1
|
Willems RAL, Biesmans C, Campello E, Simioni P, de Laat B, de Vos-Geelen J, Roest M, Ten Cate H. Cellular Components Contributing to the Development of Venous Thrombosis in Patients with Pancreatic Cancer. Semin Thromb Hemost 2024; 50:429-442. [PMID: 38049115 DOI: 10.1055/s-0043-1777304] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive type of cancer and has a poor prognosis. Patients with PDAC are at high risk of developing thromboembolic events, which is a leading cause of morbidity and mortality following cancer progression. Plasma-derived coagulation is the most studied process in cancer-associated thrombosis. Other blood components, such as platelets, red blood cells, and white blood cells, have been gaining less attention. This narrative review addresses the literature on the role of cellular components in the development of venous thromboembolism (VTE) in patients with PDAC. Blood cells seem to play an important role in the development of VTE. Altered blood cell counts, i.e., leukocytosis, thrombocytosis, and anemia, have been found to associate with VTE risk. Tumor-related activation of leukocytes leads to the release of tissue factor-expressing microvesicles and the formation of neutrophil extracellular traps, initiating coagulation and forming a scaffold for thrombi. Tissue factor-expressing microvesicles are also thought to be released by PDAC cells. PDAC cells have been shown to stimulate platelet activation and aggregation, proposedly via the secretion of podoplanin and mucins. Hypofibrinolysis, partially explained by increased plasminogen activator inhibitor-1 activity, is observed in PDAC. In short, PDAC-associated hypercoagulability is a complex and multifactorial process. A better understanding of cellular contributions to hypercoagulability might lead to the improvement of diagnostic tests to identify PDAC patients at highest risk of VTE.
Collapse
Affiliation(s)
- Ruth Anne Laura Willems
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, The Netherlands
- Thrombosis Expert Center Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM, School for Cardiovascular Diseases, Maastricht, The Netherlands
| | - Charlotte Biesmans
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, The Netherlands
- Thrombosis Expert Center Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Elena Campello
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Paolo Simioni
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Bas de Laat
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, The Netherlands
- CARIM, School for Cardiovascular Diseases, Maastricht, The Netherlands
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Judith de Vos-Geelen
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Mark Roest
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Hugo Ten Cate
- Thrombosis Expert Center Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM, School for Cardiovascular Diseases, Maastricht, The Netherlands
| |
Collapse
|
2
|
Qiu Y, Wang H, Guo Q, Liu Y, He Y, Zhang G, Yang C, Du Y, Gao F. CD44s-activated tPA/LRP1-NFκB pathway drives lamellipodia outgrowth in luminal-type breast cancer cells. Front Cell Dev Biol 2023; 11:1224827. [PMID: 37842093 PMCID: PMC10569302 DOI: 10.3389/fcell.2023.1224827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023] Open
Abstract
Some cancer cells migration and metastasis are characterized by the outgrowth of lamellipodia protrusions in which the underlying mechanism remains unclear. Evidence has confirmed that lamellipodia formation could be regulated by various adhesion molecules, such as CD44, and we previously reported that lamellipodia at the leading edge of luminal type breast cancer (BrCa) were enriched with high expression of CD44. In this study, we found that the overexpression of CD44s could promote lamellipodia formation in BrCa cells through inducing tissue type plasminogen activator (tPA) upregulation, which was achieved by PI3K/Akt signaling pathway activation. Moreover, we revealed that tPA could interact with LDL receptor related protein 1 (LRP1) to activate the downstream NFκB signaling pathway, which in turn facilitate lamellipodia formation. Notably, inhibition of the tPA/LRP1-NFkB signaling cascade could attenuate the CD44s-induced lamellipodia formation. Thus, our findings uncover a novel role of CD44s in driving lamellipodia outgrowth through tPA/LRP1-NFkB axis in luminal BrCa cells that may be helpful for seeking potential therapeutic targets.
Collapse
Affiliation(s)
- Yaqi Qiu
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Guo
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoliang Zhang
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Du
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Gao
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Mehner C, Hockla A, Coban M, Madden B, Estrada R, Radisky DC, Radisky ES. Activity-based protein profiling reveals active serine proteases that drive malignancy of human ovarian clear cell carcinoma. J Biol Chem 2022; 298:102146. [PMID: 35716777 PMCID: PMC9304776 DOI: 10.1016/j.jbc.2022.102146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is an understudied poor prognosis subtype of ovarian cancer lacking in effective targeted therapies. Efforts to define molecular drivers of OCCC malignancy may lead to new therapeutic targets and approaches. Among potential targets are secreted proteases, enzymes which in many cancers serve as key drivers of malignant progression. Here, we found that inhibitors of trypsin-like serine proteases suppressed malignant phenotypes of OCCC cell lines. To identify the proteases responsible for malignancy in OCCC, we employed activity-based protein profiling to directly analyze enzyme activity. We developed an activity-based probe featuring an arginine diphenylphosphonate warhead to detect active serine proteases of trypsin-like specificity and a biotin handle to facilitate affinity purification of labeled proteases. Using this probe, we identified active trypsin-like serine proteases within the complex proteomes secreted by OCCC cell lines, including two proteases in common, tissue plasminogen activator and urokinase-type plasminogen activator. Further interrogation of these proteases showed that both were involved in cancer cell invasion and proliferation of OCCC cells and were also detected in in vivo models of OCCC. We conclude the detection of tissue plasminogen activator and urokinase-type plasminogen activator as catalytically active proteases and significant drivers of the malignant phenotype may point to these enzymes as targets for new therapeutic strategies in OCCC. Our activity-based probe and profiling methodology will also serve as a valuable tool for detection of active trypsin-like serine proteases in models of other cancers and other diseases.
Collapse
Affiliation(s)
- Christine Mehner
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA,Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Mathew Coban
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Benjamin Madden
- Medical Genome Facility Proteomics Core, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Derek C. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA,For correspondence: Evette S. Radisky
| |
Collapse
|
4
|
Al‐Qahtani SM, Gadalla SE, Guo M, Ericsson C, Hägerstrand D, Nistér M. The association between Annexin A2 and epithelial cell adhesion molecule in breast cancer cells. Cancer Rep (Hoboken) 2022; 5:e1498. [PMID: 34240826 PMCID: PMC9124509 DOI: 10.1002/cnr2.1498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/05/2021] [Accepted: 06/14/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The epithelial cell adhesion molecule (EpCAM) is a type I transmembrane and glycosylated protein, which is overexpressed in many neoplasms. However, EpCAM has no known ligand partners and the mechanisms by which it functions are not fully understood. AIM This study was performed to discover novel partners of EpCAM, which may provide a better understanding of its functions. METHODS The membrane fraction of the ERα+ noninvasive breast cancer cell line ZR-75-1 and MCF-7 was extracted and followed by co-immunoprecipitation of EpCAM using C-10, a mouse monoclonal antibody raised against amino acids 24-93 of the EpCAM molecule. As a negative control, MDA-MB-231 and Hs578T were used since they express a negligible amount of EpCAM and are known as EpCAM-/low ERα-/low invasive and tumorigenic breast cancer cell lines. RESULTS Annexin A2 (ANXA2) was found to be selectively and differentially co-immunoprecipitated with EpCAM in the ERα+ breast cancer cells MCF-7 and ZR-75-1. ANXA2 is a multifunctional protein and known to act as a co-receptor for tissue plasminogen activator (tPA) on the surface of endothelial and cancer cells, thereby affecting fibrinolytic activity and neoangiogenesis as well as invasive and metastatic properties. In this study, the association between EpCAM and ANXA2 was found to affect the activity of tPA. CONCLUSION This study concludes that ANXA2 co-localizes with EpCAM at the plasma membrane, and the co-localization may have functional implications. Data suggest that EpCAM supports ANXA2 to function as a co-receptor for the tPA, and that EpCAM has a regulatory function on the expression and subcellular localization of ANXA2.
Collapse
Affiliation(s)
- Saad Misfer Al‐Qahtani
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
- Department of Pathology, College of Medicine and Najran University HospitalNajran UniversityNajranSaudi Arabia
| | | | - Min Guo
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| | | | | | - Monica Nistér
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
5
|
Wang X, Xue D, Zhu X, Geng R, Bao X, Chen X, Xia T. Low Expression of PLAT in Breast Cancer Infers Poor Prognosis and High Immune Infiltrating Level. Int J Gen Med 2021; 14:10213-10224. [PMID: 35221711 PMCID: PMC8867000 DOI: 10.2147/ijgm.s341959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Breast cancer accounts for the highest incidence of tumors in women. Immune infiltrating of the tumor microenvironment positively correlates with the overall survival of breast cancer patients. PLAT can affect the development of many cancers, but its mechanism in breast cancer is unclear. We assessed the correlation between PLAT and immune infiltrating in breast cancer based on the TCGA database. Patients and Methods The expression and DNA methylation of PLAT in breast cancer with different clinical characteristics was tested by Wilcoxon signed rank test and displayed by box plot. Sequentially, Kaplan–Meier plot was employed to compare the difference in overall survival rates between patients with different expressed levels. Univariate and multivariate Cox regression analyses were used to validate whether PLAT is an independent prognostic factor of breast cancer. After that, GO, KEGG, and gene-set enrichment analysis were employed to do functional enrichment analysis. Finally, TIMER, TISIDB database, and ssGSEA algorithm were used to assess the correlation between PLAT expression and various immune characteristics. The correlation between PLAT expression and DNA methylation was examined by Pearson correlation coefficient. Results PLAT displays differential expression levels in breast cancer patients with various clinical characteristics. As an independent protective factor for breast cancer, PLAT may significantly correlate with the immune status of breast cancer by adjusting many immune molecules and affecting the immune infiltration in the tumor microenvironment. DNA methylation of PLAT downregulates the gene expression and affects the prognosis of breast cancer. Conclusion PLAT can be considered a potential biomarker to predict breast cancer prognosis and might contribute to the development of immunological treatment strategies.
