51
|
Coleman SJ, Grose RP, Kocher HM. Fibroblast growth factor family as a potential target in the treatment of hepatocellular carcinoma. J Hepatocell Carcinoma 2014; 1:43-54. [PMID: 27508175 PMCID: PMC4918266 DOI: 10.2147/jhc.s48958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatocellular cancer (HCC) is currently the third leading cause of cancer death worldwide. The prognosis of patients diagnosed with late-stage disease is dismal due to high resistance to conventional systemic therapies. The introduction of sorafenib, despite its limited efficacy, as the standard systemic therapy for advanced HCC has paved a way for targeted molecular therapies for HCC. Fibroblast growth factor (FGF) signaling plays an important role in the developing embryo and the adult. The FGF signaling pathway is often hijacked by cancer cells, including HCC. Several alterations in FGF signaling correlate with poor outcome in HCC patients, suggesting that this family of signaling molecules plays an important role in the development of HCC. Multikinase inhibitors targeting FGF signaling are currently under investigation in clinical trials. This review discusses the current understanding of the biological and clinical implications of aberrant FGF signaling in the prognosis, diagnosis, and treatment of HCC.
Collapse
Affiliation(s)
- Stacey J Coleman
- Centre for Tumour Biology, Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, UK
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, UK; Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
52
|
Shi L, Feng Y, Lin H, Ma R, Cai X. Role of estrogen in hepatocellular carcinoma: is inflammation the key? J Transl Med 2014; 12:93. [PMID: 24708807 PMCID: PMC3992128 DOI: 10.1186/1479-5876-12-93] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/28/2014] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide and accounts for the third-leading cause of cancer-related deaths. Over the past decades, advances have been made in the field of surgery, but effective treatment of HCC is lacking. Due to a marked male predominance in morbidity and mortality in HCC patients, it has long been considered that sex hormones play a role in HCC development. Recently estrogen has been proven to exert protective effects against HCC through IL-6 restrictions, STAT3 inactivation and tumour-associated macrophage inhibition. While IL-6-dependent STAT3 activation is considered a key event in inflammation-induced liver cancer, the anti-inflammation effect of estrogen is well documented. The roles of the estrogen receptor and aromatase and interactions between microRNAs and estrogen in HCC have been investigated. In this review, we present a novel model to elucidate the mechanism of estrogen-mediated inhibition of HCC development through an anti-inflammation effect and provide new insights into the roles of estrogen in liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Xiujun Cai
- Chawnshang Chang Live Cancer Center, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
53
|
Fuchs BC, Hoshida Y, Fujii T, Wei L, Yamada S, Lauwers GY, McGinn CM, DePeralta DK, Chen X, Kuroda T, Lanuti M, Schmitt AD, Gupta S, Crenshaw A, Onofrio R, Taylor B, Winckler W, Bardeesy N, Caravan P, Golub TR, Tanabe KK. Epidermal growth factor receptor inhibition attenuates liver fibrosis and development of hepatocellular carcinoma. Hepatology 2014; 59:1577-90. [PMID: 24677197 PMCID: PMC4086837 DOI: 10.1002/hep.26898] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/10/2013] [Indexed: 12/13/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is the most rapidly increasing cause of cancer-related mortality in the United States. Because of the lack of viable treatment options for HCC, prevention in high-risk patients has been proposed as an alternative strategy. The main risk factor for HCC is cirrhosis and several lines of evidence implicate epidermal growth factor (EGF) in the progression of cirrhosis and development of HCC. We therefore examined the effects of the EGF receptor (EGFR) inhibitor erlotinib on liver fibrogenesis and hepatocellular transformation in three different animal models of progressive cirrhosis: a rat model induced by repeated, low-dose injections of diethylnitrosamine (DEN), a mouse model induced by carbon tetrachloride (CCl4 ), and a rat model induced by bile duct ligation (BDL). Erlotinib reduced EGFR phosphorylation in hepatic stellate cells (HSC) and reduced the total number of activated HSC. Erlotinib also decreased hepatocyte proliferation and liver injury. Consistent with all these findings, pharmacological inhibition of EGFR signaling effectively prevented the progression of cirrhosis and regressed fibrosis in some animals. Moreover, by alleviating the underlying liver disease, erlotinib blocked the development of HCC and its therapeutic efficacy could be monitored with a previously reported gene expression signature predictive of HCC risk in human cirrhosis patients. CONCLUSION These data suggest that EGFR inhibition using Food and Drug Administration-approved inhibitors provides a promising therapeutic approach for reduction of fibrogenesis and prevention of HCC in high-risk cirrhosis patients who can be identified and monitored by gene expression signatures.
Collapse
Affiliation(s)
- Bryan C. Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Yujin Hoshida
- Liver Cancer Program, Tisch Cancer Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Tsutomu Fujii
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Lan Wei
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Suguru Yamada
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Gregory Y. Lauwers
- Department of Pathology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Christopher M. McGinn
- Division of Thoracic Surgery, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Danielle K. DePeralta
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Xintong Chen
- Liver Cancer Program, Tisch Cancer Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Toshihiko Kuroda
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Michael Lanuti
- Division of Thoracic Surgery, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Anthony D. Schmitt
- Division of Thoracic Surgery, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| | - Supriya Gupta
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge MA, 02142
| | - Andrew Crenshaw
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge MA, 02142
| | - Robert Onofrio
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge MA, 02142
| | - Bradley Taylor
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge MA, 02142
| | - Wendy Winckler
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge MA, 02142
| | - Nabeel Bardeesy
- Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114
| | - Peter Caravan
- A.A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth St., Suite 2301, Charlestown MA 02129
| | - Todd R. Golub
- Cancer Program, Broad Institute, 7 Cambridge Center, Cambridge, MA, 02142,Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215,Howard Hughes Medical Institute, 20 Shattuck Street, Boston, MA, 02115
| | - Kenneth K. Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114
| |
Collapse
|
54
|
Berasain C, Elizalde M, Urtasun R, Castillo J, García-Irigoyen O, Uriarte I, Latasa MU, Prieto J, Avila MA. Alterations in the expression and activity of pre-mRNA splicing factors in hepatocarcinogenesis. Hepat Oncol 2014; 1:241-252. [PMID: 30190958 DOI: 10.2217/hep.13.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a molecularly complex tumor that is resistant to standard and targeted therapies, and thus a deadly disease. In this context, the identification of key alterations driving HCC development is therefore essential. The implementation of next-generation sequencing techniques has underscored earlier realizations of the marked dysregulation of pre-mRNA splicing in HCC. Impairments in alternative splicing may lead to the expression of protumorigenic protein isoforms and to the generation of unstable mRNA species. Mechanistically, mutations in key nucleotides are responsible for many of these alterations in different types of tumors. However, changes in the expression of factors involved in the regulation of the splicing machinery are also important determinants in the derangement of pre-mRNA splicing. Here we discuss recent reports on the alteration of splicing factors in HCC, the pathological significance of these changes, and the identification of cell signaling pathways leading to the missplicing of genes in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Carmen Berasain
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - María Elizalde
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Urtasun
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Josefa Castillo
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain
| | - Oihane García-Irigoyen
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Iker Uriarte
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Maria U Latasa
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Prieto
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Matías A Avila
- Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Division of Hepatology & Gene Therapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, CIMA, Avda, Pio XII, n55, 31008 Pamplona, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
55
|
Li Z, Luo H, Li C, Huo X, Yan C, Huang X, Al-Haddawi M, Mathavan S, Gong Z. Transcriptomic analysis of a transgenic zebrafish hepatocellular carcinoma model reveals a prominent role of immune responses in tumour progression and regression. Int J Cancer 2014; 135:1564-73. [PMID: 24550086 DOI: 10.1002/ijc.28794] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 01/23/2014] [Accepted: 01/27/2014] [Indexed: 12/23/2022]
Abstract
Using our previously established xmrk transgenic zebrafish, hepatocellular carcinoma (HCC) was generated by induced expression of xmrk, which encoded a hyperactive epidermal growth factor receptor (EGFR) homolog, and regressed by suppression of xmrk expression. To investigate molecular changes in liver tumour progression and regression, RNA-Seq was performed for induced HCC and early and late stages of liver tissues during tumour regression. We found that Xmrk-induced zebrafish HCC shared strong molecular characteristics with a human HCC subtype (S2), which shows activated Myc signalling, upregulated phosphor-S6 and epithelial cell adhesion molecule. In the HCC stage, there were enhanced proteasome, antigen processing and presentation, aminosugars metabolisms, p53 and cell cycle pathways. During tumour regression, the transcriptomic profile showed a reversed trend of molecular changes compared with human HCC progression. Interestingly, distinct immune responses in tumour progression and regression were observed, including increased major histocompatibility complex class I (MHCI) at the HCC stage, enriched immune cell trafficking signals and inflammation in early regression and enhanced MHCII in late regression. Both neutrophils and macrophages were enriched during tumour progression and regression; however, the distribution of neutrophils and macrophages in HCC was relatively uniform, whereas both types of immune cells were regionally clustered during tumour regression, especially with dominant blood vessel association of macrophage in late regression, suggesting differential functions of these immune cells in tumour progression and regression. As tumour regression in our model resembles the targeted inhibition of EGFR in cancer therapy, our observations may provide molecular insights into the targeted inhibition and highlight the importance of immune response in tumour regression.
Collapse
Affiliation(s)
- Zhen Li
- Department of Biological Sciences, National University of Singapore, Singapore; Human genetics 2, Genome Institute of Singapore, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Berasain C, Avila MA. Amphiregulin. Semin Cell Dev Biol 2014; 28:31-41. [PMID: 24463227 DOI: 10.1016/j.semcdb.2014.01.005] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/10/2014] [Accepted: 01/14/2014] [Indexed: 12/26/2022]
Abstract
Amphiregulin (AREG) is a ligand of the epidermal growth factor receptor (EGFR), a widely expressed transmembrane tyrosine kinase. AREG is synthesized as a membrane-anchored precursor protein that can engage in juxtacrine signaling on adjacent cells. Alternatively, after proteolytic processing by cell membrane proteases, mainly TACE/ADAM17, AREG is secreted and behaves as an autocrine or paracrine factor. AREG gene expression and release is induced by a plethora of stimuli including inflammatory lipids, cytokines, hormones, growth factors and xenobiotics. Through EGFR binding AREG activates major intracellular signaling cascades governing cell survival, proliferation and motility. Physiologically, AREG plays an important role in the development and maturation of mammary glands, bone tissue and oocytes. Chronic elevation of AREG expression is increasingly associated with different pathological conditions, mostly of inflammatory and/or neoplastic nature. Here we review the essential aspects of AREG structure, function and regulation, discuss the basis for its differential role within the EGFR family of ligands, and identify emerging aspects in AREG research with translational potential.
