1
|
Ke FY, Chen WY, Lin MC, Hwang YC, Kuo KT, Wu HC. Novel monoclonal antibody against integrin α3 shows therapeutic potential for ovarian cancer. Cancer Sci 2020; 111:3478-3492. [PMID: 32648337 PMCID: PMC7541015 DOI: 10.1111/cas.14566] [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: 04/15/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer has a high recurrence rate after platinum‐based chemotherapy. To improve the treatment of ovarian cancer and identify ovarian cancer‐specific antibodies, we immunized mice with the human ovarian carcinoma cell line, SKOV‐3, and generated hybridoma clones. Several rounds of screening yielded 30 monoclonal antibodies (mAbs) with no cross‐reactivity to normal cells. Among these mAbs, OV‐Ab 30‐7 was found to target integrin α3 and upregulate p53 and p21, while stimulating the apoptosis of cancer cells. We further found that binding of integrin α3 by OV‐Ab 30‐7 impaired laminin‐induced focal adhesion kinase phosphorylation. The mAb alone or in combination with carboplatin and paclitaxel inhibited tumor progression and prolonged survival of tumor‐bearing mice. Moreover, immunohistochemical staining of ovarian patient specimens revealed higher levels of integrin α3 in cancer cells compared with normal cells. By querying online clinical databases, we found that elevated ITGA3 expression in ovarian cancer is associated with poor prognosis. Taken together, our data suggest that the novel mAb, OV‐Ab 30‐7, may be considered as a potential therapeutic for ovarian cancer.
Collapse
Affiliation(s)
- Feng-Yi Ke
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Yu Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Ming-Chieh Lin
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chyi Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Kuan-Ting Kuo
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pathology and Laboratory Medicine, National Taiwan University Hospital Hsin-Chu Biomedical Park Branch, Hsinchu County, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
2
|
Lv J, Bai R, Wang L, Gao J, Zhang H. Artesunate may inhibit liver fibrosis via the FAK/Akt/β-catenin pathway in LX-2 cells. BMC Pharmacol Toxicol 2018; 19:64. [PMID: 30326962 PMCID: PMC6192352 DOI: 10.1186/s40360-018-0255-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/01/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND An increasing number of studies are investigating the effects of Chinese medicine on hepatic fibrosis, but only few studies have examined the anti-fibrogenic properties of Artesunate (ART). The aim of the present study was to explore the anti-fibrotic effects of ART on LX-2 cells, the human HSC cell line, and to determine potential molecular mechanisms via the focal adhesion kinase (FAK)/ protein kinase B (Akt)/ β-catenin pathway. METHODS LX-2 cells were stimulated with different concentration of ART (0, 12.5, 25 and 50 μg/ml) for 12, 24, 48 or 72 h, their proliferation was analyzed using the Cell Counting Kit-8 (CCK-8) assay. LX-2 cells were treated with different doses of ART (0, 12.5, 25 and 50 μg/ml) for 24 h, their apoptosis was measured using flow cytometry, the levels of mRNAs encoding collagen I or α-smooth muscle actin (α-SMA) were determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and the levels of key proteins in the FAK/Akt/β-catenin signaling pathway were assessed by western blotting. Specific inhibitors of FAK were added to the LX-2 cells cultures to explore the potential signaling. RESULTS Exposing LX-2 cells to ART efficiently inhibited their proliferation, significantly promoted early apoptosis in a dose-dependent manner, and markedly downregulated the mRNA expression of α-SMA and collagen I. In addition, ART, similar to FAK inhibitor PF562271 significantly inhibited the FAK/Akt/β-catenin signaling pathway by reducing the levels of phosphorylated FAK, Akt and GSK-3β. CONCLUSIONS Our present study shows that ART could regulate the proliferation, apoptosis and activation of LX-2. Meanwhile, the anti-fibrogenic mechanisms of ART was correlated with FAK/Akt/β-catenin pathway. Future research should verify and extend these findings, as well as explore other molecules and therefore serve as useful therapeutic targets.