Collapse
Affiliation(s)
- Xinyang Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, People’s Republic of China
| | - Dandan Xue
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xiaoxia Zhu
- Department of Thyroid and Breast, Yixing People’s Hospital Affiliated to Jiangsu University, Yixing, People’s Republic of China
| | - Rui Geng
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xin Bao
- Department of Thyroid and Breast, Yixing People’s Hospital Affiliated to Jiangsu University, Yixing, People’s Republic of China
| | - Xiang Chen
- Department of Thyroid and Breast, Yixing People’s Hospital Affiliated to Jiangsu University, Yixing, People’s Republic of China
- Correspondence: Xiang Chen Department of Thyroid and Breast, Yixing People’s Hospital Affiliated to Jiangsu University, 75 Zhen’guan Road, Yixing, 214200, People’s Republic of China Email
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, People’s Republic of China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Tiansong Xia Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People’s Republic of China Email
| |
Collapse
|
6
|
Lin YS, Kuo TT, Lo CC, Cheng WC, Chang WC, Tseng GC, Bai ST, Huang YK, Hsieh CY, Hsu HS, Jiang YF, Lin CY, Lai LC, Li XG, Sher YP. ADAM9 functions as a transcriptional regulator to drive angiogenesis in esophageal squamous cell carcinoma. Int J Biol Sci 2021; 17:3898-3910. [PMID: 34671207 PMCID: PMC8495400 DOI: 10.7150/ijbs.65488] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/29/2021] [Indexed: 11/25/2022] Open
Abstract
Hypoxia and angiogenesis play key roles in the pathogenesis of esophageal squamous cell carcinoma (ESCC), but regulators linking these two pathways to drive tumor progression remain elusive. Here we provide evidence of ADAM9's novel function in ESCC progression. Increasing expression of ADAM9 was correlated with poor clinical outcomes in ESCC patients. Suppression of ADAM9 function diminished ESCC cell migration and in vivo metastasis in ESCC xenograft mouse models. Using cellular fractionation and imaging, we found a fraction of ADAM9 was present in the nucleus and was uniquely associated with gene loci known to be linked to the angiogenesis pathway demonstrated by genome-wide ChIP-seq. Mechanistically, nuclear ADAM9, triggered by hypoxia-induced translocation, functions as a transcriptional repressor by binding to promoters of genes involved in the negative regulation of angiogenesis, and thereby promotes tumor angiogenesis in plasminogen/plasmin pathway. Moreover, ADAM9 suppresses plasminogen activator inhibitor-1 gene transcription by interacting with its transcription factors at the promoter. Our findings uncover a novel regulatory mechanism of ADAM9 as a transcriptional regulator in angiogenesis and highlight ADAM9 as a promising therapeutic target for ESCC treatment.
Collapse
Affiliation(s)
- Yu-Sen Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung 404, Taiwan.,Division of Thoracic Surgery, China Medical University Hospital, Taichung 404, Taiwan
| | - Ting-Ting Kuo
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Chia-Chien Lo
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Guan-Chin Tseng
- Department of Anatomic Pathology, Nantou Hospital of the Ministry of Health and Welfare, Nantou 540, Taiwan
| | - Shih-Ting Bai
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Kai Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | - Chih-Ying Hsieh
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Han-Shui Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan.,Institute of Emergency and Care Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Yi-Fan Jiang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Chen-Yuan Lin
- School of Pharmacy, China Medical University, Taichung 404, Taiwan.,Division of Hematology and Oncology, China Medical University Hospital, Taichung 404, Taiwan
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Xing-Guo Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | - Yuh-Pyng Sher
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung 404, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
7
|
Fang L, Xu Q, Qian J, Zhou JY. Aberrant Factors of Fibrinolysis and Coagulation in Pancreatic Cancer. Onco Targets Ther 2021; 14:53-65. [PMID: 33442266 PMCID: PMC7797325 DOI: 10.2147/ott.s281251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Aberrant factors associated with fibrinolysis and thrombosis are found in many cancer patients, which can promote metastasis and are associated with poor prognosis. The relationship between tumor-associated fibrinolysis and thrombosis is poorly understood in pancreatic cancer. This review provides a brief highlight of existing studies that the fibrinolysis and coagulation systems were activated in pancreatic cancer patients, along with aberrant high concentrations of tissue plasminogen activator (t-PA), urine plasminogen activator (u-PA), D-dimer, fibrinogen, or platelets. These factors cooperate with each other, propelling tumor cell shedding, localization, adhesion to distant metastasis. The relationship between thrombosis or fibrinolysis and cancer immune escape is also investigated. In addition, the potential prevention and therapy strategies of pancreatic cancer targeting factors in fibrinolysis and coagulation systems are also been discussed, in which we highlight two effective agents aspirin and low-molecular weight heparin (LMWH). Summarily, this review provides new directions for the research and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Lianghua Fang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| | - Qing Xu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210029, People's Republic of China
| | - Jun Qian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| | - Jin-Yong Zhou
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| |
Collapse
|
8
|
Noh K, Bach DH, Choi HJ, Kim MS, Wu SY, Pradeep S, Ivan C, Cho MS, Bayraktar E, Rodriguez-Aguayo C, Dasari SK, Stur E, Mangala LS, Lopez-Berestein G, Sood AK. The hidden role of paxillin: localization to nucleus promotes tumor angiogenesis. Oncogene 2021; 40:384-395. [PMID: 33149280 PMCID: PMC8275353 DOI: 10.1038/s41388-020-01517-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 09/17/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022]
Abstract
Paxillin (PXN), a key component of the focal adhesion complex, has been associated with cancer progression, but the underlying mechanisms are poorly understood. The purpose of this study was to elucidate mechanisms by which PXN affects cancer growth and progression, which we addressed using cancer patient data, cell lines, and orthotopic mouse models. We demonstrated a previously unrecognized mechanism whereby nuclear PXN enhances angiogenesis by transcriptionally regulating SRC expression. SRC, in turn, increases PLAT expression through NF-ĸB activation; PLAT promotes angiogenesis via LRP1 in endothelial cells. PXN silencing in ovarian cancer mouse models reduced angiogenesis, tumor growth, and metastasis. These findings provide a new understanding of the role of PXN in regulating tumor angiogenesis and growth.
Collapse
Affiliation(s)
- Kyunghee Noh
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon, Republic of Korea
| | - Duc-Hiep Bach
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hyun-Jin Choi
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Obstetrics and Gynecology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Mark S Kim
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sherry Y Wu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Cristina Ivan
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Min-Soon Cho
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Cristian Rodriguez-Aguayo
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Santosh K Dasari
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elaine Stur
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gabriel Lopez-Berestein
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Qin YY, Huang SN, Chen G, Pang YY, Li XJ, Xing WW, Wei DM, He Y, Rong MH, Tang XZ. Clinicopathological value and underlying molecular mechanism of annexin A2 in 992 cases of thyroid carcinoma. Comput Biol Chem 2020; 86:107258. [PMID: 32304977 DOI: 10.1016/j.compbiolchem.2020.107258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/30/2019] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Thyroid carcinoma (THCA) is one of the most frequent endocrine cancers and has increasing morbidity. Annexin A2 (ANXA2) has been found to be highly expressed in various cancers; however, its expression level and potential mechanism in THCA remain unknown. This study investigated the clinicopathological value and primary molecular machinery of ANXA2 in THCA. MATERIAL AND METHODS Public RNA-sequencing and microarray data were obtained and analyzed with ANXA2 expression in THCA and corresponding non-cancerous thyroid tissue. A Pearson correlation coefficient calculation was used for the acquisition of ANXA2 coexpressed genes, while edgR, limma, and Robust Rank Aggregation were employed for differentially expressed gene (DEG) in THCA. The probable mechanism of ANXA2 in THCA was predicted by gene ontology and pathway enrichment. A dual-luciferase reporter assay was employed to confirm the targeting relationships between ANXA2 and its predicted microRNA (miRNA). RESULTS Expression of ANXA2 was significantly upregulated in THCA tissues with a summarized standardized mean difference of 1.09 (P < 0.0001) based on 992 THCA cases and 589 cases of normal thyroid tissue. Expression of ANXA2 was related to pathologic stage. Subsequently, 1442 genes were obtained when overlapping 4542 ANXA2 coexpressed genes with 2248 DEGs in THCA; these genes were mostly enriched in pathways of extracellular matrix-receptor interaction, cell adhesion molecules, and complement and coagulation cascades. MiR-23b-3p was confirmed to target ANXA2 by dual-luciferase reporter assay. CONCLUSIONS Upregulated expression of ANXA2 may promote the malignant biological behavior of THCA by affecting the involving pathways or being targeted by miR-23b-3p.
Collapse
Affiliation(s)
- Yong-Ying Qin
- Department of Head and Neck Tumor Surgery, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Su-Ning Huang
- Department of Radiotherapy, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Yu-Yan Pang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Xiao-Jiao Li
- Department of PET/CT, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Wen-Wen Xing
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Dan-Ming Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Yun He
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Min-Hua Rong
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China.
| | - Xiao-Zhun Tang
- Department of Head and Neck Tumor Surgery, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China.
| |
Collapse
|
10
|
Yan M, Wang W, Zhou J, Chang M, Peng W, Zhang G, Li J, Li H, Bai C. Knockdown of PLAT enhances the anticancer effect of gefitinib in non-small cell lung cancer. J Thorac Dis 2020; 12:712-723. [PMID: 32274137 PMCID: PMC7139041 DOI: 10.21037/jtd.2019.12.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Tyrosine kinase inhibitors (TKIs), such as gefitinib, are widely used as standard treatments for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations. However, the subsequent inevitable drug resistance has become a major challenge in clinical treatment. The aim of this study was to investigate the role of tissue-type plasminogen activator (PLAT) in gefitinib resistance in NSCLC. Methods The function of PLAT was determined using gefitinib-resistant cells and a nude mouse model. The gene knockdown was achieved by Lentivirus based RNA silence technique. Expression of relevant genes and proteins, cell viability, proliferation, apoptosis, cell cycle, reactive oxygen species levels, mitochondrial membrane potential and differential gene expression was detected by RT-qPCR, western blot, cell counting kit-8 assay, EdU incorporation, flow cytometry, JC-1 dye assay and complementary DNA arrays. The effects of PLAT knockdown on tumorigenesis was analyzed in vivo. Results Gefitinib-resistant cells expressed higher levels of PLAT and that knockdown of PLAT in resistant cells restored gefitinib sensitivity. Tumor proliferation was limited in vivo following PLAT knockdown. Moreover, PLAT knockdown affected mitochondrial function, caused caspase activation and cell cycle arrest, and activated TNF-α signaling, leading to apoptosis of gefitinib-resistant PC9 cells. Conclusions Our results suggest that PLAT reduces apoptosis of NSCLC cells and knockdown of PLAT enhances anticancer effect of gefitinib by upregulating TNF-α signaling.
