1
|
Wang B, Gu B, Zhang T, Li X, Wang N, Ma C, Xiang L, Wang Y, Gao L, Yu Y, Song K, He P, Wang Y, Zhu J, Chen H. Good or bad: Paradox of plasminogen activator inhibitor 1 (PAI-1) in digestive system tumors. Cancer Lett 2023; 559:216117. [PMID: 36889376 DOI: 10.1016/j.canlet.2023.216117] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
The fibrinolytic system is involved in many physiological functions, among which the important members can interact with each other, either synergistically or antagonistically to participate in the pathogenesis of many diseases. Plasminogen activator inhibitor 1 (PAI-1) acts as a crucial element of the fibrinolytic system and functions in an anti-fibrinolytic manner in the normal coagulation process. It inhibits plasminogen activator, and affects the relationship between cells and extracellular matrix. PAI-1 not only involved in blood diseases, inflammation, obesity and metabolic syndrome but also in tumor pathology. Especially PAI-1 plays a different role in different digestive tumors as an oncogene or cancer suppressor, even a dual role for the same cancer. We term this phenomenon "PAI-1 paradox". PAI-1 is acknowledged to have both uPA-dependent and -independent effects, and its different actions can result in both beneficial and adverse consequences. Therefore, this review will elaborate on PAI-1 structure, the dual value of PAI-1 in different digestive system tumors, gene polymorphisms, the uPA-dependent and -independent mechanisms of regulatory networks, and the drugs targeted by PAI-1 to deepen the comprehensive understanding of PAI-1 in digestive system tumors.
Collapse
Affiliation(s)
- Bofang Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Baohong Gu
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Tao Zhang
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xuemei Li
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Na Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Chenhui Ma
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lin Xiang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yunpeng Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lei Gao
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yang Yu
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Kewei Song
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Puyi He
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yueyan Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jingyu Zhu
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou, Gansu, China; Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Zhang T, Wang B, Su F, Gu B, Xiang L, Gao L, Zheng P, Li XM, Chen H. TCF7L2 promotes anoikis resistance and metastasis of gastric cancer by transcriptionally activating PLAUR. Int J Biol Sci 2022; 18:4560-4577. [PMID: 35864968 PMCID: PMC9295057 DOI: 10.7150/ijbs.69933] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Gastric cancer (GC) is the most common gastrointestinal malignant tumor, and distant metastasis is a critical factor in the prognosis of patients with GC. Understanding the mechanism of GC metastasis will help improve patient prognosis. Studies have confirmed that urokinase-type plasminogen activator receptor (PLAUR) promotes GC metastasis; however, its relationship with anoikis resistance and associated mechanisms remains unclear. In this study, we demonstrated that PLAUR promotes the anoikis resistance and metastasis of GC cells and identified transcription Factor 7 Like 2 (TCF7L2) as an important transcriptional regulator of PLAUR. We also revealed that TCF7L2 is highly expressed in GC and promotes the anoikis resistance and metastasis of GC cells. Moreover, we found that TCF7L2 transcription activates PLAUR. Finally, we confirmed that TCF7L2 is an independent risk factor for poor prognosis of patients with GC. Our results show that TCF7L2 and PLAUR are candidate targets for developing therapeutic strategies for GC metastasis.
Collapse
Affiliation(s)
- Tao Zhang
- Department of oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Bofang Wang
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Fei Su
- Department of oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Baohong Gu
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lin Xiang
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lei Gao
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Peng Zheng
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xue-Mei Li
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hao Chen
- The second clinical medical college of Lanzhou university, Lanzhou , Gansu, China.,Key laboratory of digestive system tumors, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Cancer center, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Zhai BT, Tian H, Sun J, Zou JB, Zhang XF, Cheng JX, Shi YJ, Fan Y, Guo DY. Urokinase-type plasminogen activator receptor (uPAR) as a therapeutic target in cancer. J Transl Med 2022; 20:135. [PMID: 35303878 PMCID: PMC8932206 DOI: 10.1186/s12967-022-03329-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/03/2022] [Indexed: 12/22/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR) is an attractive target for the treatment of cancer, because it is expressed at low levels in healthy tissues but at high levels in malignant tumours. uPAR is closely related to the invasion and metastasis of malignant tumours, plays important roles in the degradation of extracellular matrix (ECM), tumour angiogenesis, cell proliferation and apoptosis, and is associated with the multidrug resistance (MDR) of tumour cells, which has important guiding significance for the judgement of tumor malignancy and prognosis. Several uPAR-targeted antitumour therapeutic agents have been developed to suppress tumour growth, metastatic processes and drug resistance. Here, we review the recent advances in the development of uPAR-targeted antitumor therapeutic strategies, including nanoplatforms carrying therapeutic agents, photodynamic therapy (PDT)/photothermal therapy (PTT) platforms, oncolytic virotherapy, gene therapy technologies, monoclonal antibody therapy and tumour immunotherapy, to promote the translation of these therapeutic agents to clinical applications.