Collapse
Affiliation(s)
- Jian Lv
- Department of Pharmacy, Renmin Hospital of Wuhan University, Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Ruidan Bai
- Department of Pharmacy, Renmin Hospital of Wuhan University, Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Li Wang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Jiefang Gao
- Department of Pharmacy, Renmin Hospital of Wuhan University, Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Hong Zhang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
3
|
Adiponectin as an anti-fibrotic and anti-inflammatory adipokine in the liver. CURRENT PATHOBIOLOGY REPORTS 2015; 3:243-252. [PMID: 26858914 DOI: 10.1007/s40139-015-0094-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hepatic fibrosis is a dynamic process resulting from excessive deposition of extracellular matrix in the liver; uncontrolled progression of fibrosis can eventually lead to liver cirrhosis and/or hepatocellular carcinoma. The fibrogenic process is complex and modulated by a number of both hepatic and extra-hepatic biological factors. Growing evidence indicates that adipokines, a group of cytokines produced by adipose tissue, impart dynamic functions in liver and are involved in modulation of hepatic fibrosis. In particular, two key adipokines, adiponectin and leptin, directly regulate many biological responses closely associated with development and progression of hepatic fibrosis. Leptin acts as a pro-fibrogenic cytokine, while adiponectin possesses anti-fibrogenic and anti-inflammatory properties. Adiponectin, acting via its cognate receptors, adiponectin receptors 1 and 2, potently suppresses fibrosis and inflammation in liver via multiple mechanisms. This review summarizes recent findings concerning the role of adiponectin in fibrogenic process in liver and addresses the underlying molecular mechanisms in modulation of fibrosis.
Collapse
|
4
|
Liang NL, Men R, Zhu Y, Yuan C, Wei Y, Liu X, Yang L. Visfatin: an adipokine activator of rat hepatic stellate cells. Mol Med Rep 2014; 11:1073-8. [PMID: 25351242 DOI: 10.3892/mmr.2014.2795] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 09/29/2014] [Indexed: 02/05/2023] Open
Abstract
The present study was conducted to investigate the effects of visfatin on the activation of hepatic stellate cells (HSC) and the possible underlying mechanism. HSC were isolated from the livers of Sprague‑Dawley rats by in situ perfusion of collagenase and pronase and a single‑step density Nycodenz gradient. The culture‑activated cells were serum‑starved and incubated with different concentrations of recombinant visfatin (0, 25, 50, 100 or 200 ng/ml) for 24 h. The expression of α‑smooth muscle actin (α‑SMA), collagen types I and III and connective tissue growth factor (CTGF) were then measured by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis. The results demonstrated that 100 and 200 ng/ml concentrations of visfatin induced the expression of α‑SMA in culture‑activated rat HSC, which was accompanied by a significant increase in collagen types I and III, as confirmed by western blot and RT‑qPCR analyses. In addition, treatment of the HSC with certain concentrations of visfatin upregulated the expression of CTGF. These findings suggested that visfatin activated HSC and induced the production of collagen types I and III.
Collapse
Affiliation(s)
- Ning-Lin Liang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ruoting Men
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yongjun Zhu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Cong Yuan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Wei
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
5
|
Kuo WL, Yu MC, Lee JF, Tsai CN, Chen TC, Chen MF. Imatinib mesylate improves liver regeneration and attenuates liver fibrogenesis in CCL4-treated mice. J Gastrointest Surg 2012; 16:361-369. [PMID: 22068968 DOI: 10.1007/s11605-011-1764-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 10/16/2011] [Indexed: 01/31/2023]
Abstract
BACKGROUNDS Imatinib mesylate (STI-571), a tyrosine kinase inhibitor, has previously been demonstrated to attenuate liver fibrogenesis through inhibition of the activation of hepatic stellate cells (HSCs) in CCL(4)-treated rat models. AIMS This study aimed to further evaluate the role of STI-571 in liver regeneration. MATERIALS AND METHODS All animals were divided into four groups, and mice were treated with or without CCL(4) and STI-571 (n = 6 for each group). RESULTS Activated cultured HSCs in vitro with STI-571 administration showed increased apoptosis and reduced proliferation, as determined by flow cytometric analysis, 3-(4, 5-cimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay, and confocal microscopy. STI-571 treatment attenuated liver fibrosis in vivo, as was evident in the results of histology, mRNA level, and expression analysis of smooth muscle actin and type I collagen. Mice treated with STI-571 had increased liver weight ratio and the improvement in liver regeneration was compatible with the change of serum interleukin 6 levels (p < 0.05). Further, increased apoptosis and a reduced proliferation were observed in the CCL(4)-treated mice after STI-571 treatment based on the immunohistochemical staining of Annexin V, phosphorylated STAT3, and PCNA. CONCLUSION STI-571 treatment effectively attenuated liver fibrogenesis and improved in liver regeneration in vivo and induced apoptosis in HSCs both in vitro and in vivo.