Collapse
Affiliation(s)
- Carmen Berasain
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain; CIBERehd, Clinica Universidad de Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain.
| | - Matías A Avila
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain; CIBERehd, Clinica Universidad de Navarra, Avda. Pio XII, n55, 31008 Pamplona, Spain.
| |
Collapse
|
57
|
Berasain C, Avila MA. The EGFR signalling system in the liver: from hepatoprotection to hepatocarcinogenesis. J Gastroenterol 2014; 49:9-23. [PMID: 24318021 DOI: 10.1007/s00535-013-0907-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 10/28/2013] [Indexed: 02/04/2023]
Abstract
The liver displays an outstanding wound healing and regenerative capacity unmatched by any other organ. This reparative response is governed by a complex network of inflammatory mediators, growth factors and metabolites that are set in motion in response to hepatocellular injury. However, when liver injury is chronic, these regenerative mechanisms become dysregulated, facilitating the accumulation of genetic alterations leading to unrestrained cell proliferation and the development of hepatocellular carcinoma (HCC). The epidermal growth factor receptor (EGFR or ErbB1) signaling system has been identified as a key player in all stages of the liver response to injury, from early inflammation and hepatocellular proliferation to fibrogenesis and neoplastic transformation. The EGFR system engages in extensive crosstalk with other signaling pathways, acting as a true signaling hub for other growth factors, cytokines and inflammatory mediators. Here, we briefly review essential aspects of the biology of the EGFR, the other ErbB receptors, and their ligands in liver injury, regeneration and HCC development. Some aspects of the preclinical and clinical experience with EGFR therapeutic targeting in HCC are also discussed.
Collapse
Affiliation(s)
- Carmen Berasain
- Division of Hepatology and Gene Therapy and CIBEREhd, CIMA-University of Navarra, Avda. Pio XII, n55, 31008, Pamplona, Spain,
| | | |
Collapse
|
58
|
Horváth Z, Kovalszky I, Fullár A, Kiss K, Schaff Z, Iozzo RV, Baghy K. Decorin deficiency promotes hepatic carcinogenesis. Matrix Biol 2013; 35:194-205. [PMID: 24361483 DOI: 10.1016/j.matbio.2013.11.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/26/2013] [Accepted: 11/26/2013] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma represents one of the most-rapidly spreading cancers in the world. In the majority of cases, an inflammation-driven fibrosis or cirrhosis precedes the development of the tumor. During malignant transformation, the tumor microenvironment undergoes qualitative and quantitative changes that modulate the behavior of the malignant cells. A key constituent for the hepatic microenvironment is the small leucine-rich proteoglycan decorin, known to interfere with cellular events of tumorigenesis mainly by blocking various receptor tyrosine kinases (RTK) such as EGFR, Met, IGF-IR, PDGFR and VEGFR2. In this study, we characterized cell signaling events evoked by decorin deficiency in two experimental models of hepatocarcinogenesis using thioacetamide or diethyl nitrosamine as carcinogens. Genetic ablation of decorin led to enhanced tumor occurrence as compared to wild-type animals. These findings correlated with decreased levels of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) and a concurrent elevation in retinoblastoma protein phosphorylation via cyclin dependent kinase 4. Decreased steady state p21(Waf1/Cip1) levels correlated with enhanced expression of transcription factor AP4, a known transcriptional repressor of p21(Waf1/Cip1), and enhanced c-Myc protein levels. In addition, translocation of β-catenin was a typical event in diethyl nitrosamine-evoked tumors. In parallel, decreased phosphorylation of both c-Myc and β-catenin was observed in Dcn(-/-) livers likely due to the hindered GSK3β-mediated targeting of these proteins to proteasomal degradation. We discovered that in a genetic background lacking decorin, four RTKs were constitutively activated (phosphorylated), including three known targets of decorin such as PDGFRα, EGFR, IGF-IR, and a novel RTK MSPR/RON. Our findings provide powerful genetic evidence for a crucial in vivo role of decorin during hepatocarcinogenesis as lack of decorin in the liver and hepatic stroma facilitates experimental carcinogenesis by providing an environment devoid of this potent pan-RTK inhibitor. Thus, our results support future utilization of decorin as an antitumor agent in liver cancer.
Collapse
Affiliation(s)
- Zsolt Horváth
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Alexandra Fullár
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Kiss
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zsuzsa Schaff
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
59
|
Xu WP, Yi M, Li QQ, Zhou WP, Cong WM, Yang Y, Ning BF, Yin C, Huang ZW, Wang J, Qian H, Jiang CF, Chen YX, Xia CY, Wang HY, Zhang X, Xie WF. Perturbation of MicroRNA-370/Lin-28 homolog A/nuclear factor kappa B regulatory circuit contributes to the development of hepatocellular carcinoma. Hepatology 2013; 58:1977-91. [PMID: 23728999 DOI: 10.1002/hep.26541] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/17/2003] [Accepted: 05/16/2013] [Indexed: 01/09/2023]
Abstract
UNLABELLED MicroRNA 370 (miR-370) is located within the DLK1/DIO3 imprinting region on human chromosome 14, which has been identified as a cancer-associated genomic region. However, the role of miR-370 in malignances remains controversial. Here, we report that miR-370 was repressed in human hepatocellular carcinoma (HCC) tissues and hepatoma cell lines. Using gain-of-function and loss-of-function experiments, we demonstrated that miR-370 inhibited the malignant phenotype of HCC cells in vitro. Overexpression of miR-370 inhibited growth and metastasis of HCC cells in vivo. Moreover, the RNA-binding protein, LIN28A, was identified as a direct functional target of miR-370, which, in turn, blocked the biogenesis of miR-370 by binding to its precursor. LIN28A also mediated the suppressive effects of miR-370 on migration and invasion of HCC cells by post-transcriptionally regulating RelA/p65, which is an important effector of the canonical nuclear factor kappa B (NF-κB) pathway. Interleukin-6 (IL-6), a well-known NF-κB downstream inflammatory molecule, reduced miR-370 but increased LIN28A levels in HCC. Furthermore, miR-370 levels were inversely correlated with LIN28A and IL-6 messenger RNA (mRNA) levels, whereas LIN28A mRNA expression was positively correlated with IL-6 expression in human HCC samples. Interestingly, reduction of miR-370 expression was associated with the development of HCC in rats, as well as with aggressive tumor behavior and short survival in HCC patients. CONCLUSIONS These data demonstrate the involvement of a novel regulatory circuit consisting of miR-370, LIN28A, RelA/p65 and IL-6 in HCC progression. Manipulating this feedback loop may have beneficial effect in HCC treatment.
Collapse
Affiliation(s)
- Wen-Ping Xu
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Huang P, Xu X, Wang L, Zhu B, Wang X, Xia J. The role of EGF-EGFR signalling pathway in hepatocellular carcinoma inflammatory microenvironment. J Cell Mol Med 2013; 18:218-30. [PMID: 24268047 PMCID: PMC3930409 DOI: 10.1111/jcmm.12153] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 09/04/2013] [Indexed: 12/14/2022] Open
Abstract
Epidermal growth factor (EGF) and their receptor (EGFR) play an important role in the development of cancer proliferation, and metastasis, although the mechanism remains unclear. The present study aimed at investigating the role of EGF-EGFR signalling pathway in the development of human hepatocellular carcinoma (HCC) inflammatory environment. Gene profiles of inflammatory cytokines from HCC were measured. Cell bio-behaviours of HCC with low or high metastasis were detected by the live cell monitoring system. Cell proliferation was measured by CCK8. The protein level of CXCL5 and CXCL8 was measured by ELISA. The phosphorylation of PI3K, ERK, MAPK was measured by western blot. EGF significantly induced cell proliferation in HepG2 cells, but not in HCCLM3 cells. EGF prompted the cell movement in both HepG2 and HCCLM3 and regulated the production of CXCL5 and CXCL8 from HCC, which were inhibited by EGFR inhibitor, Erk inhibitor (U0126), or PI3K inhibitors (BEZ-235 and SHBM1009). HCC proliferation, metastasis and production of inflammatory cytokines were regulated via EGF-EGFR signal pathways. CXCL5 could interact with CXCL8, possibly by CXCR2 or the cross-talk between CXCR2 and EGFR. EGF-EGFR signaling pathway can be the potential target of therapies for HCC.
Collapse
Affiliation(s)
- Peixin Huang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
61
|
Dong S, Li FQ, Zhang Q, Lv KZ, Yang HL, Gao Y, Yu JR. Expression and clinical significance of SHP2 in gastric cancer. J Int Med Res 2013; 40:2083-9. [PMID: 23321164 DOI: 10.1177/030006051204000605] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES To investigate the expression and clinical significance of the protein tyrosine phosphatase, nonreceptor type 11 (PTPN11 or SHP2) gene, which encodes Src homology 2 domain-containing phosphatase (SHP-2) in gastric cancer. METHODS SHP2 expression was detected by immunohistochemical staining and real-time quantitative reverse transcription-polymerase chain reaction in tissue samples of normal gastric mucosa and different grades of gastric cancer. Correlation between SHP2 expression and standard clinico pathological parameters was analysed. RESULTS Immunohistochemical staining revealed significantly higher rates of SHP2 expression in gastric cancer tissues (72.5%) versus normal gastric mucosa (21.9%). SHP-2 mRNA levels were also significantly higher in gastric cancer tissues versus normal gastric mucosa. SHP2 expression correlated significantly with tumour differentiation, clinical classification and lymph node metastases, but was independent of sex and age. CONCLUSIONS SHP-2 is upregulated in gastric cancer and may be related to the development of gastric cancer. SHP-2 may be a potential prognostic marker of, or a therapeutic target for, gastric cancer.