Collapse
Affiliation(s)
- Mengnan Yan
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Wang
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Medical College of Qingdao University, Yantai 264000, China
| | - Jian Zhou
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Meijia Chang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenjun Peng
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ge Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huayin Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunxue Bai
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
11
|
Al-Awadhi FH, Law BK, Paul VJ, Luesch H. Grassystatins D-F, Potent Aspartic Protease Inhibitors from Marine Cyanobacteria as Potential Antimetastatic Agents Targeting Invasive Breast Cancer. JOURNAL OF NATURAL PRODUCTS 2017; 80:2969-2986. [PMID: 29087712 PMCID: PMC5764543 DOI: 10.1021/acs.jnatprod.7b00551] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Three new modified peptides named grassystatins D-F (1-3) were discovered from a marine cyanobacterium from Guam. Their structures were elucidated using NMR spectroscopy and mass spectrometry. The hallmark structural feature in the peptides is a statine unit, which contributes to their aspartic protease inhibitory activity preferentially targeting cathepsins D and E. Grassystatin F (3) was the most potent analogue, with IC50 values of 50 and 0.5 nM against cathepsins D and E, respectively. The acidic tumor microenvironment is known to increase the activation of some of the lysosomal proteases associated with tumor metastasis such as cathepsins. Because cathepsin D is a biomarker in aggressive forms of breast cancer and linked to poor prognosis, the effects of cathepsin D inhibition by 1 and 3 on the downstream cellular substrates cystatin C and PAI-1 were investigated. Furthermore, the functional relevance of targeting cathepsin D substrates was evaluated by examining the effect of 1 and 3 on the migration of MDA-MD-231 cells. Grassystatin F (3) inhibited the cleavage of cystatin C and PAI-1, the activities of their downstream targets cysteine cathepsins and tPA, and the migration of the highly aggressive triple negative breast cancer cells, phenocopying the effect of siRNA-mediated knockdown of cathepsin D.
Collapse
Affiliation(s)
- Fatma H. Al-Awadhi
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Brian K. Law
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
- Department of Pharmacology and Therapeutics, University of Florida, 1600 Archer Road, Gainesville, Florida 32610, United States
| | - Valerie J. Paul
- Smithsonian Marine Station, 701 Seaway Drive, Fort Pierce, Florida 34949, United States
| | - Hendrik Luesch
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
- Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| |
Collapse
|
12
|
Activation of tissue plasminogen activator by metastasis-inducing S100P protein. Biochem J 2017; 474:3227-3240. [PMID: 28798096 DOI: 10.1042/bcj20170578] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/25/2022]
Abstract
S100P protein in human breast cancer cells is associated with reduced patient survival and, in a model system of metastasis, it confers a metastatic phenotype upon benign mammary tumour cells. S100P protein possesses a C-terminal lysine residue. Using a multiwell in vitro assay, S100P is now shown for the first time to exhibit a strong, C-terminal lysine-dependent activation of tissue plasminogen activator (tPA), but not of urokinase-catalysed plasminogen activation. The presence of 10 μM calcium ions stimulates tPA activation of plasminogen 2-fold in an S100P-dependent manner. S100P physically interacts with both plasminogen and tPA in vitro, but not with urokinase. Cells constitutively expressing S100P exhibit detectable S100P protein on the cell surface, and S100P-containing cells show enhanced activation of plasminogen compared with S100P-negative control cells. S100P shows C-terminal lysine-dependent enhancement of cell invasion. An S100P antibody, when added to the culture medium, reduced the rate of invasion of wild-type S100P-expressing cells, but not of cells expressing mutant S100P proteins lacking the C-terminal lysine, suggesting that S100P functions outside the cell. The protease inhibitors, aprotinin or α-2-antiplasmin, reduced the invasion of S100P-expressing cells, but not of S100P-negative control cells, nor cells expressing S100P protein lacking the C-terminal lysine. It is proposed that activation of tPA via the C-terminal lysine of S100P contributes to the enhancement of cell invasion by S100P and thus potentially to its metastasis-promoting activity.
Collapse
|
13
|
Wehner C, Janjić K, Agis H. Relevance of the plasminogen system in physiology, pathology, and regeneration of oral tissues - From the perspective of dental specialties. Arch Oral Biol 2016; 74:136-145. [PMID: 27743595 DOI: 10.1016/j.archoralbio.2016.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 09/15/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022]
Abstract
Plasmin is a proteolytic enzyme that is crucial in fibrinolysis. In oral tissues, the plasminogen system plays an essential role in physiological and pathological processes, which in addition to fibrinolysis include degradation of extracellular matrix, inflammation, immune response, angiogenesis, tissue remodeling, cell migration, and wound healing. Oral tissues reveal a change in the plasminogen system during pathological processes such as periodontitis, peri-implantitis, or pulpitis, as well as in response to mechanical load. The plasminogen system is also a key element in tissue regeneration. The number of studies investigating the plasminogen system in dentistry have grown continuously in recent years, highlighting its increasing relevance in dental medicine. In this review, we present the diverse functions of the plasminogen system in physiology and its importance for dental specialists in pathology and regeneration. We thus provide an overview of the current knowledge on the role of the plasminogen system in the different fields of dentistry, including endodontics, orthodontics, periodontics, and oral surgery.
Collapse
Affiliation(s)
- Christian Wehner
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University of Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Klara Janjić
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University of Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Hermann Agis
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University of Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
14
|
Ammendola M, Sacco R, Marech I, Sammarco G, Zuccalà V, Luposella M, Patruno R, Giordano M, Ruggieri E, Zizzo N, Gadaleta CD, Ranieri G. Microvascular density and endothelial area correlate with Ki-67 proliferative index in surgically-treated pancreatic ductal adenocarcinoma patients. Oncol Lett 2015; 10:967-971. [PMID: 26622606 DOI: 10.3892/ol.2015.3286] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 04/09/2015] [Indexed: 12/14/2022] Open
Abstract
Previous experimental and clinical data have indicated that tumour cell proliferation is associated with angiogenesis; in addition, an increased microvascular density (MVD) of tumours has been associated with poor prognosis in solid and haematological malignancies. However, limited data exists regarding the association between tumour cell proliferation and angiogenesis in primary tumour tissue from pancreatic ductal adenocarcinoma (PDAC) patients; therefore, the present study aimed to investigate this association. A series of 31 PDAC patients with stage Tumour (T)2-3 Node (N)0-1 Metastasis (M)0 were recruited into the present study and subsequently underwent surgery. PDAC tissue and adjacent normal tissue (ANT), resected during surgery, were evaluated using immunohistochemistry and image analysis methods to determine MVD, endothelial area (EA) and Ki-67 expression, which is an indicator of cell proliferation rate. The results demonstrated a correlation between the above parameters with each other as well as the main clinico-pathological features of PDAC. Significant differences were identified in MVD, EA and Ki-67 proliferation index between PDAC and ANT. It was demonstrated that MVD, EA and Ki-67 proliferation index were significantly correlated with each other in tumour tissue (r=0.69-0.81; P=0.001-0.003). However, no other significant correlations were identified. These data therefore suggested that angiogenesis and cell proliferation rate were significantly increased in PDAC compared with ANT, which provides a biological basis for the potential use of novel combinations of angiogenesis inhibitors and anti-proliferative chemotherapeutic drugs in the treatment of PDAC.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy ; Surgery Unit, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Rosario Sacco
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Ilaria Marech
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Valeria Zuccalà
- Department of Health Science, Pathology Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Maria Luposella
- Department of Medical and Surgery Science, Cardiovascular Disease Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Rosa Patruno
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Marcella Giordano
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Eustachio Ruggieri
- Surgery Unit, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Nicola Zizzo
- Chair of Pathology, 'Aldo Moro' University of Bari, Bari 70010, Italy
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| |
Collapse
|
15
|
Xu XH, Pan W, Kang LH, Feng H, Song YQ. Association of annexin A2 with cancer development (Review). Oncol Rep 2015; 33:2121-8. [PMID: 25760910 DOI: 10.3892/or.2015.3837] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/20/2015] [Indexed: 01/11/2023] Open
Abstract
Annexin A2 (ANXA2) is a well-known calcium-dependent phospholipid binding protein widely distributed in the nucleus, cytoplasm and extracellular surface of various eukaryotic cells. It has been recognized as a pleiotropic protein affecting a wide range of molecular and cellular processes. Dysregulation and abnormal expression of ANXA2 are linked to a large number of prevalent diseases, including autoimmune and neurodegenerative disease, antiphospholipid syndrome, inflammation, diabetes mellitus and a series of cancers. Accumulating data suggest that ANXA2 is aberrantly expressed in a wide spectrum of cancers, and exerts profound effects on tumor cell adhesion, proliferation, apoptosis, invasion and metastasis as well as tumor neovascularization via different modes of action. However, despite significant research, our knowledge of the mechanism by which ANXA2 participates in cancer development remains fragmented. The present review systematically summarizes the effects of ANXA2 on tumor progression, in an attempt to gain an improved understanding of the underlying mechanisms and to provide a potential effective target for cancer therapy.
Collapse
Affiliation(s)
- Xiao-Heng Xu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Li-Hua Kang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Hui Feng
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Yan-Qiu Song
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|
16
|
Kruithof EKO, Dunoyer-Geindre S. Human tissue-type plasminogen activator. Thromb Haemost 2014; 112:243-54. [PMID: 24718307 DOI: 10.1160/th13-06-0517] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 03/07/2014] [Indexed: 11/05/2022]
Abstract
Tissue-type plasminogen activator (t-PA ) plays an important role in the removal of intravascular fibrin deposits and has several physiological roles and pathological activities in the brain. Its production by many other cell types suggests that t-PA has additional functions outside the vascular and central nervous system. Activity of t-PA is regulated at the level of its gene transcription, its mRNA stability and translation, its storage and regulated release, its interaction with cofactors that enhance its activity, its inhibition by inhibitors such as plasminogen activator inhibitor type 1 or neuroserpin, and its removal by clearance receptors. Gene transcription of t-PA is modulated by a large number of hormones, growth factors, cytokines or drugs and t-PA gene responses may be tissue-specific. The aim of this review is to summarise current knowledge on t-PA function and regulation of its pericellular activity, with an emphasis on regulation of its gene expression.