Collapse
Affiliation(s)
- Wen-Ling Kuo
- Department of Surgery, Chang Gung Memorial Hospital and Medical School of Chang Gung University, Kuei-Shan, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
6
|
Kurenova EV, Hunt DL, He D, Magis AT, Ostrov DA, Cance WG. Small molecule chloropyramine hydrochloride (C4) targets the binding site of focal adhesion kinase and vascular endothelial growth factor receptor 3 and suppresses breast cancer growth in vivo. J Med Chem 2009; 52:4716-24. [PMID: 19610651 PMCID: PMC2765121 DOI: 10.1021/jm900159g] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
FAK is a tyrosine kinase that functions as a key orchestrator of signals leading to invasion and metastasis. Since FAK interacts directly with a number of critical proteins involved in survival signaling in tumor cells, we hypothesized that targeting a key protein-protein interface with druglike small molecules was a feasible strategy for inhibiting tumor growth. In this study, we targeted the protein-protein interface between FAK and VEGFR-3 and identified compound C4 (chloropyramine hydrochloride) as a drug capable of (1) inhibiting the biochemical function of VEGFR-3 and FAK, (2) inhibiting proliferation of a diverse set of cancer cell types in vitro, and (3) reducing tumor growth in vivo. Chloropyramine hydrochloride reduced tumor growth as a single agent, while concomitant administration with doxorubicin had a pronounced synergistic effect. Our data demonstrate that the FAK-VEGFR-3 interaction can be targeted by small druglike molecules and this interaction can provide the basis for highly specific novel cancer therapeutics.
Collapse
Affiliation(s)
- Elena V. Kurenova
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Darell L. Hunt
- Department of Surgery, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Dihua He
- Department of Surgery, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Andrew T. Magis
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - David A. Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - William G. Cance
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
7
|
Luo M, Guan JL. Focal adhesion kinase: a prominent determinant in breast cancer initiation, progression and metastasis. Cancer Lett 2009; 289:127-39. [PMID: 19643531 DOI: 10.1016/j.canlet.2009.07.005] [Citation(s) in RCA: 218] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 07/08/2009] [Accepted: 07/10/2009] [Indexed: 11/30/2022]
Abstract
Focal adhesion kinase (FAK) is an intracellular non-receptor tyrosine kinase. In addition to its role as a major mediator of signal transduction by integrins, FAK also participates in signaling by a wide range of extracellular stimuli including growth factors, G-protein-coupled receptor agonists, cytokines, and other inflammatory mediators. The link between FAK and breast cancers is strongly suggested by a number of reports showing that FAK gene is amplified and overexpressed in a large fraction of breast cancer specimens. In addition, increased FAK expression and activity frequently correlate with metastatic disease and poor prognosis. Since its discovery in early 1990s, numerous studies have shown a role for FAK in the regulation of cell spreading, adhesion, migration, survival, proliferation, differentiation, and angiogenesis. Many of these studies in cultured cells provided strong evidence to connect FAK expression/activation to the promotion of cancer. Recently, a prominent role of FAK in promoting mammary tumorigenesis, progression and metastasis has been unveiled by different animal models of human breast cancer, including xenograft models in immunodeficient rodents and spontaneous tumor models in transgenic mice that have specific deletion of FAK in the mammary epithelial cells during embryonic or postnatal development. These in vivo studies established FAK as a prominent determinant in mammary cancer initiation, progression and metastasis. Furthermore, a novel function of FAK in maintaining mammary cancer stem/progenitor cells in vivo has been recently reported, which may provide a novel cellular mechanism of FAK in promoting breast cancer initiation and progression. The wealth of knowledge accumulated over almost two decades of research on FAK should help to design potentially novel therapies for breast cancer.
