and Us3b, could block apoptosis induced by virus infection, and further identified molecules involved in the signaling pathways. Our results demonstrated that PRV elicits 3-phosphoinositide dependent protein kinase-1/phosphatidylinositide 3-kinases/Akt (PDK-1/PI3-K/Akt)- and nuclear factor-κB (NF-κB)-dependent signaling in the early stage of infection. Inhibition of the PI3-K/Akt or NF-κB pathway enhanced cell death but no effect was observed on virus replication or PRV gene expression. Transiently-expressed GFP- or His-tagged PRV Us3a and Us3b cDNA protect cells against PRV-, avian reovirus- or bovine ephemeral fever virus-induced apoptosis in the cell lines. Us3a and Us3b transient over-expression upregulated several anti-apopototic signaling events, and the anti-apoptosis activity of Us3a is greater than that of Us3b. Kinase activity-deficient point or double point mutated Us3a lost the kinase activity of Us3a, which showed that kinase activity is required for the anti-apoptosis effect of Us3. Akt and NF-κB activation still occurred in UV-inactivated PRV- and cycloheximide-treated cells. In vivo study showed that PRV-infected trigeminal ganglion increases the expression of anti-apoptosis signaling molecules, including Akt, PDK-1 and IκBα, which is a similar result to that seen in the in vitro experiments. Our study suggests that signaling mechanisms may play important roles in PRV pathogenesis.
Collapse
|
12
|
Lan YY, Yeh TH, Lin WH, Wu SY, Lai HC, Chang FH, Takada K, Chang Y. Epstein-Barr virus Zta upregulates matrix metalloproteinases 3 and 9 that synergistically promote cell invasion in vitro. PLoS One 2013; 8:e56121. [PMID: 23409137 PMCID: PMC3567054 DOI: 10.1371/journal.pone.0056121] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 01/04/2013] [Indexed: 02/03/2023] Open
Abstract
Zta is a lytic transactivator of Epstein-Barr virus (EBV) and has been shown to promote migration and invasion of epithelial cells. Although previous studies indicate that Zta induces expression of matrix metalloproteinase (MMP) 9 and MMP1, direct evidence linking the MMPs to Zta-induced cell migration and invasion is still lacking. Here we performed a series of in vitro studies to re-examine the expression profile and biologic functions of Zta-induced MMPs in epithelial cells derived from nasopharyngeal carcinoma. We found that, in addition to MMP9, MMP3 was a new target gene upregulated by Zta. Ectopic Zta expression in EBV-negative cells increased both mRNA and protein production of MMP3. Endogenous Zta also contributed to induction of MMP3 expression, migration and invasion of EBV-infected cells. Zta activated the MMP3 promoter through three AP-1 elements, and its DNA-binding domain was required for the promoter binding and MMP3 induction. We further tested the effects of MMP3 and MMP9 on cell motility and invasiveness in vitro. Zta-promoted cell migration required MMP3 but not MMP9. On the other hand, both MMP3 and MMP9 were essential for Zta-induced cell invasion, and co-expression of the two MMPs synergistically increased cell invasiveness. Therefore, this study provides integrated evidence demonstrating that, at least in the in vitro cell models, Zta drives cell migration and invasion through MMPs.
Collapse
Affiliation(s)
- Yu-Yan Lan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Graduate Institute of Basic Medical Sciences, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Hao Yeh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Department of Microbiology and Immunology, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Hung Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Department of Microbiology and Immunology, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yi Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Hsiao-Ching Lai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Graduate Institute of Basic Medical Sciences, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Fang-Hsin Chang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Department of Microbiology and Immunology, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Kenzo Takada
- Department of Tumor Virology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yao Chang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Graduate Institute of Basic Medical Sciences, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
13
|
Granchi C, Minutolo F. Anticancer agents that counteract tumor glycolysis. ChemMedChem 2012; 7:1318-50. [PMID: 22684868 PMCID: PMC3516916 DOI: 10.1002/cmdc.201200176] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 05/04/2012] [Indexed: 12/12/2022]
Abstract
Can we consider cancer to be a "metabolic disease"? Tumors are the result of a metabolic selection, forming tissues composed of heterogeneous cells that generally express an overactive metabolism as a common feature. In fact, cancer cells have increased needs for both energy and biosynthetic intermediates to support their growth and invasiveness. However, their high proliferation rate often generates regions that are insufficiently oxygenated. Therefore, their carbohydrate metabolism must rely mostly on a glycolytic process that is uncoupled from oxidative phosphorylation. This metabolic switch, also known as the Warburg effect, constitutes a fundamental adaptation of tumor cells to a relatively hostile environment, and supports the evolution of aggressive and metastatic phenotypes. As a result, tumor glycolysis may constitute an attractive target for cancer therapy. This approach has often raised concerns that antiglycolytic agents may cause serious side effects toward normal cells. The key to selective action against cancer cells can be found in their hyperbolic addiction to glycolysis, which may be exploited to generate new anticancer drugs with minimal toxicity. There is growing evidence to support many glycolytic enzymes and transporters as suitable candidate targets for cancer therapy. Herein we review some of the most relevant antiglycolytic agents that have been investigated thus far for the treatment of cancer.