Collapse
Affiliation(s)
- S Dong
- Department of Gastroenterological Surgery, The First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, Zhejiang Province, China
| | | | | | | | | | | | | |
Collapse
|
62
|
Jiang YX, Yu WJ, Lin DL, Li H, Li YJ. Significance of expression of EGFR, Connexin43 and E-cadherin in primary hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2013; 21:2185-2191. [DOI: 10.11569/wcjd.v21.i22.2185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of EGFR, Connexin43 (Cx43) and E-cadherin in primary hepatocellular carcinoma (HCC) and to analyze their relationship with clinicopathological characteristics of HCC.
METHODS: Immunohistochemistry was used to detect the expression of EGFR, Cx43 and E-cadherin in 74 HCC tissue specimens, 25 liver cirrhosis tissue specimens and 20 normal liver tissue specimens.
RESULTS: The positive rates of EGFR, Cx43 and E-cadherin protein expression were 66.22%, 27.03% and 40.54% in HCC, 44.00%, 68.00% and 52.00% in liver cirrhosis, and 10.00%, 95.00% and 80.00% in normal liver tissues. Expression of EGFR was significantly higher in HCC than in liver cirrhosis (P < 0.05) and normal liver tissue (both P < 0.01). Expression of Cx43 was significantly lower in HCC than in liver cirrhosis (P < 0.05) and normal liver tissue (P < 0.01). Expression of E-cadherin differed significantly between HCC and normal liver tissue (P < 0.01). Expression of EGFR, Cx43 and E-cadherin was associated with tumor size, differentiation grade, TNM stage, presence of intravascular cancer embolus, capsule involvement and infiltrative growth pattern (all P < 0.05), but not with age, gender, cirrhosis background, cancer embolus in the portal vein or serum level of alpha-fetal protein (all P > 0.05). In HCC, expression of EGFR was up-regulated, while that of Cx43 and E-cadherin was down-regulated. Expression of EGFR was negatively correlated with expression of Cx43 (r = -0.3123, P = 0.0068) and E-cadherin (r = -0.2962, P = 0.0104).
CONCLUSION: Combined detection of EGFR, Cx43 and E-cadherin expression can help reveal the carcinogenic mechanisms of HCC. EGFR, Cx43 and E-cadherin might be used as novel and useful markers for predicting the invasion, metastasis and prognosis of HCC and guiding the clinical therapy of this malignancy.
Collapse
|
63
|
Hepatitis B Virus-Encoded X Protein Downregulates EGFR Expression via Inducing MicroRNA-7 in Hepatocellular Carcinoma Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:682380. [PMID: 23840262 PMCID: PMC3693120 DOI: 10.1155/2013/682380] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 05/23/2013] [Indexed: 12/18/2022]
Abstract
Hepatitis B virus (HBV) infection accounts for over a half of cases of hepatocellular carcinoma (HCC), the most frequent malignant tumor of the liver. HBV-encoded X (HBx) plays critical roles in HBV-associated hepatocarcinogenesis. However, it is unclear whether and how HBx regulates the expression of epidermal growth factor receptor (EGFR), an important gene for cell growth. Therefore, the study aimed to investigate the association between HBx and EGFR expression. In this study, we found that HBx upregulates miR-7 expression to target 3′UTR of EGFR mRNA, which in turn results in the reduction of EGFR protein expression in HCC cells. HBx-mediated EGFR suppression renders HCC cells a slow-growth behavior. Deprivation of HBx or miR-7 expression or restoration of EGFR expression can increase the growth rate of HCC cells. Our data showed the miR-7-dependent EGFR suppression by HBx, supporting an inhibitory role of HBx in the cell growth of HCC. These findings not only identify miR-7 as a novel regulatory target of HBx, but also suggest HBx-miR-7-EGFR as a critical signaling in controlling the growth rate of HCC cells.
Collapse
|
64
|
Experimental evidence of ω-3 polyunsaturated fatty acid modulation of inflammatory cytokines and bioactive lipid mediators: their potential role in inflammatory, neurodegenerative, and neoplastic diseases. BIOMED RESEARCH INTERNATIONAL 2013; 2013:743171. [PMID: 23691510 PMCID: PMC3652138 DOI: 10.1155/2013/743171] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/27/2013] [Accepted: 03/27/2013] [Indexed: 12/19/2022]
Abstract
A large body of evidence has emerged over the past years to show the critical role played by inflammation in the pathogenesis of several diseases including some cardiovascular, neoplastic, and neurodegenerative diseases, previously not considered inflammation-related. The anti-inflammatory action of ω-3 polyunsaturated fatty acids (PUFAs), as well as their potential healthy effects against the development and progression of the same diseases, has been widely studied by our and others' laboratories. As a result, a rethinking is taking place on the possible mechanisms underlying the beneficial effects of ω-3 PUFAs against these disorders, and, in particular, on the influence that they may exert on the molecular pathways involved in inflammatory process, including the production of inflammatory cytokines and lipid mediators active in the resolving phase of inflammation. In the present review we will summarize and discuss the current knowledge regarding the modulating effects of ω-3 PUFAs on the production of inflammatory cytokines and proresolving or protective lipid mediators in the context of inflammatory, metabolic, neurodegenerative, and neoplastic diseases.
Collapse
|
65
|
Calvisi DF, Frau M, Tomasi ML, Feo F, Pascale RM. Deregulation of signalling pathways in prognostic subtypes of hepatocellular carcinoma: novel insights from interspecies comparison. Biochim Biophys Acta Rev Cancer 2013; 1826:215-37. [PMID: 23393659 DOI: 10.1016/j.bbcan.2012.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma is a frequent and fatal disease. Recent researches on rodent models and human hepatocarcinogenesis contributed to unravel the molecular mechanisms of hepatocellular carcinoma dedifferentiation and progression, and allowed the discovery of several alterations underlying the deregulation of cell cycle and signalling pathways. This review provides an interpretive analysis of the results of these studies. Mounting evidence emphasises the role of up-regulation of RAS/ERK, P13K/AKT, IKK/NF-kB, WNT, TGF-ß, NOTCH, Hedgehog, and Hippo signalling pathways as well as of aberrant proteasomal activity in hepatocarcinogenesis. Signalling deregulation often occurs in preneoplastic stages of rodent and human hepatocarcinogenesis and progressively increases in carcinomas, being most pronounced in more aggressive tumours. Numerous changes in signalling cascades are involved in the deregulation of carbohydrate, lipid, and methionine metabolism, which play a role in the maintenance of the transformed phenotype. Recent studies on the role of microRNAs in signalling deregulation, and on the interplay between signalling pathways led to crucial achievements in the knowledge of the network of signalling cascades, essential for the development of adjuvant therapies of liver cancer. Furthermore, the analysis of the mechanisms involved in signalling deregulation allowed the identification of numerous putative prognostic markers and novel therapeutic targets of specific hepatocellular carcinoma subtypes associated with different biologic and clinical features. This is of prime importance for the selection of patient subgroups that are most likely to obtain clinical benefit and, hence, for successful development of targeted therapies for liver cancer.
Collapse
Affiliation(s)
- Diego F Calvisi
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | | | | | | | | |
Collapse
|
66
|
Abstract
Erlotinib is a small-molecular inhibitor of epidermal growth factor receptor (EGFR). Here, we identify that cancerous inhibitor of protein phosphatase 2A (CIP2A) is a major determinant mediating erlotinib-induced apoptosis in hepatocellular carcinoma (HCC). Erlotinib showed differential effects on apoptosis in 4 human HCC cell lines. Erlotinib induced significant apoptosis in Hep3B and PLC5 cell lines; however, Huh-7 and HA59T cell lines showed resistance to erlotinib-induced apoptosis at all tested doses. Down-regulation of CIP2A, a cellular inhibitor of protein phosphatase 2A (PP2A), mediated the apoptotic effect of erlotinib in HCC. Erlotinib inhibited CIP2A in a dose- and time-dependent manner in all sensitive HCC cells whereas no alterations in CIP2A were found in resistant cells. Overexpression of CIP2A upregulated phospho-Akt and protected Hep3B cells from erlotinib-induced apoptosis. In addition, silencing CIP2A by siRNA restored the effects of erlotinib in Huh-7 cells. Moreover, adding okadaic acid, a PP2A inhibitor, abolished the effects of erlotinib on apoptosis in Hep3B cells; and forskolin, a PP2A agonist enhanced the effect of erlotinib in resistant HA59T cells. Combining Akt inhibitor MK-2206 with erlotinib restored the sensitivity of HA59T cells to erlotinib. Furthermore, in vivo xenograft data showed that erlotinib inhibited the growth of PLC5 tumor but had no effect on Huh-7 tumor. Erlotinib downregulated CIP2A and upregulated PP2A activity in PLC5 tumors, but not in Huh-7 tumors. In conclusion, inhibition of CIP2A determines the effects of erlotinib on apoptosis in HCC. CIP2A may be useful as a therapeutic biomarker for predicting clinical response to erlotinib in HCC treatment.
Collapse
|
67
|
Yang Z, Wu Q, Shi Y, Nie Y, Wu K, Fan D. Epidermal growth factor 61A>G polymorphism is associated with risk of hepatocellular carcinoma: a meta-analysis. Genet Test Mol Biomarkers 2012; 16:1086-91. [PMID: 22852842 DOI: 10.1089/gtmb.2012.0050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The association between hepatocellular carcinoma (HCC) and the 61A>G polymorphism in the epidermal growth factor (EGF) gene has been analyzed in several studies, but results have been inconsistent. The aim of this study was to integrate previous findings and explore whether this polymorphism is associated with susceptibility to HCC. A meta-analysis was performed by searching PubMed, Web of Science, and Cochrane Library databases. Data were extracted using predefined form and pooled odds ratios (OR) with 95% confidence intervals (CI) and were calculated to evaluate the strength of this association. Five studies involving 690 cases, 514 healthy controls, and 1419 controls with cancer-free liver diseases were identified. On the basis of healthy controls, the significant main effects on HCC risk were observed in a heterozygote comparison (OR=1.76, 95% CI 1.07-2.90, p=0.02) and a dominant genetic model (OR=1.65, 95% CI 1.03-2.66, p=0.04). On the basis of the controls with cancer-free liver diseases, a significantly increased risk of HCC was found in all the genetic models. Subgroup analyses stratified by ethnicity and etiology of HCC also showed positive associations. The EGF 61G allele is a risk factor for developing HCC without the influence of ethnic and etiological diversity.