Collapse
Affiliation(s)
- E K O Kruithof
- Egbert K.O. Kruithof, Division of Angiology and Hemostasis, Department of internal medicine, University Hospital of Geneva and Faculty of Medicine of the University of Geneva, University Medical Center CMU 9094, 1 Rue Michel Servet, CH1211 Geneva 4, Switzerland, Tel.: +41 22 3795493 or +41 22 3795567, E-mail:
| | - S Dunoyer-Geindre
- Sylvie Dunoyer-Geindre, Division of Angiology and Hemostasis, Department of internal medicine, University Hospital of Geneva and Faculty of Medicine of the University of Geneva, University Medical Center CMU 9094, 1 Rue Michel Servet, CH1211 Geneva 4, Switzerland, Tel.: +41 22 3795493 or +41 22 3795567, E-mail:
| |
Collapse
|
17
|
Regulation of protein translation and c-Jun expression by prostate tumor overexpressed 1. Oncogene 2013; 33:1124-34. [PMID: 23455324 DOI: 10.1038/onc.2013.51] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 12/17/2012] [Accepted: 01/11/2013] [Indexed: 12/18/2022]
Abstract
Prostate tumor overexpressed-1 (PTOV1), a modulator of the Mediator transcriptional regulatory complex, is expressed at high levels in prostate cancer and other neoplasias in association with a more aggressive disease. Here we show that PTOV1 interacts directly with receptor of activated protein C kinase 1 (RACK1), a regulator of protein kinase C and Jun signaling and also a component of the 40S ribosome. Consistent with this interaction, PTOV1 was associated with ribosomes and its overexpression promoted global protein synthesis in prostate cancer cells and COS-7 fibroblasts in a mTORC1-dependent manner. Transfection of ectopic PTOV1 enhanced the expression of c-Jun protein without affecting the levels of c-Jun or RACK1 mRNA. Conversely, knockdown of PTOV1 caused significant declines in global protein synthesis and c-Jun protein levels. High levels of PTOV1 stimulated the motility and invasiveness of prostate cancer cells, which required c-Jun, whereas knockdown of PTOV1 strongly inhibited the tumorigenic and metastatic potentials of PC-3 prostate cancer cells. In human prostate cancer samples, the expression of high levels of PTOV1 in primary and metastatic tumors was significantly associated with increased nuclear localization of active c-Jun. These results unveil new functions of PTOV1 in the regulation of protein translation and in the progression of prostate cancer to an invasive and metastatic disease.
Collapse
|
18
|
Fernández-Miranda G, Méndez R. The CPEB-family of proteins, translational control in senescence and cancer. Ageing Res Rev 2012; 11:460-72. [PMID: 22542725 DOI: 10.1016/j.arr.2012.03.004] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/14/2012] [Accepted: 03/27/2012] [Indexed: 12/31/2022]
Abstract
Cytoplasmic elongation of the poly(A) tail was originally identified as a mechanism to activate maternal mRNAs, stored as silent transcripts with short poly(A) tails, during meiotic progression. A family of RNA-binding proteins named CPEBs, which recruit the translational repression or cytoplasmic polyadenylation machineries to their target mRNAs, directly mediates cytoplasmic polyadenylation. Recent years have witnessed an explosion of studies showing that CPEBs are not only expressed in a variety of somatic tissues, but have essential functions controlling gene expression in time and space in the adult organism. These "new" functions of the CPEBs include regulating the balance between senescence and proliferation and its pathological manifestation, tumor development. In this review, we summarize current knowledge on the functions of the CPEB-family of proteins in the regulation of cell proliferation, their target mRNAs and the mechanism controlling their activities.
Collapse
|
19
|
Bournet B, Pointreau A, Souque A, Oumouhou N, Muscari F, Lepage B, Senesse P, Barthet M, Lesavre N, Hammel P, Levy P, Ruszniewski P, Cordelier P, Buscail L. Gene expression signature of advanced pancreatic ductal adenocarcinoma using low density array on endoscopic ultrasound-guided fine needle aspiration samples. Pancreatology 2011; 12:27-34. [PMID: 22487470 DOI: 10.1016/j.pan.2011.12.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS The purpose of this study was to investigate the clinical feasibility and utility of low-density array analysis on samples obtained from endoscopic ultrasound-guided fine needle aspiration biopsy in locally advanced and/or metastatic pancreatic ductal adenocarcinoma and chronic pancreatitis. PATIENTS AND METHODS In this prospective multicenter study, we quantified candidate gene expression in biopsies sampled from 44 locally advanced and/or metastatic pancreatic carcinoma and from 17 pseudotumoural chronic pancreatitis using dedicated low-density array microfluidic plates. RESULTS We first demonstrated that 18S gene expression is stable and comparable in normal pancreas and pancreatic cancer tissues. Next, we found that eight genes (S100P, PLAT, PLAU, MSLN, MMP-11, MMP-7, KRT7, KRT17) were significantly over expressed in pancreatic cancer samples when compared to pseudotumoural chronic pancreatitis (p value ranging from 0.0007 to 0.0215): Linear discriminative analysis identified S100P, PLAT, MSLN, MMP-7, KRT7 as highly explicative variables. The area under receiver operating curve establishes the clinical validity of the potential diagnostic markers identified in this study (values ranging from 0.69 to 0.76). In addition, combination of S100P and KRT7 gave better diagnosis performances (Area Under Receiver Operating Curve 0.81, sensitivity 81%, specificity 77%). CONCLUSION We demonstrate that molecular studies on EUS-guided FNA material are feasible for the identification and quantification of markers in PDAC patients diagnosed with non-resectable tumours. Using low-density array, we isolated a molecular signature of advanced pancreatic carcinoma including mostly cancer invasion-related genes. This work stems for the use of novel biomarkers for the molecular diagnosis of patient with solid pancreatic masses.
Collapse
Affiliation(s)
- B Bournet
- INSERM UMR1037, Cancer Research Center of Toulouse, CHU Rangueil, 1 avenue Jean Poulhès, Bât. L3, BP 84225, 31432 Toulouse Cedex 4, France3
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ong PS, Chan SY, Ho PC. Microarray analysis revealed dysregulation of multiple genes associated with chemoresistance to As(2)O(3) and increased tumor aggressiveness in a newly established arsenic-resistant ovarian cancer cell line, OVCAR-3/AsR. Eur J Pharm Sci 2011; 45:367-78. [PMID: 22178533 DOI: 10.1016/j.ejps.2011.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 11/30/2011] [Accepted: 12/03/2011] [Indexed: 01/07/2023]
Abstract
The potential of arsenic trioxide (As(2)O(3)) for use as a novel therapy for ovarian cancer treatment has been increasingly recognized. In this study, we developed an arsenic-resistant OVCAR-3 subline (OVCAR-3/AsR) and aimed to identify the molecular mechanisms and signaling pathways contributing to the development of acquired arsenic chemoresistance in ovarian cancer. OVCAR-3/AsR cells were obtained following continual exposure of parental OVCAR-3 cells to low dose As(2)O(3) for 12months. Cytotoxicity of OVCAR-3/AsR cells to As(2)O(3), paclitaxel and cisplatin was investigated. Cell apoptosis and cell cycle distribution following As(2)O(3) treatment of OVCAR-3/AsR cells was also analyzed using flow cytometry. Subsequently, cDNA microarray analysis was performed from the RNA samples of OVCAR-3 and OVCAR-3/AsR cells in duplicate experiments. Microarray data were analyzed using Genespring® and Pathway Studio® Softwares. OVCAR-3/AsR cells showed 9-fold greater resistance to As(2)O(3) and lack of collateral resistance to cisplatin and paclitaxel. Compared with parental OVCAR-3 cells, OVCAR-3/AsR had significantly lower apoptotic rates following As(2)O(3) treatment. These cells were also arrested at both the S phase and G(2)/M phase of the cell cycle after exposure to high concentrations of As(2)O(3). Gene expression profiling revealed significant differences in expression levels of 397 genes between OVCAR-3/AsR and OVCAR-3 cells. The differentially regulated transcripts genes have functional ontologies related to continued cancer cell growth, cell survival, tumor metastasis and tumor aggressiveness. Additionally, numerous gene targets of the nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor showed elevated expression in OVCAR-3/AsR cells. Subsequent pathway analysis further revealed a gene network involving interleukin 1-alpha (IL1A) in mediating the arsenic-resistant phenotype. These results showed that changes in multiple genes and an increased in tumor aggressiveness occurred during the development of acquired chemoresistance to As(2)O(3) in ovarian cancer cells. The functional relevance of these genetic changes should be validated in future studies.
Collapse
Affiliation(s)
- Pei-Shi Ong
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117 543, Singapore.
| | | | | |
Collapse
|
21
|
Ortiz-Zapater E, Pineda D, Martínez-Bosch N, Fernández-Miranda G, Iglesias M, Alameda F, Moreno M, Eliscovich C, Eyras E, Real FX, Méndez R, Navarro P. Key contribution of CPEB4-mediated translational control to cancer progression. Nat Med 2011; 18:83-90. [PMID: 22138752 DOI: 10.1038/nm.2540] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 09/28/2011] [Indexed: 02/06/2023]
Abstract
Malignant transformation, invasion and angiogenesis rely on the coordinated reprogramming of gene expression in the cells from which the tumor originated. Although deregulated gene expression has been extensively studied at genomic and epigenetic scales, the contribution of the regulation of mRNA-specific translation to this reprogramming is not well understood. Here we show that cytoplasmic polyadenylation element binding protein 4 (CPEB4), an RNA binding protein that mediates meiotic mRNA cytoplasmic polyadenylation and translation, is overexpressed in pancreatic ductal adenocarcinomas and glioblastomas, where it supports tumor growth, vascularization and invasion. We also show that, in pancreatic tumors, the pro-oncogenic functions of CPEB4 originate in the translational activation of mRNAs that are silenced in normal tissue, including the mRNA of tissue plasminogen activator, a key contributor to pancreatic ductal adenocarcinoma malignancy. Taken together, our results document a key role for post-transcriptional gene regulation in tumor development and describe a detailed mechanism for gene expression reprogramming underlying malignant tumor progression.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Cancer Research Programme, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sharma M, Blackman MR, Sharma MC. Antibody-directed neutralization of annexin II (ANX II) inhibits neoangiogenesis and human breast tumor growth in a xenograft model. Exp Mol Pathol 2011; 92:175-84. [PMID: 22044461 DOI: 10.1016/j.yexmp.2011.10.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 10/08/2011] [Indexed: 12/11/2022]
Abstract
Activation of the fibrinolytic pathway has long been associated with human breast cancer. Plasmin is the major end product of the fibrinolytic pathway and is critical for normal physiological functions. The mechanism by which plasmin is generated in breast cancer is not yet fully described. We previously identified annexin II (ANX II), a fibrinolytic receptor, in human breast tumor tissue samples and observed a strong positive correlation with advanced stage cancer (Sharma et al., 2006a). We further demonstrated that tissue plasminogen activator (tPA) binds to ANX II in invasive breast cancer MDA-MB231cells, which leads to plasmin generation (Sharma et al., 2010). We hypothesize that ANX II-dependent plasmin generation in breast tumor is necessary to trigger the switch to neoangiogenesis, thereby stimulating a more aggressive cancer phenotype. Our immunohistochemical studies of human breast tumor tissues provide compelling evidence of a strong positive correlation between ANX II expression and neoangiogenesis, and suggest that ANX II is a potential target to slow or inhibit breast tumor growth by inhibiting neoangiogenesis. We now report that administration of anti-ANX II antibody potently inhibits the growth of human breast tumor in a xenograft model. Inhibition of tumor growth is at least partly due to attenuation of neoangiogenic activity within the tumor. In vitro studies demonstrate that anti-ANX II antibody inhibits angiogenesis on three dimensional matrigel cultures by eliciting endothelial cell (EC) death likely due to apoptosis. Taken together, these data suggest that selective disruption of the fibrinolytic activity of ANX II may provide a novel strategy for specific inhibition of neoangiogenesis in human breast cancer.