Collapse
Affiliation(s)
- Carlotta Granchi
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa (Italy)
| | - Filippo Minutolo
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa (Italy)
| |
Collapse
|
14
|
Dayoub R, Wagner H, Bataille F, Stöltzing O, Spruss T, Buechler C, Schlitt HJ, Weiss TS. Liver regeneration associated protein (ALR) exhibits antimetastatic potential in hepatocellular carcinoma. Mol Med 2010; 17:221-8. [PMID: 21152698 DOI: 10.2119/molmed.2010.00117] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 12/02/2010] [Indexed: 01/17/2023] Open
Abstract
Augmenter of liver regeneration (ALR), which is critically important in liver regeneration and hepatocyte proliferation, is highly expressed in cirrhotic livers and hepatocellular carcinomas (HCC). In the current study, the functional role of ALR in hepatocancerogenesis was analyzed in more detail. HepG2 cells, in which the cytosolic 15 kDa ALR isoform was reexpressed stably, (HepG2-ALR) were used in migration and invasion assays using modified Boyden chambers. Epithelial-mesenchymal transition (EMT) markers were determined in HepG2-ALR cells in vitro and in HepG2-ALR tumors grown in nude mice. ALR protein was quantified in HCC and nontumorous tissues by immunohistochemistry. HepG2-ALR, compared with HepG2 cells, demonstrated reduced cell motility and increased expression of the epithelial cell markers E-cadherin and Zona occludens-1 (ZO-1), whereas SNAIL, a negative regulator of E-cadherin, was diminished. Matrix metalloproteinase MMP1 and MMP3 mRNA expression and activity were reduced. HepG2-ALR cell-derived subcutaneously grown tumors displayed fewer necrotic areas, more epithelial-like cell growth and fewer polymorphisms and atypical mitotic figures than tumors derived from HepG2 cells. Analysis of tumor tissues of 53 patients with HCC demonstrated an inverse correlation of ALR protein with histological angioinvasion and grading. The 15 kDa ALR isoform was found mainly in HCC tissues without histological angioinvasion 0. In summary the present data indicate that cytosolic ALR reduces hepatoma cell migration, augments epithelial growth and, therefore, may act as an antimetastatic and EMT reversing protein.
Collapse
Affiliation(s)
- Rania Dayoub
- Center for Liver Cell Research, University Medical Center Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Ahluwalia M, de Groot J, Liu W(M, Gladson CL. Targeting SRC in glioblastoma tumors and brain metastases: rationale and preclinical studies. Cancer Lett 2010; 298:139-49. [PMID: 20947248 PMCID: PMC3212431 DOI: 10.1016/j.canlet.2010.08.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 08/22/2010] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is an extremely aggressive, infiltrative tumor with a poor prognosis. The regulatory approval of bevacizumab for recurrent GBM has confirmed that molecularly targeted agents have potential for GBM treatment. Preclinical data showing that SRC and SRC-family kinases (SFKs) mediate intracellular signaling pathways controlling key biologic/oncogenic processes provide a strong rationale for investigating SRC/SFK inhibitors, e.g., dasatinib, in GBM and clinical studies are underway. The activity of these agents against solid tumors suggests that they may also be useful in treating brain metastases. This article reviews the potential for using SRC/SFK inhibitors to treat GBM and brain metastases.