Collapse
Affiliation(s)
- Zhiping Yang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | |
Collapse
|
68
|
Enterococcus faecalis enhances cell proliferation through hydrogen peroxide-mediated epidermal growth factor receptor activation. Infect Immun 2012; 80:3545-58. [PMID: 22851748 DOI: 10.1128/iai.00479-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enterococcus faecalis is a member of the intestinal and oral microbiota that may affect the etiology of colorectal and oral cancers. The mechanisms by which E. faecalis may contribute to the initiation and progression of these cancers remain uncertain. Epidermal growth factor receptor (EGFR) signaling is postulated to play a crucial role in oral carcinogenesis. A link between E. faecalis and EGFR signaling in oral cancer has not been elucidated. The present study aimed to evaluate the association between E. faecalis and oral cancer and to determine the underlying mechanisms that link E. faecalis to EGFR signaling. We report the high frequency of E. faecalis infection in oral tumors and the clinical association with EGFR activation. Using human oral cancer cells, we support the clinical findings and demonstrate that E. faecalis can induce EGFR activation and cell proliferation. E. faecalis activates EGFR through production of H(2)O(2), a signaling molecule that activates several signaling pathways. Inhibitors of H(2)O(2) (catalase) and EGFR (gefitinib) significantly blocked E. faecalis-induced EGFR activation and cell proliferation. Therefore, E. faecalis infection of oral tumor tissues suggests a possible association between E. faecalis infection and oral carcinogenesis. Interaction of E. faecalis with host cells and production of H(2)O(2) increase EGFR activation, thereby contributing to cell proliferation.
Collapse
|
69
|
Sanchez-Guerrero E, Chen E, Kockx M, An SW, Chong BH, Khachigian LM. IL-1beta signals through the EGF receptor and activates Egr-1 through MMP-ADAM. PLoS One 2012; 7:e39811. [PMID: 22792188 PMCID: PMC3391205 DOI: 10.1371/journal.pone.0039811] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 05/27/2012] [Indexed: 01/22/2023] Open
Abstract
The immediate-early gene Egr-1 controls the inducible expression of many genes implicated in the pathogenesis of a range of vascular disorders, yet our understanding of the mechanisms controlling the rapid expression of this prototypic zinc finger transcription factor is poor. Here we show that Egr-1 expression induced by IL-1beta is dependent on metalloproteinases (MMP) and a disintegrin and a metalloproteinase (ADAM). Pharmacologic MMP/ADAM inhibitors and siRNA knockdown prevent IL-1beta induction of Egr-1. Further, IL-1beta activates Egr-1 via the epidermal growth factor receptor (EGFR). This is blocked by EGFR tyrosine kinase inhibition and EGFR knockdown. IL-1beta induction of Egr-1 expression is reduced in murine embryonic fibroblasts (mEFs) deficient in ADAM17 despite unbiased expression of EGFR and IL-1RI in ADAM17-deficient and wild-type mEFs. Finally, we show that IL-1beta-inducible wound repair after mechanical injury requires both EGFR and MMP/ADAM. This study reports for the first time that Egr-1 induction by IL-1beta involves EGFR and MMP/ADAM-dependent EGFR phosphorylation.
Collapse
Affiliation(s)
| | - Elya Chen
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Maaike Kockx
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Si-Wei An
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Beng H. Chong
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Levon M. Khachigian
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
70
|
Esmat G, El-Bendary M, Zakarya S, Ela MA, Zalata K. Role of Helicobacter pylori in patients with HCV-related chronic hepatitis and cirrhosis with or without hepatocellular carcinoma: possible association with disease progression. J Viral Hepat 2012; 19:473-9. [PMID: 22676359 DOI: 10.1111/j.1365-2893.2011.01567.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of Helicobacter hepaticus as a causal agent of hepatitis and hepatocellular carcinoma (HCC) in mice has stimulated interest in looking for Helicobacter species in human liver samples. In this study, we searched for association between H. pylori and HCV-related liver disease. Liver specimens were collected from eighty-five patients; they were divided into five different groups according to liver pathology (METAVIR system). Group I (the 1st control group) consisted of 16 patients with chronic hepatitis C without histological activity. Group II consisted of 25 patients with chronic active hepatitis C, Group III, 17 patients with HCV-related cirrhosis and Group IV, 16 patients with HCV-related cirrhosis and HCC. Group V (2nd control group) consisted of 11 patients suffering from gastro duodenal and gall bladder diseases but negative for HCV. All cases were tested by polymerase chain reaction on liver samples for the presence of H. pylori DNA Cag A gene. Routine biochemical, radiological and RT-PCR for HCV RNA were also performed for all cases. The positivity of H. pylori PCR CagA gene in liver tissue was directly proportional to the severity of liver pathology, this being 75%, 52.9% and 32% in groups IV, III and II, respectively, which was more significant than the 1st and 2nd control groups (P < 0.001). There was a significant difference between H. pylori PCR values when compared to METAVIR staging (F) in different groups (P = 0.001). Helicobacter pylori PCR (Cag A gene) was positive in about 28.2% cases of late fibrosis (F3 + F4) while positivity was (5.9%) in early fibrosis (F1 + F2) (P = 0.0001). There was significant difference between H. pylori PCR (Cag A gene) in liver tissue and METAVIR activity in different groups (P = 0.002) as most of H. pylori PCR-positive cases were METAVIR activity A1 and A2 (15.3% and 12.9%, respectively). There was no association between H. pylori PCR and quantitative HCV RNA (P = 0.531). Also there was no significant difference of Child-Pugh staging in the H. pylori PCR-positive group when compared to the negative group (P = 0.996). There may be an association between the presence of H. pylori (Cag A gene) in the liver and disease progression in HCV-related chronic hepatitis and cirrhosis with and without HCC.
Collapse
Affiliation(s)
- G Esmat
- Department of Tropical Medicine, Cairo University Hospital, Cairo Faculty of Medicine, Cairo, Egypt
| | | | | | | | | |
Collapse
|
71
|
Han W, Carpenter RL, Cao X, Lo HW. STAT1 gene expression is enhanced by nuclear EGFR and HER2 via cooperation with STAT3. Mol Carcinog 2012; 52:959-69. [PMID: 22693070 DOI: 10.1002/mc.21936] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/04/2012] [Accepted: 05/21/2012] [Indexed: 01/05/2023]
Abstract
Both EGFR and HER2 are important mediators of tumorigenesis and tumor progression. Despite their best-characterized roles as plasma membrane-bound receptors, both receptors undergo nuclear translocation though the impact of this process remains unclear. In this study, we provide evidence showing that EGFR upregulates expression of signal transducer and activator of transcription 1 (STAT1), a transcription factor responding to inflammatory signals and regulating genes involved in inflammatory response. EGFR regulation of STAT1 expression is primarily attributed to the nuclear activity of EGFR. The oncogenic transcription factor STAT3 binds to the STAT1 promoter and synergizes with nuclear EGFR to significantly enhance STAT1 gene expression. Structural characterization of the human STAT1 gene promoter indicates the presence of four functional STAT3-binding sites in the promoter and their importance in STAT1 co-regulation by EGFR and STAT3. The constitutively activated EGFR variant, EGFRvIII, also cooperates with STAT3 to activate the STAT1 gene promoter through the identified STAT3-binding sites within the promoter. Using human breast cancer cell lines, we further found a positive association between levels of STAT1, EGFR, and p-STAT3. Furthermore, we found that STAT1 expression is transcriptionally upregulated by HER2 and heregulin stimulation in breast cancer cells, and the level is further augmented by activated STAT3. In summary, we report in this study that STAT1 expression is upregulated by nuclear EGFR, EGFRvIII and HER2, and that STAT3 synergizes with the three receptors to further enhance STAT1 expression. These novel findings establish a novel link between the mitogenic ErbB signaling pathway and the inflammatory pathway mediated by STAT1.
Collapse
Affiliation(s)
- Woody Han
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710
| | | | | | | |
Collapse
|
72
|
Sung TI, Wang YJ, Chen CY, Hung TL, Guo HR. Increased serum level of epidermal growth factor receptor in liver cancer patients and its association with exposure to arsenic. THE SCIENCE OF THE TOTAL ENVIRONMENT 2012; 424:74-78. [PMID: 22446113 DOI: 10.1016/j.scitotenv.2012.02.079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 02/29/2012] [Accepted: 02/29/2012] [Indexed: 05/31/2023]
Abstract
Arsenic is a human carcinogen and can activate epidermal growth factor receptor (EGFR) in human cell lines. As EGFR is associated with the occurrence of cancers, we conducted a study to evaluate whether serum EGFR may increase in liver cancer patients, particularly in those with exposure to arsenic. We recruited 100 patients of liver cancer and 100 age- and sex-matched controls in Taiwan and determined EGFR levels in sera by enzyme-linked immunosorbent assay. The patients had higher EGFR levels (668.1 vs. 243.1 fmol/mL, p<0.01), and after adjusting for hepatitis B and C, they still had an average EGFR level 406.1 fmol/mL higher than that of the controls (p<0.01). When we compared 22 patients residing in an endemic area of arsenic intoxication to 22 age- and sex-matched patients residing outside the area, we found that patients from the endemic area had higher EGFR levels (882.8 vs. 511.6 fmol/mL, p = 0.04). We concluded that EGFR is over-expressed in patients of liver cancer, particularly in those with exposure to arsenic, and therefore, serum EGFR level is not only a potential biomarker of liver cancer, but also a potential biomarker of cancers associated with arsenic exposure.