Collapse
Affiliation(s)
- Meena Sharma
- University of Pennsylvania, School of Medicine, PA, USA
| | | | | |
Collapse
|
23
|
Abstract
The dismal prognosis of pancreatic adenocarcinoma is due in part to a lack of molecular information regarding disease development. Established cell lines remain a useful tool for investigating these molecular events. Here we present a review of available information on commonly used pancreatic adenocarcinoma cell lines as a resource to help investigators select the cell lines most appropriate for their particular research needs. Information on clinical history; in vitro and in vivo growth characteristics; phenotypic characteristics, such as adhesion, invasion, migration, and tumorigenesis; and genotypic status of commonly altered genes (KRAS, p53, p16, and SMAD4) was evaluated. Identification of both consensus and discrepant information in the literature suggests careful evaluation before selection of cell lines and attention be given to cell line authentication.
Collapse
|
24
|
Begum FD, Høgdall E, Christensen IJ, Kjaer SK, Blaakaer J, Christensen L, Høgdall C. Serum tetranectin is a significant prognostic marker in ovarian cancer patients. Acta Obstet Gynecol Scand 2010; 89:190-8. [PMID: 20121334 DOI: 10.3109/00016340903530936] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To evaluate the prognostic value of preoperative serum tetranectin (TN) in Danish ovarian cancer (OvCa) patients. Design. Population-based, multidisciplinary Danish case-control study of OvCa. PARTICIPANTS A total of 445 primary OvCa patients diagnosed at one of the gynecological departments in 18 regional hospitals around Denmark during the period 1994-1999. METHODS Serum levels of TN were evaluated preoperatively and tested for possible association with prognosis. MAIN OUTCOME MEASURES Disease specific survival. RESULTS During the observation period (median 45.9 months, range 0.2-121) 278 OvCa-related deaths were seen. Univariate analysis of TN and CA125 demonstrated a significant association with survival using the Cox proportional hazards model, when stratified for adjuvant treatment (TN: p < 0.0001, hazard ratio = 0.44; 95% confidence interval 0.33-0.60 and CA125: p < 0.0001, hazard ratio = 1.19; 95% confidence interval 1.11-1.27). Disease specific survival curves for patients with tumors in the early stages showed no significant association with survival, neither for TN (p = 0.68) nor for CA125 (p = 0.07). For the stage III group, a significant association with survival was found for TN (p = 0.027), but not for CA125 (p = 0.37). Multivariate Cox analysis identified TN, age, residual tumor, International Federation of Gynecology and Obstetrics stage and grade but not serum CA125 as independent prognostic variables. CONCLUSION Preoperative serum TN is a useful prognostic indicator of advanced stage for patients with OvCa.
Collapse
Affiliation(s)
- Farah Diba Begum
- The Gynecologic Clinic, The Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
25
|
Sharma M, Ownbey RT, Sharma MC. Breast cancer cell surface annexin II induces cell migration and neoangiogenesis via tPA dependent plasmin generation. Exp Mol Pathol 2010; 88:278-86. [PMID: 20079732 DOI: 10.1016/j.yexmp.2010.01.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 01/05/2010] [Indexed: 01/14/2023]
Abstract
Annexin II, an abundant phospholipids binding cell surface protein, binds tPA and functions as a regulator of fibrinolysis. Annexin II also mediates angiogenesis and enhances tumor growth and metastasis. However, the mechanism supporting this role is not known. Using human breast cancer model we show that invasive human breast cancer cells (MDA-MB231) synthesize annexin II and tissue plasminogen activator (tPA). In vitro both annexin II and tPA interacts which in turn convert zymogen plasminogen to reactive enzyme plasmin. Cell surface produced plasmin inhibited the migration of MDA-MB231 cells. Silencing of annexin II gene in MDA-MB231 cells abolished tPA binding therefore inhibited tPA dependent plasmin generation. These annexin II suppressed MDA-MB231 cells showed reduced motility. Immunohistochemical analysis of prediagnosed clinical specimens showed abundant secretion of tPA and expression of annexin II on the surface of invasive human breast cancer cells which correlates with neovascularization of the tumor. Taken together, these data indicate that annexin II may regulate localized plasmin generation in breast cancer. This may be an early event switching breast cancer from the prevascular phase to the vascular phase and thus contributing to aggressive cancer with the possibility of metastasis. The data provide a mechanism explaining the role of annexin II in breast cancer progression and suggest that annexin II may be an attractive target for therapeutic strategies aimed to inhibit angiogenesis and breast cancer.
Collapse
Affiliation(s)
- Meena Sharma
- University of Pennsylvania, Philadelphia, PA 19102, USA.
| | | | | |
Collapse
|
26
|
Feng H, Li X, Niu D, Chen WN. Protein profile in HBx transfected cells: a comparative iTRAQ-coupled 2D LC-MS/MS analysis. J Proteomics 2009; 73:1421-32. [PMID: 20026004 DOI: 10.1016/j.jprot.2009.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 12/04/2009] [Accepted: 12/07/2009] [Indexed: 01/19/2023]
Abstract
The x protein of HBV (HBx) has been involved in the development of hepatocellular carcinoma (HCC), with a possible link to individual genotypes. Nevertheless, the underlying mechanism remains obscure. In this study, we aim to identify the HBx-induced protein profile in HepG2 cells by LC-MS/MS proteomics analysis. Our results indicated that proteins were differentially expressed in HepG2 cells transfected by HBx of various genotypes. Proteins associated with cytoskeleton were found to be either up-regulated (MACF1, HMGB1, Annexin A2) or down-regulated (Lamin A/C). These may in turn result in the decrease of focal adhesion and increase of cell migration in response to HBx. Levels of other cellular proteins with reported impact on the function of extracellular matrix (ECM) proteins and cell migration, including Ca(2+)-binding proteins (S100A11, S100A6, and S100A4) and proteasome protein (PSMA3), were affected by HBx. The differential protein profile identified in this study was also supported by our functional assay which indicated that cell migration was enhanced by HBx. Our preliminary study provided a new platform to establish a comprehensive cellular protein profile by LC-MS/MS proteomics analysis. Further downstream functional assays, including our reported cell migration assay, should provide new insights in the association between HCC and HBx.
Collapse
Affiliation(s)
- Huixing Feng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | | | | | | |
Collapse
|
27
|
Roda O, Ortiz-Zapater E, Martínez-Bosch N, Gutiérrez-Gallego R, Vila-Perelló M, Ampurdanés C, Gabius HJ, André S, Andreu D, Real FX, Navarro P. Galectin-1 is a novel functional receptor for tissue plasminogen activator in pancreatic cancer. Gastroenterology 2009; 136:1379-90, e1-5. [PMID: 19171142 DOI: 10.1053/j.gastro.2008.12.039] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 09/19/2008] [Accepted: 12/11/2008] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Tissue plasminogen activator (tPA) exerts many different functions in addition to its role in fibrinolysis. In pancreatic ductal adenocarcinoma (PDA), tPA is overexpressed and plays an important role in proliferation, invasion, and angiogenesis. tPA interaction with cell membrane receptors has been related to increased proteolytic activity and to signal transduction through nonenzymatic mechanisms. The aim was to analyze the role of galectin-1 (Gal-1), an endogenous lectin that also is overexpressed in PDA, as a new functional receptor for tPA. METHODS Gal-1/tPA interaction was analyzed using surface plasmon resonance and pull-down assays. Pancreatic cells and tumors were used to study Gal-1 expression and localization by Western blot and immunostaining. Down-regulation of Gal-1 by small interference RNA was used to analyze the involvement of Gal-1/tPA interaction in extracellular signal-regulated kinase 1/2 activation, cell proliferation, and invasion in pancreatic and fibroblastic cells. RESULTS Gal-1/tPA interaction is direct, specific, and of high affinity. Gal-1 moderately increases the catalytic activity of tPA. High Gal-1 levels were detected in PDA cells in culture, where it concentrates at the migration front, and in tissues, where it is expressed in epithelial cells and in the stroma. Down-regulation of Gal-1 abolished the effects of tPA on extracellular signal-regulated kinase 1/2 activation, cell proliferation, and invasion, both in pancreatic and in tumor-derived fibroblasts. CONCLUSIONS These findings support a new molecular mechanism by which Gal-1 interaction with tPA contributes to PDA progression involving both transformed epithelial cells and tumor fibroblasts.
Collapse
Affiliation(s)
- Oriol Roda
- Cancer Research Programme, Municipal Institute of Medical Research, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Takano S, Togawa A, Yoshitomi H, Shida T, Kimura F, Shimizu H, Yoshidome H, Ohtsuka M, Kato A, Tomonaga T, Nomura F, Miyazaki M. Annexin II overexpression predicts rapid recurrence after surgery in pancreatic cancer patients undergoing gemcitabine-adjuvant chemotherapy. Ann Surg Oncol 2008; 15:3157-68. [PMID: 18712570 DOI: 10.1245/s10434-008-0061-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Revised: 06/13/2008] [Accepted: 06/15/2008] [Indexed: 01/16/2023]
Abstract
BACKGROUND Gemcitabine has been shown to exhibit significant clinical activity against pancreatic cancer and has become a first-line chemotherapeutic for this disease in recent years. However, there are still many patients who do not respond to this treatment and it is expected to improve the clinical outcome if we can develop a method to predict the efficacy of gemcitabine before treatment. The purpose of this study was to determine novel factors that make pancreatic cancer resistant to gemcitabine. MATERIALS AND METHODS Using the high-resolution proteomic approach, agarose two-dimensional gel electrophoresis, we compared protein profiling of a gemcitabine-resistant pancreatic cancer cell line with its wild-type. RESULTS We identified Annexin II as an up-regulated protein in the gemcitabine-resistant pancreatic cancer cell line. Immunohistochemistry demonstrated that Annexin II was mainly expressed at the cell surface of pancreatic cancer cells. Interestingly, Annexin II overexpression in cancer cells was significantly associated with rapid recurrence after gemcitabine adjuvant chemotherapy in postoperative patients (P = .0078), and its staining was also an independent prognostic indicator of recurrence in pancreatic cancer patients who underwent adjuvant gemcitabine treatment after curative surgery on multivariate analysis (P = .0047). In addition, inhibition of Annexin II expression by siRNA in pancreatic cancer cell lines increased the cytotoxic efficacy of gemcitabine. These results indicate that Annexin II overexpression may induce gemcitabine resistance in pancreatic cancer resulting in rapid recurrence. CONCLUSIONS Analysis of Annexin II expression in cancer tissues may predict the clinical outcome of gemcitabine treatment, leading to the development of a new method for tailor-made treatment for this disease.