Collapse
Affiliation(s)
- Ming Luo
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
8
|
Elsharkawy AM, Oakley F, Mann DA. The role and regulation of hepatic stellate cell apoptosis in reversal of liver fibrosis. Apoptosis 2006; 10:927-39. [PMID: 16151628 DOI: 10.1007/s10495-005-1055-4] [Citation(s) in RCA: 325] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Liver fibrosis and its end-stage disease cirrhosis are major world health problems arising from chronic injury of the liver by a variety of etiological factors including viruses, alcohol and drug abuse, the metabolic syndrome, autoimmune disease and hereditary disorders of metabolism. Fibrosis is a progressive pathological process in which wound-healing myofibroblasts of the liver respond to injury by promoting replacement of the normal hepatic tissue with a scar-like matrix composed of cross-linked collagen. Until recently it was believed that this process was irreversible. However emerging experimental and clinical evidence is starting to show that even cirrhosis is potentially reversible. Key to this is the discovery that reversion of fibrosis is accompanied by clearance of hepatic stellate cells (HSC) by apoptosis. Furthermore, proof-of-concept studies in rodents have demonstrated that experimental augmentation of HSC apoptosis will promote the resolution of fibrosis. Consequently there is now considerable interest in determining the molecular events that regulate HSC apoptosis and the discovery of drugs that will stimulate HSC apoptosis in a selective manner. This review will consider the regulatory role played by growth factors (e.g. NGF, IGF-1, TGFbeta), death receptor ligands (TRAIL, FAS), components and regulators of extracellular matrix (integrins, collagen, matrix metalloproteinases and their tissue inhibitors) and signal transduction proteins and transcription factors (Rho/Rho kinase, Jun N-terminal Kinase (JNK), IkappaKinase (IKK), NF-kappa B). The potential for known pharmacological agents such as gliotoxin, sulfasalazine, benzodiazepine ligands, curcumin and tanshinone I to induce HSC apoptosis and therefore to be used therapeutically will be explored.
Collapse
Affiliation(s)
- A M Elsharkawy
- Liver Group, Division of Infection, Inflammation and Repair, University of Southampton, Southampton General Hospital, Level D, South Academic Block, Southampton, SO16 6YD, UK
| | | | | |
Collapse
|
9
|
Sawai H, Okada Y, Funahashi H, Matsuo Y, Takahashi H, Takeyama H, Manabe T. Activation of focal adhesion kinase enhances the adhesion and invasion of pancreatic cancer cells via extracellular signal-regulated kinase-1/2 signaling pathway activation. Mol Cancer 2005; 4:37. [PMID: 16209712 PMCID: PMC1266395 DOI: 10.1186/1476-4598-4-37] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2005] [Accepted: 10/06/2005] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Interaction with integrin and focal adhesion kinase (FAK) regulates the cancer cell adhesion and invasion into extracellular matrix (ECM). In addition, phosphorylation of FAK correlates with the increase of cell motility and invasion. Adhesion and spreading of cancer cells on a variety of ECM proteins, including collagen type IV (Coll IV), leads to an increase in tyrosine phosphorylation and activation of FAK. In this study, we investigated the mechanism of activation of FAK and its downstream extracellular signal-regulated kinase (ERK)-1/2 signaling following stimulation by interleukin (IL)-1alpha and adhesion to ECM with subsequent enhancement of pancreatic cancer cell adhesion and invasion. RESULTS In immunoblotting analysis, all three pancreatic cancer cell lines (AsPC-1, BxPC-3, and Capan-2) expressed the protein of FAK and beta1 integrin. Enhancement of FAK protein association with beta1 integrin when cells were plated on Coll IV was more increased by stimulation with IL-1alpha. Preincubation with anti-beta1 integrin antibody and FAK siRNA transfection inhibited the association of FAK with beta1 integrin of pancreatic cancer cells. FAK phosphorylation was observed by adhesion to Coll IV, furthermore, stronger FAK phosphorylation was observed by stimulation with IL-1alpha of pancreatic cancer cells adhered to Coll IV in time-dependent manner. Genistein, a tyrosine kinase inhibitor, markedly inhibited the FAK phosphorylation. IL-1alpha stimulation and Coll IV adhesion enhanced the activation of Ras, as evidenced by the increased Ras-GTP levels in pancreatic cancer cells. Activation of Ras correlated with the phosphorylation of ERK. While not statistical affecting the apoptosis of pancreatic cancer cells, IL-1alpha-induced adhesion and invasion on Coll IV were inhibited with FAK gene silencing by siRNA, beta1 integrin blocking, and inhibition of FAK phosphorylation. PD98059, a MEK inhibitor, also inhibited IL-1alpha-induced enhancement of adhesion and invasion in pancreatic cancer cells. CONCLUSION Our results demonstrated that activation of FAK is involved with the aggressive capability in pancreatic cancer through Ras/ERK signaling pathway. Based on our results, we suggest that the modification of IL-1, FAK, and integrins functions might be a novel therapeutic approach to aggressive spread of pancreatic cancer.