Collapse
Affiliation(s)
- Tzu-I Sung
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
73
|
Lee S, Springstead JR, Parks BW, Romanoski CE, Palvolgyi R, Ho T, Nguyen P, Lusis AJ, Berliner JA. Metalloproteinase processing of HBEGF is a proximal event in the response of human aortic endothelial cells to oxidized phospholipids. Arterioscler Thromb Vasc Biol 2012; 32:1246-54. [PMID: 22402363 DOI: 10.1161/atvbaha.111.241257] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Atherosclerosis is a chronic inflammatory disease initiated by monocyte recruitment and retention in the vessel wall. An important mediator of monocyte endothelial interaction is the chemokine interleukin (IL)-8. The oxidation products of phospholipids, including oxidized 1-palmitoyl-2-arachidonyl-sn-glycerol-3-phosphocholine (Ox-PAPC), accumulate in atherosclerotic lesions and strongly induce IL-8 in human aortic endothelial cells (HAECs). The goal of this study was to identify the proximal events leading to induction of IL-8 by Ox-PAPC in vascular endothelial cells. METHODS AND RESULTS In a systems genetics analysis of HAECs isolated from 96 different human donors, we showed that heparin-binding EGF-like growth factor (HBEGF) transcript levels are strongly correlated to IL-8 induction by Ox-PAPC. The silencing and overexpression of HBEGF in HAECs confirmed the role of HBEGF in regulating IL-8 expression. HBEGF has been shown to be stored in an inactive form and activation is dependent on processing by a dysintegrin and metalloproteinases (ADAM) to a form that can activate the epidermal growth factor (EGF) receptor. Ox-PAPC was shown to rapidly induce HBEGF processing and EGF receptor activation in HAECs. Using siRNA we identified 3 ADAMs that regulate IL-8 induction and directly demonstrated that Ox-PAPC increases ADAM activity in the cells using a substrate cleavage assay. We provide evidence for one mechanism of Ox-PAPC activation of ADAM involving covalent binding of Ox-PAPC to cysteine on ADAM. Free thiol cysteine analogs showed inhibition of IL-8 induction by Ox-PAPC, and both a cysteine analog and a cell surface thiol blocker strongly inhibited ADAM activity induction by Ox-PAPC. Using microarray analyses, we determined that this ADAM pathway may regulate at least 30% of genes induced by Ox-PAPC in HAECs. CONCLUSIONS This study is the first report demonstrating a role for the ADAM-HBEGF-EGF receptor axis in Ox-PAPC induction of IL-8 in HAECs. These studies highlight a role for specific ADAMs as initiators of Ox-PAPC action and provide evidence for a role of covalent interaction of Ox-PAPC in activation of ADAMs.
Collapse
Affiliation(s)
- Sangderk Lee
- Department of Pathology, University of California-Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Li Z, Huang X, Zhan H, Zeng Z, Li C, Spitsbergen JM, Meierjohann S, Schartl M, Gong Z. Inducible and repressable oncogene-addicted hepatocellular carcinoma in Tet-on xmrk transgenic zebrafish. J Hepatol 2012; 56:419-25. [PMID: 21888874 DOI: 10.1016/j.jhep.2011.07.025] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 06/24/2011] [Accepted: 07/16/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Liver cancer, mainly hepatocellular carcinoma, is a major malignancy and currently there are no effective treatment protocols due to insufficient understanding of hepatocarcinogenesis. As a potentially high-throughput and cost-effective experimental model, the zebrafish is increasingly recognized for disease studies. Here, we aim at using the zebrafish to generate a convenient hepatocellular carcinoma model. METHODS Using the Tet-on system for liver-specific expression of fish oncogene xmrk, a hyperactive version of epidermal growth factor receptor homolog, we have generated transgenic zebrafish with inducible development of liver cancer. RESULTS Liver tumors were rapidly induced with 100% penetrance in both juvenile and adult xmrk transgenic fish. Histological examination indicated that they all showed features of hepatocellular carcinoma. The induced liver tumors regressed rapidly upon inducer withdrawal. During the tumor induction stage, we detected increased cell proliferation and activation of Xmrk downstream targets Erk and Stat5, which were important for liver tumorigenesis as proved by inhibition experiments. When tumors regressed, there were decreased phosphorylated Erk and Stat5 accompanied with an increase in apoptosis. CONCLUSIONS Our zebrafish model demonstrates the potential of a hyperactivated epidermal growth factor receptor pathway in initiating heptocarcinogenesis. It provides clear evidence for the requirement of only a single oncogene for HCC initiation and maintenance and is thus a convenient model for further investigation of oncogene addiction and future anti-cancer drug screening.
Collapse
Affiliation(s)
- Zhen Li
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Jyonouchi H, Geng L, Streck DL, Toruner GA. Immunological characterization and transcription profiling of peripheral blood (PB) monocytes in children with autism spectrum disorders (ASD) and specific polysaccharide antibody deficiency (SPAD): case study. J Neuroinflammation 2012; 9:4. [PMID: 22226452 PMCID: PMC3275444 DOI: 10.1186/1742-2094-9-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/07/2012] [Indexed: 12/24/2022] Open
Abstract
Introduction There exists a small subset of children with autism spectrum disorders (ASD) characterized by fluctuating behavioral symptoms and cognitive skills following immune insults. Some of these children also exhibit specific polysaccharide antibody deficiency (SPAD), resulting in frequent infection caused by encapsulated organisms, and they often require supplemental intravenous immunoglobulin (IVIG) (ASD/SPAD). This study assessed whether these ASD/SPAD children have distinct immunological findings in comparison with ASD/non-SPAD or non-ASD/SPAD children. Case description We describe 8 ASD/SPAD children with worsening behavioral symptoms/cognitive skills that are triggered by immune insults. These ASD/SPAD children exhibited delayed type food allergy (5/8), treatment-resistant seizure disorders (4/8), and chronic gastrointestinal (GI) symptoms (5/8) at high frequencies. Control subjects included ASD children without SPAD (N = 39), normal controls (N = 37), and non-ASD children with SPAD (N = 12). Discussion and Evaluation We assessed their innate and adaptive immune responses, by measuring the production of pro-inflammatory and counter-regulatory cytokines by peripheral blood mononuclear cells (PBMCs) in responses to agonists of toll like receptors (TLR), stimuli of innate immunity, and T cell stimulants. Transcription profiling of PB monocytes was also assessed. ASD/SPAD PBMCs produced less proinflammatory cytokines with agonists of TLR7/8 (IL-6, IL-23), TLR2/6 (IL-6), TLR4 (IL-12p40), and without stimuli (IL-1ß, IL-6, and TNF-α) than normal controls. In addition, cytokine production of ASD/SPAD PBMCs in response to T cell mitogens (IFN-γ, IL-17, and IL-12p40) and candida antigen (Ag) (IL-10, IL-12p40) were less than normal controls. ASD/non-SPAD PBMDs revealed similar results as normal controls, while non-ASD/SPAD PBMCs revealed lower production of IL-6, IL-10 and IL-23 with a TLR4 agonist. Only common features observed between ASD/SPAD and non-ASD/SPAD children is lower IL-10 production in the absence of stimuli. Transcription profiling of PB monocytes revealed over a 2-fold up (830 and 1250) and down (653 and 1235) regulation of genes in ASD/SPAD children, as compared to normal (N = 26) and ASD/non-SPAD (N = 29) controls, respectively. Enriched gene expression of TGFBR (p < 0.005), Notch (p < 0.01), and EGFR1 (p < 0.02) pathways was found in the ASD/SPAD monocytes as compared to ASD/non-SPAD controls. Conclusions The Immunological findings in the ASD/SPAD children who exhibit fluctuating behavioral symptoms and cognitive skills cannot be solely attributed to SPAD. Instead, these findings may be more specific for ASD/SPAD children with the above-described clinical characteristics, indicating a possible role of these immune abnormalities in their neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Harumi Jyonouchi
- Division of Allergy/Immunology and Infectious Diseases, Department of pediatrics, UMDN-NJMS, 185 South Orange Ave, Newark, NJ 07101-1709, USA.
| | | | | | | |
Collapse
|
76
|
Li Q, Yu CH, Yu JH, Liu L, Xie SS, Li WW, Yang X, Fan WB, Gai ZT, Chen SJ, Kato N. ABO blood group and the risk of hepatocellular carcinoma: a case-control study in patients with chronic hepatitis B. PLoS One 2012; 7:e29928. [PMID: 22235351 PMCID: PMC3250489 DOI: 10.1371/journal.pone.0029928] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 12/06/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Studies have observed an association between the ABO blood group and risk of certain malignancies. However, no studies of the association with hepatocellular carcinoma (HCC) risk are available. We conducted this hospital-based case-control study to examine the association with HCC in patients with chronic hepatitis B (CHB). METHODS From January 2004 to December 2008, a total of 6275 consecutive eligible patients with chronic hepatitis B virus (HBV) infection were recruited. 1105 of them were patients with HBV-related HCC and 5,170 patients were CHB without HCC. Multivariate logistic regression models were used to investigate the association between the ABO blood group and HCC risk. RESULTS Compared with subjects with blood type O, the adjusted odds ratio (AOR) for the association of those with blood type A and HCC risk was 1.39 [95% confidence interval (CI), 1.05-1.83] after adjusting for age, sex, type 2 diabetes, cirrhosis, hepatitis B e antigen, and HBV DNA. The associations were only statistically significant [AOR (95%CI) = 1.56(1.14-2.13)] for men, for being hepatitis B e antigen positive [AOR (95%CI) = 4.92(2.83-8.57)], for those with cirrhosis [AOR (95%CI), 1.57(1.12-2.20)], and for those with HBV DNA≤10(5)copies/mL [AOR (95%CI), 1.58(1.04-2.42)]. Stratified analysis by sex indicated that compared with those with blood type O, those with blood type B also had a significantly high risk of HCC among men, whereas, those with blood type AB or B had a low risk of HCC among women. CONCLUSIONS The ABO blood type was associated with the risk of HCC in Chinese patients with CHB. The association was gender-related.