Collapse
Affiliation(s)
- Shigetsugu Takano
- The Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Roda O, Chiva C, Espuña G, Gabius HJ, Real FX, Navarro P, Andreu D. A proteomic approach to the identification of new tPA receptors in pancreatic cancer cells. Proteomics 2008; 6 Suppl 1:S36-41. [PMID: 16544279 DOI: 10.1002/pmic.200500376] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We have developed a strategy to identify putative tissue-type plasminogen activator (tPA)receptors present in pancreatic cancer cells by affinity capture with tPA-Sepharose followed by 2-DE and MALDI-MS PMF. Proteins pulled down from either total lysates or raft membrane fractions were characterized and compared with those from a total lysate of an endothelial cell line (HUVEC) to identify pancreas-restricted tPA receptors. A total of 31 proteins were found by this approach, including annexin A2, already described as a tPA receptor in pancreas and endothelial cells, other proteins acting as tPA receptors (i.e., enolase, cytokeratins 8 and 18) in other tissues, and additional proteins not previously identified as candidate tPA receptors. Confirmation of the results was performed for some of these proteins using immunoblotting. These studies are the basis for further functional analyses on the role of these proteins in the biological effects of tPA.
Collapse
Affiliation(s)
- Oriol Roda
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
30
|
Ortiz-Zapater E, Peiró S, Roda O, Corominas JM, Aguilar S, Ampurdanés C, Real FX, Navarro P. Tissue plasminogen activator induces pancreatic cancer cell proliferation by a non-catalytic mechanism that requires extracellular signal-regulated kinase 1/2 activation through epidermal growth factor receptor and annexin A2. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1573-84. [PMID: 17456763 PMCID: PMC1854952 DOI: 10.2353/ajpath.2007.060850] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue plasminogen activator (tPA) is overexpressed in pancreatic ductal carcinoma and is involved in tumor progression. This effect is probably mediated through the activation of angiogenesis, cell invasion, and cell proliferation. Previous studies support the notion that the effects of tPA on cell invasion require its proteolytic activity. Here, we report the molecular mechanism responsible for the proliferative effects of tPA on pancreatic tumor cells. tPA activates the extracellular signal-regulated kinase 1/2 signaling pathway in a manner that is independent of its catalytic activity. We also show that at least two membrane receptors, epidermal growth factor receptor and annexin A2, which are overexpressed in pancreatic cancer, are involved in the transduction of tPA signaling in pancreatic tumors. This observation suggests the establishment of an amplification loop in tumor cell proliferation. Double immunofluorescence experiments showed co-localization of tPA/epidermal growth factor receptor and tPA/annexin A2 in pancreas cancer cells. These results add novel insights into the non-catalytic functions of tPA in cancer and the molecular mechanisms behind the effects of this protease on cell proliferation, including a role for epidermal growth factor receptor.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Unitat de Biologia Cel.lular i Molecular, Institut Municipal d'Investigació Mèdica, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Siri S, Chen MJ, Chen TT. Inhibition of human breast cancer cell (MBA-MD-231) invasion by the Ea4-peptide of rainbow trout pro-IGF-I. J Cell Biochem 2007; 99:1363-73. [PMID: 16795042 DOI: 10.1002/jcb.21011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It was shown previously that Ea4-peptide of trout pro-IGF-I exerted mitogenic activity in non-transformed cells and inhibited colony formation in a soft agar medium of established human cancer cells. Here we report that the same peptide inhibits the invasion of human breast cancer cells (MDA-MB-231) through a matrigel membrane in a dose-dependent manner. The expression of urokinase-type plasminogen activator (uPA), tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor 1 (PAI1) genes in MDA-MB-231 cells were downregulated by treatment with rtEa4-peptide. The inhibition of expression of these genes in response to rtEa4-peptide treatment was reduced to the control level when inhibitors for c-Jun N-terminal kinase 1/2 (JNK1/2), mitogen activated protein kinase kinase 1/2 (Mek1/2), p38 mitogen activated protein kinase (p38 MAPK), phosphatidylinositol 3-kinase (PI3K), and phosphokinase C (PKC) were used. These results suggest that inhibition of invasion of MDA-MB-231 cells by rtEa4-peptide may be mediated via the suppression of uPA, tPA, and PAI1 gene activities through signal transduction pathways.
Collapse
Affiliation(s)
- Sineenat Siri
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA
| | | | | |
Collapse
|
32
|
Ratel D, Mihoubi S, Beaulieu E, Durocher Y, Rivard GE, Gingras D, Béliveau R. VEGF increases the fibrinolytic activity of endothelial cells within fibrin matrices: involvement of VEGFR-2, tissue type plasminogen activator and matrix metalloproteinases. Thromb Res 2007; 121:203-12. [PMID: 17512973 DOI: 10.1016/j.thromres.2007.03.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Revised: 03/02/2007] [Accepted: 03/28/2007] [Indexed: 11/30/2022]
Abstract
Proteolysis of fibrin matrices by endothelial cells plays essential roles in the migratory and morphogenic differentiation processes underlying angiogenesis. Using an in vitro fibrinolysis model consisting of human umbilical vein endothelial cells (HUVECs) embedded in a three dimensional fibrin matrix, we show that VEGF, an angiogenic cytokine that plays a crucial role in the onset of angiogenesis, is a potent activator of HUVEC-mediated fibrinolysis. This VEGF-dependent fibrin degradation was completely abrogated by inhibitors of either the plasminogen activator/plasmin or matrix metalloproteinases (MMP) proteolytic systems, suggesting the involvement of both classes of proteases in fibrin degradation. Accordingly, VEGF-induced fibrinolysis correlated with an increase in the expression of tPA and of some MMPs, such as MT2-MMP and was completely blocked by a neutralizing antibody against tPA. Overall, these results indicate that efficient proteolysis of three dimensional fibrin matrices during VEGF-mediated angiogenesis involves a complex interplay between the MMP and plasmin-mediated proteolytic systems.
Collapse
Affiliation(s)
- David Ratel
- Laboratoire de Médecine Moléculaire Ste-Justine-UQAM, Centre de Cancérologie Charles-Bruneau, Hôpital Ste-Justine, 3175 Chemin Côte-Ste-Catherine, Montréal, Qc, Canada H3T 1C5
| | | | | | | | | | | | | |
Collapse
|
33
|
Koide N, Yamada T, Shibata R, Mori T, Fukuma M, Yamazaki K, Aiura K, Shimazu M, Hirohashi S, Nimura Y, Sakamoto M. Establishment of perineural invasion models and analysis of gene expression revealed an invariant chain (CD74) as a possible molecule involved in perineural invasion in pancreatic cancer. Clin Cancer Res 2006; 12:2419-26. [PMID: 16638847 DOI: 10.1158/1078-0432.ccr-05-1852] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE Perineural invasion causes frequent local recurrence even after resection and a poor prognosis for pancreatic cancer. We established perineural invasion models and analyzed the molecular mechanism of perineural invasion in pancreatic cancer. EXPERIMENTAL DESIGN Seven pancreatic cancer cell lines with or without human peripheral nerves were s.c. implanted in nonobese diabetes/severe combined immunodeficient mice. We compared expression profiles among high and low perineural invasion cell lines by using an oligonucleotide microarray. We examined up-regulation of the invariant chain (CD74) in high perineural invasion cell lines in mRNA and protein levels and surgical cases immunohistochemically. RESULTS Four of seven pancreatic cancer cell lines (CaPan1, CaPan2, CFPAC, and MPanc96) showed perineural invasion to s.c. transplanted human peripheral nerves. Moreover, CaPan1 and CaPan2 (high perineural invasion group) also resulted in a high frequency of perineural invasion to mouse s.c. peripheral nerves, whereas three pancreatic cancer cell lines HPAFII, AsPC1, and Panc1 (low perineural invasion group) did not show perineural invasion to either human or mouse nerves. We identified 37 up-regulated genes and 12 down-regulated genes in the high perineural invasion group compared with the low perineural invasion group. Among them, CD74 was up-regulated in the high perineural invasion group in mRNA and protein levels. Furthermore, immunohistochemical expression of CD74 in clinical cases revealed its significant overexpression in pancreatic cancer with perineural invasion (P < 0.008). CONCLUSIONS This is the first report of perineural invasion models using human pancreatic cancer cell lines. In combination with gene expression profiling, it was indicated that CD74 could be a candidate molecule involved in perineural invasion. These models provide new approaches for study of perineural invasion in pancreatic cancer.
Collapse
MESH Headings
- Aged
- Animals
- Antigens, Differentiation, B-Lymphocyte/analysis
- Antigens, Differentiation, B-Lymphocyte/genetics
- Blotting, Western
- Cell Line, Tumor
- Cluster Analysis
- Disease Models, Animal
- Female
- Gene Expression Regulation, Neoplastic/genetics
- Histocompatibility Antigens Class II/analysis
- Histocompatibility Antigens Class II/genetics
- Humans
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Middle Aged
- Neoplasm Invasiveness
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Oligonucleotide Array Sequence Analysis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Peripheral Nerves/metabolism
- Peripheral Nerves/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Norimasa Koide
- Departments of Pathology and Surgery, School of Medicine, Keio University, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Laurell H, Bouisson M, Berthelemy P, Rochaix P, Dejean S, Besse P, Susini C, Pradayrol L, Vaysse N, Buscail L. Identification of biomarkers of human pancreatic adenocarcinomas by expression profiling and validation with gene expression analysis in endoscopic ultrasound-guided fine needle aspiration samples. World J Gastroenterol 2006; 12:3344-51. [PMID: 16733850 PMCID: PMC4087864 DOI: 10.3748/wjg.v12.i21.3344] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To compare gene expression profiles of pancreatic adenocarcinoma tissue specimens, human pancreatic and colon adenocarcinoma and leukemia cell lines and normal pancreas samples in order to distinguish differentially expressed genes and to validate the differential expression of a subset of genes by quantitative real-time RT-PCR (RT-QPCR) in endoscopic ultrasound-guided fine needle aspiration (EUS-guided FNA) specimens.
METHODS: Commercially dedicated cancer cDNA macroarrays (Atlas Human Cancer 1.2) containing 1176 genes were used. Different statistical approaches (hierarchical clustering, principal component analysis (PCA) and SAM) were used to analyze the expression data. RT-QPCR and immunohistochemical studies were used for validation of results.