Collapse
Affiliation(s)
- Hirozumi Sawai
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Yuji Okada
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hitoshi Funahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hiromitsu Takeyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Tadao Manabe
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| |
Collapse
|
10
|
McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer - a new therapeutic opportunity. Nat Rev Cancer 2005; 5:505-15. [PMID: 16069815 DOI: 10.1038/nrc1647] [Citation(s) in RCA: 803] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Focal-adhesion kinase (FAK) is an important mediator of growth-factor signalling, cell proliferation, cell survival and cell migration. Given that the development of malignancy is often associated with perturbations in these processes, it is not surprising that FAK activity is altered in cancer cells. Mouse models have shown that FAK is involved in tumour formation and progression, and other studies showing that FAK expression is increased in human tumours make FAK a potentially important new therapeutic target.
Collapse
Affiliation(s)
- Gordon W McLean
- The Beatson Institute for Cancer Research, Cancer Research UK Beatson Laboratories, Garscube Estate Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
| | | | | | | | | | | |
Collapse
|
11
|
Kurenova E, Xu LH, Yang X, Baldwin AS, Craven RJ, Hanks SK, Liu ZG, Cance WG. Focal adhesion kinase suppresses apoptosis by binding to the death domain of receptor-interacting protein. Mol Cell Biol 2004; 24:4361-71. [PMID: 15121855 PMCID: PMC400455 DOI: 10.1128/mcb.24.10.4361-4371.2004] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Tumor cells resist the apoptotic stimuli associated with invasion and metastasis by activating survival signals that suppress apoptosis. Focal adhesion kinase (FAK), a tyrosine kinase that is overexpressed in a variety of human tumors, mediates one of these survival signals. Attenuation of FAK expression in tumor cells results in apoptosis that is mediated by caspase 8- and FADD-dependent pathways, suggesting that death receptor pathways are involved in the process. Here, we report a functional link between FAK and death receptors. We have demonstrated that FAK binds to the death domain kinase receptor-interacting protein (RIP). RIP is a major component of the death receptor complex and has been shown to interact with Fas and tumor necrosis factor receptor 1 through its binding to adapter proteins. We have shown that RIP provides proapoptotic signals that are suppressed by its binding to FAK. We thus propose that FAK overexpression in human tumors provides a survival signal function by binding to RIP and inhibiting its interaction with the death receptor complex.
Collapse
Affiliation(s)
- Elena Kurenova
- Department of Surgery, University of Florida, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhou X, Murphy FR, Gehdu N, Zhang J, Iredale JP, Benyon RC. Engagement of alphavbeta3 integrin regulates proliferation and apoptosis of hepatic stellate cells. J Biol Chem 2004; 279:23996-4006. [PMID: 15044441 DOI: 10.1074/jbc.m311668200] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Hepatic stellate cells are the major source of the extracellular matrix that accumulates in fibrotic liver. During progressive liver fibrosis, hepatic stellate cells proliferate, but during resolution of fibrosis there is extensive stellate cell apoptosis that coincides with degradation of the liver scar. We have examined the possibility that the fate of stellate cells is influenced by the extracellular matrix through the intermediary of alpha(v)beta(3) integrin. alpha(v)beta(3) integrin was expressed by activated, myofibroblastic rat and human stellate cells in culture. Antagonism of this integrin using neutralizing antibodies, echistatin, or small inhibitory RNA to silence alpha(v) subunit expression inhibited stellate cell proliferation and their expression of proliferating cell nuclear antigen and activated forms of p44 and p42 MAPK. These alpha(v)beta(3) antagonists also increased apoptosis of cultured stellate cells, and this was associated with an increase in the BAX/BCL-2 protein ratio, induction of nuclear DNA fragmentation, and activation of intracellular caspase-3. Expression of tissue inhibitor of metalloproteinases-1 by activated stellate cells was reduced by the alpha(v)beta(3) antagonists, while matrix metalloproteinase-9 synthesis was enhanced. Stellate cells incubated with active recombinant matrix metalloproteinase-9 showed enhanced apoptosis, while cells treated with a synthetic inhibitor of this protease showed increased survival. Our studies suggest that alpha(v)beta(3) integrin regulates the fate of hepatic stellate cells. Degradation of alpha(v)beta(3) ligands surrounding activated stellate cells during resolution of liver fibrosis might decrease alpha(v)beta(3) integrin ligation, suppressing stellate cell proliferation and inducing a fibrolytic, matrix metalloproteinase-secreting phenotype that may prime stellate cells for apoptosis.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Liver Research Group, University Division of Infection, Inflammation and Repair, Southampton General Hospital, United Kingdom
| | | | | | | | | | | |
Collapse
|
13
|
Wang DS, Dou KF, Li KZ, Gao ZQ, Song ZS, Liu ZC. Hepatocellular apoptosis after hepatectomy in obstructive jaundice in rats. World J Gastroenterol 2003; 9:2737-41. [PMID: 14669324 PMCID: PMC4612043 DOI: 10.3748/wjg.v9.i12.2737] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the hepatocellular apoptosis after hepatectomy in obstructive jaundice and biliary decompression rats.