Collapse
Affiliation(s)
- Qiang Li
- Division of Liver Disease, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
| | - Cui-Hua Yu
- Department of Pharmacy, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
| | - Jin-Hong Yu
- Division of Liver Disease, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
| | - Li Liu
- Division of Liver Disease, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
| | - Shuang-Shuang Xie
- Division of Liver Disease, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
| | - Wen-Wen Li
- Unit of Disease Control Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Xia Yang
- Unit of Hyperbaric Medicine, Shandong Province Hospital, Jinan, China
| | - Wen-Bo Fan
- Unit of Digestive Disease, Department of Internal Medicine, Qihe People's Hospital, Shandong, China
| | - Zhong-Tao Gai
- Department of Infectious Disease, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
- * E-mail: (S-JC); mailto: (Z-TG)
| | - Shi-Jun Chen
- Division of Liver Disease, Jinan Infectious Disease Hospital, Shandong University, Jinan, China
- * E-mail: (S-JC); mailto: (Z-TG)
| | - Naoya Kato
- Unit of Disease Control Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
77
|
Montaseri A, Busch F, Mobasheri A, Buhrmann C, Aldinger C, Rad JS, Shakibaei M. IGF-1 and PDGF-bb suppress IL-1β-induced cartilage degradation through down-regulation of NF-κB signaling: involvement of Src/PI-3K/AKT pathway. PLoS One 2011; 6:e28663. [PMID: 22194879 PMCID: PMC3237481 DOI: 10.1371/journal.pone.0028663] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 11/12/2011] [Indexed: 01/24/2023] Open
Abstract
Objective Interleukin-1β (IL-1β) is a pro-inflammatory cytokine that plays a key role in the pathogenesis of osteoarthritis (OA). Growth factors (GFs) capable of antagonizing the catabolic actions of cytokines may have therapeutic potential in the treatment of OA. Herein, we investigated the potential synergistic effects of insulin-like growth factor (IGF-1) and platelet-derived growth factor (PDGF-bb) on different mechanisms participating in IL-1β-induced activation of nuclear transcription factor-κB (NF-κB) and apoptosis in chondrocytes. Methods Primary chondrocytes were treated with IL-1β to induce dedifferentiation and co-treated with either IGF-1 or/and PDGF-bb and evaluated by immunoblotting and electron microscopy. Results Pretreatment of chondrocytes with IGF-1 or/and PDGF-bb suppressed IL-1β-induced NF-κB activation via inhibition of IκB-α kinase. Inhibition of IκB-α kinase by GFs led to the suppression of IκB-α phosphorylation and degradation, p65 nuclear translocation and NF-κB-regulated gene products involved in inflammation and cartilage degradation (COX-2, MMPs) and apoptosis (caspase-3). GFs or BMS-345541 (specific inhibitor of the IKK) reversed the IL-1β-induced down-regulation of collagen type II, cartilage specific proteoglycans, β1-integrin, Shc, activated MAPKinase, Sox-9 and up-regulation of active caspase-3. Furthermore, the inhibitory effects of IGF-1 or/and PDGF-bb on IL-1β-induced NF-κB activation were sensitive to inhibitors of Src (PP1), PI-3K (wortmannin) and Akt (SH-5), suggesting that the pathway consisting of non-receptor tyrosine kinase (Src), phosphatidylinositol 3-kinase and protein kinase B must be involved in IL-1β signaling. Conclusion The results presented suggest that IGF-1 and PDGF-bb are potent inhibitors of IL-1β-mediated activation of NF-κB and apoptosis in chondrocytes, may be mediated in part through suppression of Src/PI-3K/AKT pathway, which may contribute to their anti-inflammatory effects.
Collapse
Affiliation(s)
- Azadeh Montaseri
- Department of Anatomical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Franziska Busch
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Ali Mobasheri
- Division of Veterinary Medicine, School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington, United Kingdom
| | - Constanze Buhrmann
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Constance Aldinger
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Jafar Soleimani Rad
- Department of Anatomical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
78
|
Caja L, Sancho P, Bertran E, Ortiz C, Campbell JS, Fausto N, Fabregat I. The tyrphostin AG1478 inhibits proliferation and induces death of liver tumor cells through EGF receptor-dependent and independent mechanisms. Biochem Pharmacol 2011; 82:1583-92. [DOI: 10.1016/j.bcp.2011.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 12/15/2022]
|
79
|
Urtasun R, Latasa MU, Demartis MI, Balzani S, Goñi S, Garcia-Irigoyen O, Elizalde M, Azcona M, Pascale RM, Feo F, Bioulac-Sage P, Balabaud C, Muntané J, Prieto J, Berasain C, Avila MA. Connective tissue growth factor autocriny in human hepatocellular carcinoma: oncogenic role and regulation by epidermal growth factor receptor/yes-associated protein-mediated activation. Hepatology 2011; 54:2149-58. [PMID: 21800344 DOI: 10.1002/hep.24587] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
UNLABELLED The identification of molecular mechanisms involved in the maintenance of the transformed phenotype of hepatocellular carcinoma (HCC) cells is essential for the elucidation of therapeutic strategies. Here, we show that human HCC cells display an autocrine loop mediated by connective tissue growth factor (CTGF) that promotes DNA synthesis and cell survival. Expression of CTGF was stimulated by epidermal growth factor receptor (EGFR) ligands and was dependent on the expression of the transcriptional coactivator, Yes-associated protein (YAP). We identified elements in the CTGF gene proximal promoter that bound YAP-enclosing complexes and were responsible for basal and EGFR-stimulated CTGF expression. We also demonstrate that YAP expression can be up-regulated through EGFR activation not only in HCC cells, but also in primary human hepatocytes. CTGF contributed to HCC cell dedifferentiation, expression of inflammation-related genes involved in carcinogenesis, resistance toward doxorubicin, and in vivo HCC cell growth. Importantly, CTGF down-regulated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 2 expression and was involved in the reduced sensitivity of these cells toward TRAIL-mediated apoptosis. CONCLUSION We have identified autocrine CTGF as a novel determinant of HCC cells' neoplastic behavior. Expression of CTGF can be stimulated through the EGFR-signaling system in HCC cells in a novel cross-talk with the oncoprotein YAP. Moreover, to our knowledge, this is the first study that identifies a signaling mechanism triggering YAP gene expression in healthy and transformed liver parenchymal cells.
Collapse
Affiliation(s)
- Raquel Urtasun
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Dissecting the effect of targeting the epidermal growth factor receptor on TGF-β-induced-apoptosis in human hepatocellular carcinoma cells. J Hepatol 2011; 55:351-8. [PMID: 21147185 DOI: 10.1016/j.jhep.2010.10.041] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 10/06/2010] [Accepted: 10/25/2010] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS Transforming growth factor-beta (TGF-β) induces apoptosis in hepatocytes, a process that is inhibited by the epidermal growth factor receptor (EGFR) pathway. The aim of this work was to ablate EGFR in hepatocellular carcinoma (HCC) cells to understand its role in impairing TGF-β-induced cell death. METHODS Response to TGF-β in terms of apoptosis was analyzed in different HCC cell lines and the effect of canceling EGFR expression was evaluated. RESULTS TGF-β induces apoptosis in some HCC cells (such as Hep3B, PLC/PRF/5, Huh7, or SNU449), but it also mediates survival signals, coincident with the up-regulation of EGFR ligands. Inhibition of the EGFR, either by targeted knock-down with specific siRNA or by pharmacological inhibition, significantly enhances apoptotic response. TGF-β treatment in EGFR targeted knock-down cells correlates with higher levels of the NADPH oxidase NOX4 and changes in the expression profile of BCL-2 and IAP families. However, other HCC cells, such as HepG2, which show over activation of the Ras/ERKs pathway, SK-Hep1, with an endothelial phenotype, or SNU398, where the TGF-β-Smad signaling is altered, show apoptosis resistance that is not restored through EGFR blockade. CONCLUSIONS The inhibition of EGFR in HCC may enhance TGF-β-induced pro-apoptotic signaling. However, this effect may only concern those tumors with an epithelial phenotype which do not bear alterations in TGF-β signaling nor exhibit an over-activation of the survival pathways downstream of the EGFR.
Collapse
|
81
|
Epidermal Growth Factor Receptor (EGFR) Crosstalks in Liver Cancer. Cancers (Basel) 2011; 3:2444-61. [PMID: 24212818 PMCID: PMC3757426 DOI: 10.3390/cancers3022444] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 05/06/2011] [Accepted: 05/12/2011] [Indexed: 01/11/2023] Open
Abstract
Hepatocarcinogenesis is a complex multistep process in which many different molecular pathways have been implicated. Hepatocellular carcinoma (HCC) is refractory to conventional chemotherapeutic agents, and the new targeted therapies are meeting with limited success. Interreceptor crosstalk and the positive feedback between different signaling systems are emerging as mechanisms of targeted therapy resistance. The identification of such interactions is therefore of particular relevance to improve therapeutic efficacy. Among the different signaling pathways activated in hepatocarcinogenesis the epidermal growth factor receptor (EGFR) system plays a prominent role, being recognized as a “signaling hub” where different extracellular growth and survival signals converge. EGFR can be transactivated in response to multiple heterologous ligands through the physical interaction with multiple receptors, the activity of intracellular kinases or the shedding of EGFR-ligands. In this article we review the crosstalk between the EGFR and other signaling pathways that could be relevant to liver cancer development and treatment.
Collapse
|
82
|
Ghosh P, Garcia-Marcos M, Farquhar MG. GIV/Girdin is a rheostat that fine-tunes growth factor signals during tumor progression. Cell Adh Migr 2011; 5:237-48. [PMID: 21546796 DOI: 10.4161/cam.5.3.15909] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
GIV/Girdin is a multidomain signaling molecule that enhances PI3K-Akt signals downstream of both G protein-coupled and growth factor receptors. We previously reported that GIV triggers cell migration via its C-terminal guanine-nucleotide exchange factor (GEF) motif that activates Gαi. Recently we discovered that GIV's C-terminus directly interacts with the epidermal growth factor receptor (EGFR), and when its GEF function is intact, a Gαi-GIV-EGFR signaling complex assembles. By coupling G proteins to growth factor receptors, GIV is uniquely poised to intercept the incoming receptor-initiated signals and modulate them via G protein intermediates. Subsequent work has revealed that expression of the highly specialized C-terminus of GIV undergoes a bipartite dysregulation during oncogenesis-full length GIV with an intact C-terminus is expressed at levels ~20-50-fold above normal in highly invasive cancer cells and metastatic tumors, but its C-terminus is truncated by alternative splicing in poorly invasive cancer cells and non-invasive tumors. The consequences of such dysregulation on graded signal transduction and cellular phenotypes in the normal epithelium and its implication during tumor progression are discussed herein. Based on the fact that GIV grades incoming signals initiated by ligand-activated receptors by linking them to cyclical activation of G proteins, we propose that GIV is a molecular rheostat for signal transduction.