RESULTS: RT-QPCR validated the increased expression of LCN2 (lipocalin 2) and for the first time PLAT (tissue-type plasminogen activator or tPA) in malignant pancreas as compared with normal pancreas. Immunohistochemical analysis confirmed the increased expression of LCN2 protein localized in epithelial cells of ducts invaded by carcinoma. The analysis of PLAT and LCN2 transcripts in 12 samples obtained through EUS-guided FNA from patients with pancreatic adenocarcinoma showed significantly increased expression levels in comparison with those found in normal tissues, indicating that a sufficient amount of high quality RNA can be obtained with this technique.
CONCLUSION: Expression profiling is a useful method to identify biomarkers and potential target genes. Molecular analysis of EUS-guided FNA samples in pancreatic cancer appears as a valuable strategy for the diagnosis of pancreatic adenocarcinomas.
Collapse
Affiliation(s)
- Henrik Laurell
- INSERM U531, IFR31, Institut Louis Bugnard, Toulouse, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
OBJECTIVES Gastroesophageal reflux disease (GERD) is an important cause of non-cardiac chest pain (NCCP), and its detection can require ambulatory pH monitoring. The purpose of this study was to determine the advantages of a wireless ambulatory pH monitoring system and 2 days of recording in diagnosing GERD in NCCP patients. METHODS Results from ambulatory pH studies using the BRAVO capsule were reviewed from 62 subjects referred for evaluation of NCCP after non-diagnostic response to proton pump inhibitor therapy. Acid exposure time (AET) and symptom-reflux association tests were calculated after 1 day of recording and compared to the final outcome from the 2-day study. RESULTS Extending the recording time increased the number of subjects having elevated AET from 16 after 1 day to 22 after 2 days of recording, a 9.7% gain in subjects (95% CI 4.6-19.6%). The number of chest pain episodes doubled from 1 to 2 days, and 4 subjects (7.3%) developed symptoms only on the second day of monitoring. Statistically significant reflux-symptom association probabilities surfaced in an additional 13 subjects (21.0%; 95% CI 12.7-32.7%) by the conclusion of the 2-day study. The effect primarily was to identify significant associations in patients with lower proportions of reflux-associated symptoms. Taken together, 19.4% (95% CI 11.5-30.9%) of the subject group gained meaningful information suggesting a reflux diagnosis by extending the pH monitoring time to 2 days. CONCLUSIONS Extending monitoring to 2 days with a wireless pH monitoring system increases the detection of GERD in a clinically significant proportion of patients with NCCP.
Collapse
Affiliation(s)
- Chandra Prakash
- Division of Gastroenterology, Washington University School of Medicine, 660 S. Euclid Avenue, CB 8124, St. Louis, MO 63110, USA
| | | |
Collapse
|
36
|
Bredenoord AJ, Weusten BLAM, Timmer R, Conchillo JM, Smout AJPM. Addition of esophageal impedance monitoring to pH monitoring increases the yield of symptom association analysis in patients off PPI therapy. Am J Gastroenterol 2006; 101:453-9. [PMID: 16464226 DOI: 10.1111/j.1572-0241.2006.00427.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The additional yield of esophageal impedance monitoring in identification of reflux as the cause of reflux symptoms is unknown. OBJECTIVES To compare the yield of symptom-reflux association analysis of combined esophageal pH-impedance data with the yield of analysis of pH data alone. METHODS In 60 patients with symptoms of heartburn and regurgitation combined, 24-h pH-impedance monitoring was performed. Acid-suppressive medication was stopped 1 wk in advance. Patients (48) with at least one symptom during the measurement period were selected for further analysis. Patients were instructed to note the time and nature of their symptoms. Eleven types of reflux episodes were defined, based on combinations of magnitude of the pH drop, nadir pH, and nature of the refluxate (gas and liquid) on impedance tracings. Symptom association analysis-symptom index, the symptom sensitivity index, and the symptom association probability (SAP)-was performed for each definition of reflux. RESULTS The proportion of patients with a positive SAP (> or =95.0%) varied between 62.5% and 77.1%, depending on the definition of reflux episodes. When both pH and impedance parameters were used to identify reflux, a higher proportion of patients had a positive SAP than with pH alone (77.1%vs 66.7%, p < 0.05). Symptom association analysis for acidic and weakly acidic reflux separately did not result in a higher yield than analysis with all reflux episodes pooled, regardless of pH. CONCLUSION In patients off proton pump inhibitor, the addition of impedance monitoring to esophageal pH monitoring leads to an increase in the proportion of patients in whom an association between reflux episodes and symptoms can be identified.
Collapse
Affiliation(s)
- Albert J Bredenoord
- Department of Gastroenterology, St. Antonius Hospital, PO Box 2500, 3430 EM Nieuwegein, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Mimeault M, Brand RE, Sasson AA, Batra SK. Recent advances on the molecular mechanisms involved in pancreatic cancer progression and therapies. Pancreas 2005; 31:301-16. [PMID: 16258363 DOI: 10.1097/01.mpa.0000175893.04660.1b] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review describes the recent advances in the molecular events involved in pancreatic cancer initiation, progression, and metastasis. Additionally, the importance of deregulated cellular signaling elements as potential targets for developing novel therapeutic strategies against incurable forms of pancreatic cancer is reported. The emphasis is on the critical functions gained by numerous growth factors and their receptors, such as epidermal growth factor receptor, hedgehog signaling, and proangiogenic agents such as vascular endothelial factor and interleukin-8 for the sustained growth, survival, and metastasis of pancreatic cancer cells. The molecular mechanisms associated with antitumoral properties and the clinical benefits of gemcitabine alone or in combination with other cytotoxic agents for the treatment of pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | | | |
Collapse
|
38
|
Wu F, Qu B, Gong YX, Wang XF, Yang CM, Wang HL. Clinical study of t-PA and u-PA expression in patients with gastrointestinal cancer. Chin J Cancer Res 2005. [DOI: 10.1007/s11670-005-0051-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
39
|
Sheikh AM, Ochi H, Masuda J. Lysophosphatidylcholine induces tPA gene expression through CRE-dependent mechanism. Biochem Biophys Res Commun 2005; 329:71-7. [PMID: 15721275 DOI: 10.1016/j.bbrc.2005.01.094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Indexed: 11/21/2022]
Abstract
Lysophosphatidylcholine (lysoPC) is implicated in the development of atherosclerosis and certain autoimmune diseases, and is reported to induce tissue-type plasminogen activator (tPA) at the protein level in endothelial cells. This study was designed to investigate the effect of lysoPC on tPA gene expression and the underlying molecular mechanisms in cultured endothelial cells. LysoPC transiently induced the mRNA expression of tPA in endothelial cells. LysoPC also induced the mRNA expression of urokinase-type plasminogen activator (uPA), uPA receptor, and plasminogen activator inhibitor-1, but the kinetics were different from that of tPA. Promoter analysis revealed that the cyclic AMP-responsive element of the tPA gene (tPACRE) is required for lysoPC-induced tPA expression. Furthermore, an electrophoresis mobility shift assay showed that lysoPC increased the binding activity of CRE binding protein to tPACRE. These results indicated that lysoPC transcriptionally upregulated the gene expression of tPA in endothelial cells, at least in part, via tPACRE activation.
Collapse
Affiliation(s)
- Abdullah Md Sheikh
- Department of Laboratory Medicine, Shimane University School of Medicine, Izumo, Japan
| | | | | |
Collapse
|
40
|
Abstract
Proteases play an important role in cancer invasion and metastasis. In this review we specifically discuss the role of these proteases in pancreatic carcinoma. Serine proteases and matrix metalloproteinases are the focus of intense research, as they appear to be related to the process of tumor progression. We discuss the diagnostic and prognostic value of these two groups of proteases. Lastly, we report on the current and recent clinical trials focusing on inhibition of these proteases.
Collapse
Affiliation(s)
- Lane C Patten
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 6550 Fannin Street, Houston, Texas 77030, USA
| | | |
Collapse
|
41
|
Aguilar S, Corominas JM, Malats N, Pereira JA, Dufresne M, Real FX, Navarro P. Tissue plasminogen activator in murine exocrine pancreas cancer: selective expression in ductal tumors and contribution to cancer progression. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1129-39. [PMID: 15466380 PMCID: PMC1618622 DOI: 10.1016/s0002-9440(10)63374-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tissue plasminogen activator (tPA) is absent from normal human pancreas and is expressed in 95% of human pancreatic adenocarcinomas. We have analyzed the expression of components of the tPA system in murine pancreatic tumors and the role of tPA in neoplastic progression. Transgenic mice expressing T antigen and c-myc under the control of the elastase promoter (Ela1-TAg and Ela1-myc, respectively) were used. tPA was undetectable in normal pancreas, acinar dysplasia, ductal complexes, and in all acinar tumors. By contrast, it was consistently detected in Ela1-myc tumors showing ductal differentiation. Crossing transgenic Ela1-myc with tPA-/- mice had no effect on the proportion of ductal tumors, indicating that tPA is not involved in the acinar-to-ductal transition. Ela1-myc:tPA-/- mice showed an increased survival in comparison to control mice. All ductal tumors, and none of the acinar tumors, overexpressed the tPA receptor annexin A2, suggesting its participation in the effects mediated by tPA. Our findings indicate that murine and human pancreatic ductal tumors share molecular alterations in the tPA system that may play a role in tumor progression.
Collapse
Affiliation(s)
- Susana Aguilar
- Unitat de Biologia Cellular i Molecular, Institut Municipal d'Investigació Mèdica, Dr. Aiguader, 80, 08003-Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
42
|
Boucher MJ, Jean D, Vézina A, Rivard N. Dual role of MEK/ERK signaling in senescence and transformation of intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2004; 286:G736-46. [PMID: 14701721 DOI: 10.1152/ajpgi.00453.2003] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The mitogen-activated protein kinase cascade operates downstream of Ras to convey cell-surface signals to the nucleus via nuclear translocation of ERK1 and ERK2. We and others have recently demonstrated that activation of ERK1/2 by growth factors is required for proliferation of intestinal epithelial crypt cells. However, it remained to be established whether ERK1/2 activation alone was sufficient to trigger intestinal epithelial cell (IEC) proliferation. To this aim, retrovirus encoding the hemagglutinin-tagged MAPK/ERK kinase (MEK)1 wild type (wtMEK), the upstream activator of ERK1/2, or a constitutively active mutant of MEK1 (MEK1-S218D/S222D; caMEK) were used to infect nonimmortalized human normal intestinal epithelial crypt cell cultures [human intestinal epithelial cells (HIEC)] and rodent immortalized intestinal crypt cells (IEC-6). Stable expression of caMEK but not wtMEK in HIEC led to the irreversible arrest of cellular proliferation (premature senescence). Concomitant with the onset of cell-cycle arrest was the induction of the cyclin-dependent kinase inhibitors p21(Cip), p53, and p16(INK4A). By contrast, overexpression of caMEK in IEC-6 cells induced growth factor relaxation for DNA synthesis, promoted morphological transformation and growth in soft agar, and did not affect expression of p21(Cip), p53, and p16(INK4A). We provided evidences that ERK1b, an alternatively spliced isoform of ERK1, is activated and may contribute to the deregulation of contact inhibition cell growth and transformation of these cells. Constitutive activation of MEK in IECs can produce either premature senescence or forced mitogenesis depending on the integrity of a senescence program controlled by the cell cycle inhibitors p53, p16(INK4A), and p21(CIP).