METHODS: After bile duct ligation for 7 days, rats were randomly divided into OB group in which the rats underwent 70% hepatectomy, OB-CD group in which the rats underwent hepatectomy accompanied by choledochoduodenostomy, CD-Hx group in which the rats underwent choledochoduodenostomy and then received 70% hepatectomy on the fifth day after biliary decompression. The control group (Hx group) only underwent hepatectomy.
RESULTS: The level of total serum bilirubin and serum enzymes was significantly lower in CD-Hx group than in OB-CD and OB groups on day 1, 3 and 5 after hepatectomy. The apoptotic index was significantly lower in CD-Hx group than in OB-CD and OB groups on day 3 and 5. The oligonucleosomal DNA fragments and Caspase-3 activity were also lower in CD-Hx group than in OB-CD and OB groups 3 days after hepatectomy, without differences between CD-Hx and Hx groups.
CONCLUSION: Hepatocellular apoptosis plays vital roles in jaundice rats, and biliary decompression is more effective in treatment of patients with severe jaundice before operation.
Collapse
Affiliation(s)
- De-Sheng Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, Shannxi Province, China.
| | | | | | | | | | | |
Collapse
|
14
|
Li MY, Deng H, Zhao JM, Dai D, Tan XY. Peroxisome proliferator-activated receptor gamma ligands inhibit cell growth and induce apoptosis in human liver cancer BEL-7402 cells. World J Gastroenterol 2003; 9:1683-8. [PMID: 12918101 PMCID: PMC4611524 DOI: 10.3748/wjg.v9.i8.1683] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the characteristics of PPAR gamma ligands induced apoptosis in liver cancer cells.
METHODS: The effects of ligands for each of the PPAR gamma ligands on DNA synthesis and cell viability were examined in BEL-7402 liver cancer cells. Apoptosis was characterized by Hochest33258 staining, DNA fragmentation, TUNEL and ELISA, and cell cycle kinetics by FACS. Modulation of apoptosis related caspases expression by PPAR gamma ligands was examined by Western blot.
RESULTS: PPARgamma ligands, 15-deoxy-12, 14-prostaglandin J2 (15d-PGJ2) and troglitazone (TGZ), suppressed DNA synthesis of BEL-7402 cells. Both 15d-PGJ2 and TGZ induced BEL-7402 cell death in a dose dependent manner, which was associated with an increase in fragmented DNA and TUNEL-positive cells. At concentrations of 10 and 30 µM, 15d-PGJ2 or troglitazone increased the proportion of cells with G0/G1 phase DNA content and decreased those with S phase DNA content. There was no significant change in the proportion of cells with G2/M DNA content. The activities of Caspases-3, -6, -7 and -9 were increased by 15d-PGJ2 and TGZ treatment, while the activity of Caspase 8 had not significantly changed.
CONCLUSION: The present results suggest the potential usefulness of PPAR gamma ligands for chemoprevention and treatment of liver cancers.
Collapse
Affiliation(s)
- Ming-Yi Li
- Department of General Surgery, Affiliated Hospital of Guangdong Medical College, Zhangjiang 524001, Guangdong Province, China.
| | | | | | | | | |
Collapse
|
15
|
Liu QH, Li DG, Huang X, You HN, Pan Q, Xu LM, Xu QF, Lu HM. Effect of Activin on extracelluar matrix secretion in isolated rat hepatic stellate cell. Shijie Huaren Xiaohua Zazhi 2003; 11:745-748. [DOI: 10.11569/wcjd.v11.i6.745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of activin A on the extracelluar matrix secretion of rat hepatic stellate cell.