Collapse
Affiliation(s)
- Pradipta Ghosh
- Department of Medicine, School of Medicine, University of California-San Diego, La Jolla, CA, USA.
| | | | | |
Collapse
|
83
|
Bhui K, Tyagi S, Srivastava AK, Singh M, Roy P, Singh R, Shukla Y. Bromelain inhibits nuclear factor kappa-B translocation, driving human epidermoid carcinoma A431 and melanoma A375 cells through G(2)/M arrest to apoptosis. Mol Carcinog 2011; 51:231-43. [PMID: 21432909 DOI: 10.1002/mc.20769] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 01/29/2011] [Accepted: 02/18/2011] [Indexed: 12/13/2022]
Abstract
Bromelain, obtained from pineapple, is already in use clinically as adjunct in chemotherapy. Our objective was to test its ability to act as a sole anti-cancer agent. Therefore, we describe its anti-proliferative, anti-inflammatory and subsequent anti-cancer effects in vitro, against human epidermoid carcinoma-A431 and melanoma-A375 cells. Bromelain exhibited reduction in proliferation of both these cell-lines and suppressed their potential for anchorage-independent growth. Further, suppression of inflammatory signaling by bromelain was evident by inhibition of Akt regulated-nuclear factor-kappaB activation via suppression of inhibitory-kappaBα phosphorylation and concomitant reduction in cyclooxygenase-2. Since, the inflammatory cascade is well-known to be closely allied to cancer; we studied the effect of bromelain on events/molecules central to it. Bromelain caused depletion of intracellular glutathione and generation of reactive oxygen-species followed by mitochondrial membrane depolarization. This led to bromelain-induced cell-cycle arrest at G(2)/M phase which was mediated by modulation of cyclin B1, phospho-cdc25C, Plk1, phospho-cdc2, and myt1. This was subsequently followed by induction of apoptosis, indicated by membrane-blebbing, modulation of Bax-Bcl-2 ratio, Apaf-1, caspase-9, and caspase-3; chromatin-condensation, increase in caspase-activity and DNA-fragmentation. Bromelain afforded substantial anti-cancer potential in these settings; hence we suggest it as a potential prospect for anti-cancer agent besides only an additive in chemotherapy.
Collapse
Affiliation(s)
- Kulpreet Bhui
- Proteomics Laboratory, Indian Institute of Toxicology Research, Lucknow, India
| | | | | | | | | | | | | |
Collapse
|
84
|
Cheng CY, Kuo CT, Lin CC, Hsieh HL, Yang CM. IL-1beta induces expression of matrix metalloproteinase-9 and cell migration via a c-Src-dependent, growth factor receptor transactivation in A549 cells. Br J Pharmacol 2010; 160:1595-610. [PMID: 20649564 DOI: 10.1111/j.1476-5381.2010.00858.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Interleukin (IL)-1beta-induced matrix metalloproteinase (MMP-9) expression is regulated by mitogen activated protein kinases (MAPKs) and NF-kappaB. IL-1beta also stimulates transactivation of growth factor receptors and phosphatidylinositol 3-kinase (PI3K)/Akt., leading to the expression of inflammatory proteins. Here, we investigated whether these transactivation mechanisms participated in IL-1beta-induced MMP-9 expression in A549 cells. EXPERIMENTAL APPROACH A549 cells were treated with/without pharmacological inhibitors and neutralizing antibody or transfected with dominant negative mutants and siRNA of particular protein kinases before stimulation with IL-1beta. Cell migration was measured by in vitro scratch assay. Expression and enzymatic activity of MMP-9 were analysed by Western blot and gelatin zymography. Transcriptional activity of MMP-9 was analysed by RT-PCR, chromatin immunoprecipitation and promoter assays. KEY RESULTS Inhibition of MMP-9 expression by inhibitors of Src (PP1), platelet-derived growth factor (PDGF) receptor and epithelial growth factor (EGF) receptor or transfection with siRNA for Src and Akt prevented IL-1beta-induced migration of A549 cells. These tyrosine kinases were involved through phosphorylation of Src, PDGF, or EGF receptors (EGFRs) via the formation of Src/PDGFR or Src/EGFR complexes, attenuated by PP1. IL-1beta-induced MMP-9 expression through EGFR transactivation was diminished by inhibitors of MMPs and heparin-binding EGF-like factor (HB-EGF), or a neutralizing HB-EGF antibody. IL-1beta-stimulated activation and translocation of Akt and NF-kappaB (p65); the recruitment of activated NF-kappaB (p65) to the MMP-9 promoter region was attenuated by LY294002. CONCLUSIONS AND IMPLICATIONS IL-1beta-induced MMP-9 expression and cell migration was mediated through c-Src-dependent transactivation of EGFR/PDGFR/PI3K/Akt linking to the NF-kappaB pathway in A549 cells.
Collapse
Affiliation(s)
- Ching-Yi Cheng
- Department of Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | | | |
Collapse
|
85
|
Hagymási K, Tulassay Z. [New possibilities of targeted therapy in the treatment of hepatocellular carcinoma with the help of molecular biology]. Orv Hetil 2010; 151:1763-8. [PMID: 20940115 DOI: 10.1556/oh.2010.28984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) has a poor prognosis. Approximately 85% of patients are not candidates for curative treatments at the time of diagnosis; hence palliative modalities (transcatheter arterial chemoembolisation, radiofrequency ablation, systemic chemotherapy) are used. Systemic chemotherapies have disappointing results. The increasing knowledge in the molecular biology of HCC will increase the possibilities of targeted therapy. The multi-tyrosine kinase inhibitor sorafenib is the only drug which has approved. The VEGF-inhibitors (bevacizumab, sunitinib), EGFR-blocker agents (erlotinib), as well as the inhibition of mTOR (rapamycin) are promising. Combination of sorafenib or other anti-angiogenic agents with local ablative procedure (transcatheter arterial chemoembolisation, radiofrequency ablation), or with curative hepatectomy also can be favorable. Alteration of Wnt pathway, retinoid compounds, inhibition of the cell cycle as well as the proteosome, and epigenetic therapy can be other potential promising targets in HCC.
Collapse
Affiliation(s)
- Krisztina Hagymási
- Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest.
| | | |
Collapse
|
86
|
Helicobacter pylori accelerates hepatic fibrosis by sensitizing transforming growth factor-β1-induced inflammatory signaling. J Transl Med 2010; 90:1507-16. [PMID: 20531291 DOI: 10.1038/labinvest.2010.109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Our earlier report has shown that Helicobacter pylori promoted hepatic fibrosis in a murine model. Herein, in order to elucidate the mechanism by which H. pylori accelerate liver fibrosis, the authors investigated the changes in expression levels of mitogen-activated protein kinases (MAPKs), p53-related proteins, antioxidants, and proinflammatory cytokines in liver samples. H. pylori infection enhanced CCl4-induced MAP kinase activation and p53 signaling pathway as well as Bax- and proliferating-cell nuclear antigen expressions, whereas H. pylori alone induced neither of these expressions nor hepatic fibrosis. Moreover, mRNA expressions of inflammatory cytokines, glutathione peroxidase expression, and the proliferative index were strongly augmented in livers of the H. pylori with CCl4 treatment group compared with those of the CCl4-alone treatment group, whereas there was no difference in apoptotic index between the two groups. Interestingly, H. pylori treatment increased the number of α-fetoprotein-expressing hepatocytes independently of CCl4 intoxication. In vitro analyses, using an immortalized rat hepatic stellate cell (HSC) line, revealed that H. pylori lysates increased the proliferation of HSCs, which was boosted by the addition of transforming growth factor-beta1 (TGF-β1). Furthermore, the treatment of H. pylori lysates promoted the translocation of nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) into the nucleus based on an increase in the degradation of NF-κB inhibitor alpha, in the presence of TGF-β1, as did H2O2 treatment. In conclusion, H. pylori infection along with an elevated TGF-β1 may accelerate hepatic fibrosis through increased TGF-β1-induced pro-inflammatory signaling pathways in HSCs. Moreover, H. pylori infection might increase the risk of TGF-β1-mediated tumorigenesis by disturbing the balance between apoptosis and proliferation of hepatocytes.
Collapse
|
87
|
Bergkvist GT, Yool DA. Epidermal growth factor receptor as a therapeutic target in veterinary oncology. Vet Comp Oncol 2010; 9:81-94. [PMID: 21569194 DOI: 10.1111/j.1476-5829.2010.00237.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor that stimulates cell proliferation and survival and becomes dysregulated in a range of solid tumours in man. It is recognized as a key oncogenic driver and has become a favoured therapeutic target and a prognostic and predictive marker of cancer in man. In animals, EGFR dysregulation is emerging as a potential factor in the development of a number of naturally occurring tumours including mammary, lung, glial and epithelial cancers. Comparative analyses suggest that these diseases share many features with equivalent diseases in man and EGFR may have value as a prognostic or a biological marker of animal disease. There is still little direct evidence that EGFR is a critical oncogenic driver in naturally occurring animal disease and there are no veterinary trials of EGFR-targeted therapy. These will be critical steps in establishing a role for EGFR in veterinary oncology.
Collapse
Affiliation(s)
- G T Bergkvist
- Royal (Dick) School of Veterinary Studies and Roslin Institute, Division of Veterinary Clinical Sciences, The University of Edinburgh, Hospital for Small Animals, Easter Bush Veterinary Centre, Roslin, Midlothian, UK.
| | | |
Collapse
|
88
|
Berasain C, Goñi S, Castillo J, Latasa MU, Prieto J, Ávila MA. Impairment of pre-mRNA splicing in liver disease: Mechanisms and consequences. World J Gastroenterol 2010; 16:3091-102. [PMID: 20593494 PMCID: PMC2896746 DOI: 10.3748/wjg.v16.i25.3091] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pre-mRNA splicing is an essential step in the process of gene expression in eukaryotes and consists of the removal of introns and the linking of exons to generate mature mRNAs. This is a highly regulated mechanism that allows the alternative usage of exons, the retention of intronic sequences and the generation of exonic sequences of variable length. Most human genes undergo splicing events, and disruptions of this process have been associated with a variety of diseases, including cancer. Hepatocellular carcinoma (HCC) is a molecularly heterogeneous type of tumor that usually develops in a cirrhotic liver. Alterations in pre-mRNA splicing of some genes have been observed in liver cancer, and although still scarce, the available data suggest that splicing defects may have a role in hepatocarcinogenesis. Here we briefly review the general mechanisms that regulate pre-mRNA splicing, and discuss some examples that illustrate how this process is impaired in liver tumorigenesis, and may contribute to HCC development. We believe that a more thorough examination of pre-mRNA splicing is still needed to accurately draw the molecular portrait of liver cancer. This will surely contribute to a better understanding of the disease and to the development of new effective therapies.