Collapse
Affiliation(s)
- Manmeet Ahluwalia
- Cleveland Clinic Main Campus, Mail Code ND40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-444-6145
| | - John de Groot
- The Brain Tumor Center, The University of Texas, M.D. Anderson Cancer Center, 1515, Holcombe Blvd., Unit 431, Houston, TX 77030, Phone: 713-792-7255
| | - Wei (Michael) Liu
- Lerner Research Institute, Department of Cancer Biology, Cleveland Clinic Mail Code NB40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-636-9494
| | - Candece L Gladson
- Lerner Research Institute, Department of Cancer Biology, Cleveland Clinic Mail Code NB40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-636-9493, Fax: 216-445-6269
| |
Collapse
|
16
|
Lin PY, Liu HJ, Liao MH, Chang CD, Chang CI, Cheng HL, Lee JW, Shih WL. Activation of PI 3-kinase/Akt/NF-kappaB and Stat3 signaling by avian reovirus S1133 in the early stages of infection results in an inflammatory response and delayed apoptosis. Virology 2010; 400:104-14. [PMID: 20170934 DOI: 10.1016/j.virol.2010.01.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 01/05/2010] [Accepted: 01/20/2010] [Indexed: 01/08/2023]
Abstract
Avian reovirus (ARV) strain S1133 causes apoptosis in host cells in the middle to late stages of infection. This study investigated the early-stage biological response and intracellular signaling in ARV S1133-infected Vero and chicken cells. Treatment with conditioned medium from ARV S1133-infected cells increased the chemotactic activity of U937 cells. Neutralizing antibodies against IL-1beta and IL-6 showed that both cytokines contribute to viral-induced inflammation but neither affect cell survival. Inhibition of Akt, NF-kappaB, and Stat3 released the chemotactic activity and anti-apoptotic effect elicited by ARV S1133. ARV S1133 activated PI 3-kinase-dependent Akt/NF-kappaB and p70 S6 kinase, as well as Stat3; however, p70 S6 kinase was not involved in ARV S1133-mediated effects. DF1 cells over-expressing constitutively active PI 3-kinase and Stat3 showed association with enhancement of anti-apoptotic activity. In conclusion, in the early stages of ARV S1133 infection, activation of cell survival signals contributes to virus-induced inflammation and anti-apoptotic response.
Collapse
Affiliation(s)
- Ping-Yuan Lin
- Graduate Institute and Department of Life Science, Tzu-Chi University, Hualien, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Shih WL, Liao MH, Lin PY, Chang CI, Cheng HL, Yu FL, Lee JW. PI 3-kinase/Akt and STAT3 are required for the prevention of TGF-beta-induced Hep3B cell apoptosis by autocrine motility factor/phosphoglucose isomerase. Cancer Lett 2009; 290:223-37. [PMID: 19819066 DOI: 10.1016/j.canlet.2009.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 09/16/2009] [Accepted: 09/17/2009] [Indexed: 11/17/2022]
Abstract
We established Hep3B cells stably-expressing wild-type and mutated AMF/PGI with differing enzymatic activities in order to investigate how AMF/PGI affects TGF-beta-induced apoptosis, and demonstrated that AMF/PGI against TGF-beta-induced apoptosis was correlated with its enzymatic activity. AMF/PGI did not alter TGF-beta-receptor expression nor affect TGF-beta-induced PAI-1 gene promoter or Smad3/4 activity. AMF/PGI induced PI 3-kinase activity, IRS and Akt phosphorylation, which can further regulate BAD phosphorylation. Constitutively-active p110 enhanced AMF/PGI-mediated anti-apoptosis activity, and dominant negative Akt alleviated anti-TGF-beta-induced apoptosis. We also demonstrated that STAT3 is a weak anti-apoptotic agent but has an increased anti-apoptotic effect in cooperation with PI 3-kinase/Akt.
Collapse
Affiliation(s)
- Wen-Ling Shih
- National Pingtung University of Science and Technology, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
18
|
Grise F, Bidaud A, Moreau V. Rho GTPases in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2009; 1795:137-51. [PMID: 19162129 DOI: 10.1016/j.bbcan.2008.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 12/21/2008] [Accepted: 12/24/2008] [Indexed: 01/05/2023]
Abstract
Rho GTPases are major regulators of signal transduction pathways and play key roles in processes including actin dynamics, cell cycle progression, cell survival and gene expression, whose deregulation may lead to tumorigenesis. A growing number of in vitro and in vivo studies using tumor-derived cell lines, primary tumors and animal cancer models strongly suggest that altered Rho GTPase signaling plays an important role in the initiation as well as in the progression of hepatocellular carcinoma (HCC), one of the deadliest human cancers in the world. These alterations can occur at the level of the GTPases themselves or of one of their regulators or effectors. The participation into the tumorigenic process can occur either through the over-expression of one of these components which presents an oncogenic activity as illustrated with RhoA and C or through the attenuation of the expression of a component presenting tumor suppressor activity as for Cdc42 or the RhoGAP, DLC-1. Consequently, these observations reflect the heterogeneity and the complexity of liver carcinogenesis. Recently, pharmacological approaches targeting Rho GTPase signaling have been used in HCC-derived models with relative success but remain to be validated in more physiologically relevant systems. Therefore, therapeutic approaches targeting Rho GTPase signaling may provide a novel alternative for anti-HCC therapy.
Collapse
Affiliation(s)
- Florence Grise
- INSERM, U889, Bordeaux, 33076 Bordeaux, France; Université Victor Segalen Bordeaux 2, Bordeaux, 33076 Bordeaux, France
| | | | | |
Collapse
|