METHODS Hepatic stellate cells were isolated and purified from normal male Sprague-Dawley rat liver by a combination of pronase-collagenase perfusion and density gradient centrifugation. Passaged hepatic stellate cells were divided randomly into eight groups: control group(A group), ACTA 1 μg/L group (B group), ACTA 10 μg/L group(C group), ACTA 100 μg/L group (D group), TGF β1 10 μg/L group(E group), TGF β1 10 μg/L plus ACTA 1 μg/L group(F group), TGF β1 10 μg/L plus ACTA 10 μg/L group(G group), TGF β1 10 μg/L plus ACTA 100 μg/L group(H group). 24 h after incubation secretion of procollagen Ⅲ, collagen Ⅳ and mRNA of collagen Ⅲ in hepatic stellate cells were detected by radioimmunoassays and semi-quantitative RT-PCR method respectively.
RESULTS Extracellular matrix secretion in passaged hepatic stellate cells was enhanced by activin A according to its concentration, the capacity of extracellular matrix secretion by 100 μg/L activin A was equal to that of 10 μg/L TGF β1, extracellular matrix secretion and type Ⅲ collagen mRNA expression in passaged hepatic stellate cells was enhanced by activin A and TGFβ1 in a synergistic manner.
CONCLUSION Activin A may contribute to hepatic fibrogenesis.
Collapse
Affiliation(s)
- Qing-Hua Liu
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Ding-Guo Li
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Xin Huang
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Han-Ning You
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Qin Pan
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Lei-Ming Xu
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Qin-Fang Xu
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| | - Han-Ming Lu
- Department of Gastroenterology of Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
| |
Collapse
|
16
|
Liu XJ, Yang L, Mao YQ, Wang Q, Huang MH, Wang YP, Wu HB. Effects of the tyrosine protein kinase inhibitor genistein on the proliferation, activation of cultured rat hepatic stellate cells. World J Gastroenterol 2002; 8:739-45. [PMID: 12174389 PMCID: PMC4656331 DOI: 10.3748/wjg.v8.i4.739] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Hepatic stellate cell (HSC) plays a pivotal role in liver fibrosis and is considered as the therapeutic target for the treatment of hepatic fibrosis. Tyrosine protein kinase plays an important role in the proliferation, activation of HSC. The purpose of the study is to investigate the effects of the tyrosine protein kinase inhibitor genistein on the proliferation and activation of cultured rat HSC.
METHODS: Rat HSC were isolated from Wistar rats by in situ perfusion of collagenase and pronase and single-step density Nycodenz gradient. Culture-activated HSC were serum-starved and incubated with 10-9 to 10-5 mol/L concentration of genistein for 24, 48 or 72 h. In PDGF-induced HSC proliferation, HSC were stimulated with 10 μg·L-1 PDGF-BB for 15 min, and then treated with genistein for the same time. Cell proliferation was measured by MTT assay and based on flow cytometric analysis of cell cycle. The α-smooth muscle actin (α-SMA) expression in HSC was studied with confocal laser microscopy and flow cytometry. c-fos, c-jun and cyclin D1 expression in HSC was also detected by flow cytometry.
RESULTS: Genistein inhibited basal and PDGF-induced proliferation of HSC at the concentration of 10-8 to 10-5 mol/L, and treatment with 10-7 mol/L concentration of genistein for 48 h inhibited the HSC proliferation significantly (the inhibition rate was 70.3%, P < 0.05). Immunofluorescence detected by confocal laser microscopy and flow cytometry showed that treatment with 10-7 mol/L genistein for 48 h suppressed the expression of α-SMA significantly in HSC (the specific fluorescence intensity were 60.2 ± 21.5 vs 35.3 ± 11.6 and 12.8 ± 10.4 vs 9.54 ± 6.39, respectively, both P < 0.05). The intensity of c-fos, c-jun and cyclin D1 expression of HSCs treated with 10-7 mol/L genistein for 48 h was also significantly decreased compared with the controls.
CONCLUSION: Genistein influences proliferation of HSC, suppresses the expression of α-SMA in HSC and t inhibits the intensity of c-fos, c-jun and cyclin D1 expression of HSCs. Genistein has therapeutic potential against liver fibrosis.
Collapse
Affiliation(s)
- Xiao-Jing Liu
- Laboratory of Department of Internal Medicine, West China Hospital, Sichuan University, 37 Wainan Guoxueshang, Chengdu 610041, Sichuan Province, China.
| | | | | | | | | | | | | |
Collapse
|