Collapse
|
89
|
Chalaris A, Adam N, Sina C, Rosenstiel P, Lehmann-Koch J, Schirmacher P, Hartmann D, Cichy J, Gavrilova O, Schreiber S, Jostock T, Matthews V, Häsler R, Becker C, Neurath MF, Reiss K, Saftig P, Scheller J, Rose-John S. Critical role of the disintegrin metalloprotease ADAM17 for intestinal inflammation and regeneration in mice. ACTA ACUST UNITED AC 2010; 207:1617-24. [PMID: 20603312 PMCID: PMC2916135 DOI: 10.1084/jem.20092366] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The protease a disintegrin and metalloprotease (ADAM) 17 cleaves tumor necrosis factor (TNF), L-selectin, and epidermal growth factor receptor (EGF-R) ligands from the plasma membrane. ADAM17 is expressed in most tissues and is up-regulated during inflammation and cancer. ADAM17-deficient mice are not viable. Conditional ADAM17 knockout models demonstrated proinflammatory activities of ADAM17 in septic shock via shedding of TNF. We used a novel gene targeting strategy to generate mice with dramatically reduced ADAM17 levels in all tissues. The resulting mice called ADAM17ex/ex were viable, showed compromised shedding of ADAM17 substrates from the cell surface, and developed eye, heart, and skin defects as a consequence of impaired EGF-R signaling caused by failure of shedding of EGF-R ligands. Unexpectedly, although the intestine of unchallenged homozygous ADAM17ex/ex mice was normal, ADAM17ex/ex mice showed substantially increased susceptibility to inflammation in dextran sulfate sodium colitis. This was a result of impaired shedding of EGF-R ligands resulting in failure to phosphorylate STAT3 via the EGF-R and, consequently, in defective regeneration of epithelial cells and breakdown of the intestinal barrier. Besides regulating the systemic availability of the proinflammatory cytokine TNF, our results demonstrate that ADAM17 is needed for vital regenerative activities during the immune response. Thus, our mouse model will help investigate ADAM17 as a potential drug target.
Collapse
Affiliation(s)
- Athena Chalaris
- Institute of Biochemistry, Christian-Albrechts-University Kiel, D-24098 Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Albitar L, Pickett G, Morgan M, Wilken JA, Maihle NJ, Leslie KK. EGFR isoforms and gene regulation in human endometrial cancer cells. Mol Cancer 2010; 9:166. [PMID: 20579378 PMCID: PMC2907331 DOI: 10.1186/1476-4598-9-166] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 06/25/2010] [Indexed: 11/10/2022] Open
Abstract
Background Epidermal growth factor (EGF) and its receptor (EGFR) constitute a principal growth-promoting pathway in endometrial cancer cells. Pre-clinical studies were undertaken to compare the expression of EGFR isoforms and the downstream effects of activating or blocking EGFR function in Ishikawa H cells, derived from a moderately differentiated type I endometrioid adenocarcinoma, or in Hec50co cells, derived from a poorly differentiated type II adenocarcinoma with papillary serous sub-differentiation. Results We investigated whether EGFR mutations are present in the tyrosine kinase domain (exons 18-22) of EGFR and also whether EGFR isoforms are expressed in the Ishikawa H or Hec50co cell lines. Sequence of the EGFR tyrosine kinase domain proved to be wild type in both cell lines. While both cell lines expressed full-length EGFR (isoform A), EGFR and sEGFR (isoform D) were expressed at significantly lower levels in Hec50co cells compared to Ishikawa H cells. Analysis of gene expression following EGF vs. gefitinib treatment (a small molecule EGFR tyrosine kinase inhibitor) was performed. Early growth response 1, sphingosine kinase 2, dual specificity phosphatase 6, and glucocorticoid receptor DNA binding factor 1 are members of a cluster of genes downstream of EGFR that are differentially regulated by treatment with EGF compared to gefitinib in Ishikawa H cells, but not in Hec50co cells. Conclusions Type I Ishikawa H and type II Hec50co endometrial carcinoma cells both express EGFR and sEGFR, but differ markedly in their responsiveness to the EGFR inhibitor gefitinib. This difference is paralleled by differences in the expression of sEGFR and EGFR, as well as in their transcriptional response following treatment with either EGF or gefitinib. The small cluster of differently regulated genes reported here in these type I vs. type II endometrial cancer-derived cell lines may identify candidate biomarkers useful for predicting sensitivity to EGFR blockade.
Collapse
Affiliation(s)
- Lina Albitar
- The Reproductive Molecular Biology Laboratory, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | | | |
Collapse
|
91
|
El Zein N, D'Hondt S, Sariban E. Crosstalks between the receptors tyrosine kinase EGFR and TrkA and the GPCR, FPR, in human monocytes are essential for receptors-mediated cell activation. Cell Signal 2010; 22:1437-47. [PMID: 20566383 DOI: 10.1016/j.cellsig.2010.05.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 05/18/2010] [Indexed: 12/22/2022]
Abstract
The G-protein coupled receptor (GPCR) fMLP receptor (FPR) and the two receptors tyrosine kinase (RTK), the nerve growth factor (NGF) receptor TrkA and the epidermal growth factor (EGF) receptor (EGFR) are involved in reactive oxygen species (ROS), matrix metalloproteinase-9 (MMP-9) production and CD11b membrane integrin upregulation. We show that in monocytes the three receptors crosstalk each other to modulate these pro-inflammatory mediators. Tyrphostin AG1478, the EGFR inhibitor, inhibits fMLP and NGF-associated ROS production, fMLP-associated CD11b upregulation and NGF-induced TrkA phosphorylation; K252a, the NGF receptor inhibitor, inhibits fMLP or EGF-associated ROS production, CD11b expression and EGF-induced EGFR phosphorylation; cyclosporine H, the FPR inhibitor inhibits EGF or NGF-associated ROS production, EGF-associated CD11b upregulation and prevents EGFR and TrkA phosphorylation by their respective ligand EGF and NGF. In response to fMLP, TrkA phosphorylation is inhibited by the EGFR inhibitor while EGFR phosphorylation is inhibited by the TrkA inhibitor. Receptor crosstalks are Src and ERK dependent. Down-regulation of each receptor by specific siRNA suppresses the ability of the two other receptors to promote ligand-mediated ERK phosphorylation and pro-inflammatory activities including ROS, MMP-9 production and CD11b upregulation. Thus, in monocytes GPCR ligands' activity involves activation of RTK while RTK-ligands activity engages GPCR-signalling molecules.
Collapse
Affiliation(s)
- Nabil El Zein
- Pediatric Oncology Laboratory, 1020 Brussels, Belgium.
| | | | | |
Collapse
|
92
|
Zhang Y, Jiang LC, Hu QG, Liu XW, Xiong J, Zheng QC. Involvement of Cdc42 in epidermal growth factor-stimulated formation of filopodia in HepG2 cells. Shijie Huaren Xiaohua Zazhi 2010; 18:443-446. [DOI: 10.11569/wcjd.v18.i5.443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the role of Cdc42 in epidermal growth factor (EGF)-stimulated formation of filopodia in HepG2 cells.
METHODS: HepG2 cells were divided into four groups: normal control group, EGF-treated group, small interfering RNA (siRNA) transfection group, and EGF-treated siRNA transfection group. The siRNA targeting the Cdc42 gene was introduced into HepG2 cells by liposome-mediated transfection. The expression of Cdc42 mRNA and protein was detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot, respectively. Actin-tracker green fluorescent probe was used to detect the formation of filopodia in HepG2 cells. Cell migration was measured by Transwell migration assay.
RESULTS: No significant differences were noted in the expression levels of total Cdc42 protein and Cdc42 mRNA between the EGF-treated group and the normal control group. However, the expression level of active Cdc42 protein was significantly higher in the EGF-treated group than in the normal control group (0.713 ± 0.021 vs 0.423 ± 0.015, P < 0.05). Transfection of siRNA targeting the Cdc42 gene could not only inhibit the expression of Cdc42 protein and mRNA (0.118 ± 0.017 vs 1.128 ± 0.024, and 0.351 ± 0.021 vs 0.936 ± 0.024, respectively; both P < 0.05) but also reduce filopodia formation and cell migration in EGF-treated cells (98.43 ± 3.11 vs 61.09 ± 3.58, 50.53 ± 2.34 and 62.73 ± 2.64, respectively; all P < 0.05).
CONCLUSION: Cdc42 activation is crucial for EGF-stimulated filopodia formation and cell migration in HepG2 cells.
Collapse
|
93
|
Friso A, Tomanin R, Salvalaio M, Scarpa M. Genistein reduces glycosaminoglycan levels in a mouse model of mucopolysaccharidosis type II. Br J Pharmacol 2010; 159:1082-91. [PMID: 20136838 DOI: 10.1111/j.1476-5381.2009.00565.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Mucopolysaccharidoses (MPS) are lysosomal storage disorders resulting from a deficit of specific lysosomal enzymes catalysing glycosaminoglycan (GAG) degradation. The typical pathology involves most of the organ systems, including the brain, in its severe forms. The soy isoflavone genistein has recently attracted considerable attention as it can reduce GAG synthesis in vitro. Furthermore, genistein is able to cross the blood-brain barrier in the rat. The present study was undertaken to assess the ability of genistein to reduce urinary and tissue GAG levels in vivo. EXPERIMENTAL APPROACH We used mice with genetic deletion of iduronate-2-sulphatase (one of the GAG catabolizing enzymes) which provide a model of MPS type II. Two doses of genistein, 5 or 25 mg.kg(-1).day(-1), were given, in the diet for 10 or 20 weeks. Urinary and tissue GAG content was evaluated by biochemical and histochemical procedures. KEY RESULTS Urinary GAG levels were reduced after 10 weeks' treatment with genistein at either 5 or 25 mg.kg(-1).day(-1). In tissue samples from liver, spleen, kidney and heart, a reduction in GAG content was observed with both dosages, after 10 weeks' treatment. Decreased GAG deposits in brain were observed after genistein treatment in some animals. CONCLUSIONS AND IMPLICATIONS There was decreased GAG storage in the MPSII mouse model following genistein administration. Our results would support the use of this plant-derived isoflavone in a combined therapeutic protocol for treatment of MPS.
Collapse
Affiliation(s)
- A Friso
- Department of Pediatrics, University of Padova, Italy
| | | | | | | |
Collapse
|