Collapse
Affiliation(s)
- Marie-Josée Boucher
- Département d'Anatomie et de Biologie Cellulaire, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
| | | | | | | |
Collapse
|
43
|
Sato N, Fukushima N, Matsubayashi H, Goggins M. Identification of maspin and S100P as novel hypomethylation targets in pancreatic cancer using global gene expression profiling. Oncogene 2004; 23:1531-8. [PMID: 14716296 DOI: 10.1038/sj.onc.1207269] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
DNA hypomethylation is one of the major epigenetic alterations in human cancers. We have previously shown that genes identified as hypomethylated in pancreatic cancer are expressed in pancreatic cancer cell lines, but not in normal pancreatic ductal epithelium and can be reexpressed in nonexpressing cells using 'epigenetic modifying agents' such as DNA methyltransferase inhibitors. To identify additional targets for aberrant hypomethylation in pancreatic cancer, we used oligonucleotide microarrays to screen for genes that displayed expression patterns associated with hypomethylation. This analysis identified a substantial number of candidates including previously reported hypomethylated genes. A subset of eight genes were selected for further methylation analysis, and two cancer-related genes, maspin and S100P, were found to be aberrantly hypomethylated in a large fraction of pancreatic cancer cell lines and primary pancreatic carcinomas. Combined treatment with 5-aza-2'-deoxycytidie and trichostatin A resulted in synergistic induction of maspin and S100P mRNA in MiaPaCa2 cells where both genes were methylated. Furthermore, there was an inverse correlation between methylation and mRNA expression level for maspin and S100P in a large panel of pancreatic cancer cell lines. We also found a significant difference in the methylation patterns of maspin and two previously identified hypomethylated genes (trefoil factor 2 and lipocalin 2) between pancreatic and breast cancer cell lines, suggesting cancer-type specificity for some hypomethylation patterns. Thus, our present results confirm that DNA hypomethylation is a frequent epigenetic event in pancreatic cancer, and suggest that gene expression profiling may help to identify potential targets affected by this epigenetic alteration.
Collapse
Affiliation(s)
- Norihiro Sato
- 1Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | |
Collapse
|
44
|
Yoshida D, Watanabe K, Takahashi H, Sugisaki Y, Teramoto A. Apoptotic induction by BE16627B on human malignant glioma cell lines by an anti-matrix metalloproteinase agent. Brain Tumor Pathol 2003; 20:13-9. [PMID: 14604227 DOI: 10.1007/bf02478942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We have elucidated the pharmacological action of the anti-matrix metalloproteinase inhibitor BE16627B on glioma cells. The study was limited to the noncytotoxic dose range. The aim of the study was to investigate whether the cytotoxicity of BE16627B, an anti-MMP agent, is related to apoptosis in the human malignant glioma cell lines U87MG, U251MG, and U373MG. MTT assay was performed to detect the cytotoxic dose range. Agarose gel electrophoresis was performed with purified genomic DNA following exposure to 20 to 500 microM BE16627B for 24 h, compared with 0 microM for the control group. Transmission electron microscopy (TEM) was employed to study nuclear fragmentation following exposure to 0, 20, and 500 microM of the agent for 24 h. An in situ endolabeling assay was performed to determine the index of apoptotic induction. MTT assay revealed that concentrations of 100 microM and above were cytotoxic. DNA laddering was demonstrated in agarose gel electrophoresis. TEM disclosed condensing and fragmentation of the chromatin. None of these changes were observed in the control group and the noncytotoxic dose group. The in situ endolabeling study disclosed that the apoptotic index was significantly elevated by cytotoxic doses of this agent (U373MG; control, 4.0%; 500 microM, 68.5%). These results indicated that cytotoxic concentrations of BE16627B induced apoptosis in human malignant glioma cell lines. In our previous report, this agent inhibited activity of MMP in noncytotoxic concentrations. Further study should be done to determine the pharmacological action of toxic BE16627B.
Collapse
Affiliation(s)
- Daizo Yoshida
- Department of Neurosurgery, Nippon Medical School, 1-1-5 Sendagi. Bunkyo-ku, Tokyo 113-8603, Japan
| | | | | | | | | |
Collapse
|
45
|
Lugea A, Gukovsky I, Gukovskaya AS, Pandol SJ. Nonoxidative ethanol metabolites alter extracellular matrix protein content in rat pancreas. Gastroenterology 2003; 125:1845-59. [PMID: 14724836 DOI: 10.1053/j.gastro.2003.09.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS The mechanisms involved in ethanol-induced pancreas fibrosis are poorly understood. Here we show that fatty acid ethyl esters (FAEEs), nonoxidative ethanol metabolites, increase extracellular matrix (ECM) protein levels in pancreas. METHODS Rat pancreatic acini were incubated for 1-4 hours with FAEEs or acetaldehyde. In another set of experiments, rats received an intravenous infusion of FAEEs for 6 hours. Collagens were assessed by a hydroxyproline assay. Laminin and fibronectin were analyzed by Western blotting. Gene expression of ECM proteins was measured by conventional and real-time reverse-transcription polymerase chain reaction (RT-PCR). Matrix metalloproteinase (MMP), plasmin, and urokinase-type plasminogen activator (uPA) activities were determined by zymography and fluorogenic assays. RESULTS FAEEs increased collagen, laminin, and fibronectin levels in pancreatic acini without affecting messenger RNA (mRNA) expression for these proteins. Actinomycin D, a transcriptional inhibitor, did not block the increase in ECM proteins induced by FAEEs. FAEEs reduced the activity of the serine protease, plasmin, and that of the uPA. Consistent with these results, the serine protease inhibitor aprotinin reproduced the effects of FAEEs and prevented the further increase in ECM proteins induced by FAEEs. In vivo administration of FAEEs reduced plasmin and uPA activities and increased ECM protein levels in pancreas. Acetaldehyde had minor effects on ECM protein levels and did not affect plasmin activity. CONCLUSIONS FAEEs increase ECM protein levels in pancreas. The results suggest that this effect is caused primarily by an inhibition in ECM degradation via serine proteases including the plasminogen system.
Collapse
Affiliation(s)
- Aurelia Lugea
- University of Southern California-University of California Research Center for Alcoholic Liver and Pancreatic Diseases, Veterans Affairs Greater Los Angeles Healthcare System, 90073, USA.
| | | | | | | |
Collapse
|
46
|
Nagaoka MR, Kouyoumdjian M, Borges DR. Hepatic clearance of tissue-type plasminogen activator and plasma kallikrein in experimental liver fibrosis. Liver Int 2003; 23:476-83. [PMID: 14986822 DOI: 10.1111/j.1478-3231.2003.00872.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
UNLABELLED We have previously shown that tissue-type plasminogen activator (tPA) and rat plasma kallikrein (RPK) share a common, but not unique, pathway for liver clearance. AIM To evaluate the hepatic clearance of both proteases in experimental liver fibrosis. METHODS The hepatic clearance of these proteases was studied in porcine serum-induced liver fibrosis using the isolated and perfused rat liver model. To better interpret the results, we also studied four other experimental groups: the turpentine oil-induced acute-phase response (AP group), AP group followed by GdCl3 administration (AP/Gd group), CCl4-induced cirrhosis (CCl4 group) and normal group. RESULTS The tPA clearance decreased significantly by both fibrotic and cirrhotic rat livers whereas the RPK clearance was not altered by the fibrotic rat liver. The hepatic clearance of tPA was reduced in the AP and AP/Gd groups; on the other hand, RPK clearance was increased in the AP group and, interestingly, this effect was neutralized by concomitant GdCl3 administration. CONCLUSIONS We observed that tPA and RPK clearances were affected differently by fibrosis as well as by different stimuli of the acute-phase response, despite the fact that they share a common hepatic clearance mechanism in normal livers, and they were equally affected in cirrhosis.
Collapse
Affiliation(s)
- Marcia R Nagaoka
- Laboratory of Experimental Hepatology, Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | | | | |
Collapse
|
47
|
Farrow B, O'Connor KL, Hashimoto K, Iwamura T, Evers BM. Selective activation of PPARgamma inhibits pancreatic cancer invasion and decreases expression of tissue plasminogen activator. Surgery 2003; 134:206-12. [PMID: 12947319 DOI: 10.1067/msy.2003.221] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND PPARgamma is a ligand-activated transcription factor with antitumor effects; its ability to inhibit pancreatic cancer invasion is unknown. The purpose of this study was to define the inhibitory effect of PPARgamma ligands on pancreatic cancer invasion and the expression of invasion-related genes. METHODS Western blotting was used to establish expression of PPARgamma in AsPC-1 and SUIT-2 cells. AsPC-1 cells were treated with nontoxic doses of PPARgamma ligands (15d-PGJ(2), troglitazone, or rosiglitazone) and Matrigel Invasion chambers were used to assess invasion in vitro. A microarray for genes that contribute to invasion was used to investigate the antiinvasive targets of PPARgamma. Gene array results were confirmed by use of ribonuclease protection assay or Northern blotting. RESULTS Rosiglitazone and 15d-PGJ(2) decreased AsPC-1 cell invasion; GW9662, which inhibits PPARgamma, reversed this effect. The expression of tissue plasminogen activator (tPA) was decreased by rosiglitazone treatment, which was confirmed by Northern blotting. Secreted levels of tPA in AsPC-1 conditioned media were also decreased. CONCLUSIONS We demonstrate, for the first time, that secretion of the invasive factor tPA was decreased by rosiglitazone treatment in AsPC-1 cells. PPARgamma ligands inhibit pancreatic cancer cell invasion, suggesting that these agents may represent novel strategies to treat pancreatic cancer.
Collapse
Affiliation(s)
- Buckminster Farrow
- Department of Surgery, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | |
Collapse
|
48
|
Analysis of genes associated with lymphatic metastasis in pancreatic carcinoma using cDNA microarray. Chin J Cancer Res 2003. [DOI: 10.1007/s11670-003-0011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|