Collapse
Affiliation(s)
- Bing-Tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-Bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-Fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-Xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Ya-Jun Shi
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yu Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-Yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| |
Collapse
|
4
|
Experimental and Clinical Evidence Supports the Use of Urokinase Plasminogen Activation System Components as Clinically Relevant Biomarkers in Gastroesophageal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13164097. [PMID: 34439251 PMCID: PMC8393967 DOI: 10.3390/cancers13164097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Patients with gastric and oesophageal adenocarcinomas (GOCs) have short life expectancies as their tumours spread to other sites early. This is facilitated by the increased expression of the urokinase plasminogen activation system (uPAS); a feature of the majority of GOCs. There is increasing appreciation of the importance of uPAS expression in a range of cell types within the tumour microenvironment. Abundant clinical evidence indicates that altered expression of uPAS proteins is associated with worse outcomes, including time to tumour recurrence and patient survival. Emerging technologies, including liquid biopsy, suggest a role of uPAS for the detection of circulating tumour cells, which are responsible for the dissemination of cancers. We review and summarise pre-clinical and clinical data that supports the use of uPAS as a biomarker in GOC. Abstract Gastric and oesophageal cancers (GOCs) are lethal cancers which metastasise early and recur frequently, even after definitive surgery. The urokinase plasminogen activator system (uPAS) is strongly implicated in the invasion and metastasis of many aggressive tumours including GOCs. Urokinase plasminogen activator (uPA) interaction with its receptor, urokinase plasminogen activator receptor (uPAR), leads to proteolytic activation of plasminogen to plasmin, a broad-spectrum protease which enables tumour cell invasion and dissemination to distant sites. uPA, uPAR and the plasminogen activator inhibitor type 1 (PAI-1) are overexpressed in some GOCs. Accumulating evidence points to a causal role of activated receptor tyrosine kinase pathways enhancing uPAS expression in GOCs. Expression of these components are associated with poorer clinicopathological features and patient survival. Stromal cells, including tumour-associated macrophages and myofibroblasts, also express the key uPAS proteins, supporting the argument of stromal involvement in GOC progression and adverse effect on patient survival. uPAS proteins can be detected on circulating leucocytes, circulating tumour cells and within the serum; all have the potential to be developed into circulating biomarkers of GOC. Herein, we review the experimental and clinical evidence supporting uPAS expression as clinical biomarker in GOC, with the goal of developing targeted therapeutics against the uPAS.
Collapse
|
5
|
Zheng S, Zhang Z, Ding N, Sun J, Lin Y, Chen J, Zhong J, Shao L, Lin Z, Xue M. Identification of the angiogenesis related genes for predicting prognosis of patients with gastric cancer. BMC Gastroenterol 2021; 21:146. [PMID: 33794777 PMCID: PMC8017607 DOI: 10.1186/s12876-021-01734-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Angiogenesis is a key factor in promoting tumor growth, invasion and metastasis. In this study we aimed to investigate the prognostic value of angiogenesis-related genes (ARGs) in gastric cancer (GC). METHODS mRNA sequencing data with clinical information of GC were downloaded from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. The differentially expressed ARGs between normal and tumor tissues were analyzed by limma package, and then prognosis‑associated genes were screened using Cox regression analysis. Nine angiogenesis genes were identified as crucially related to the overall survival (OS) of patients through least absolute shrinkage and selection operator (LASSO) regression. The prognostic model and corresponding nomograms were establish based on 9 ARGs and verified in in both TCGA and GEO GC cohorts respectively. RESULTS Eighty-five differentially expressed ARGs and their enriched pathways were confirmed. Significant enrichment analysis revealed that ARGs-related signaling pathway genes were highly related to tumor angiogenesis development. Kaplan-Meier analysis revealed that patients in the high-risk group had worse OS rates compared with the low-risk group in training cohort and validation cohort. In addition, RS had a good prognostic effect on GC patients with different clinical features, especially those with advanced GC. Besides, the calibration curves verified fine concordance between the nomogram prediction model and actual observation. CONCLUSIONS We developed a nine gene signature related to the angiogenesis that can predict overall survival for GC. It's assumed to be a valuable prognosis model with high efficiency, providing new perspectives in targeted therapy.
Collapse
Affiliation(s)
- Sheng Zheng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Zizhen Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Ning Ding
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Jiawei Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Yifeng Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Jingyu Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Jing Zhong
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Liming Shao
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Zhenghua Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Meng Xue
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Gastroenterology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Fernandez-Botran R, Wellmann IA, Une C, Méndez-Chacón E, Hernández de Rodas E, Bhandari B, Villagrán de Tercero CI. Seroprevalence of Helicobacter pylori/CagA Antibodies in Guatemalan Gastric Cancer Patients: Association of Seropositivity with Increased Plasma Levels of Pepsinogens but not Soluble Urokinase Plasminogen Activator Receptor. Am J Trop Med Hyg 2020; 103:260-265. [PMID: 32314688 DOI: 10.4269/ajtmh.19-0934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Infection by Helicobacter pylori is a major risk factor for gastric cancer (GC), the second leading cause of cancer-related death worldwide. Although biomarkers such as pepsinogens (PGs) and soluble urokinase plasminogen activator receptor (suPAR) may have diagnostic and/or prognostic value in patients with GC, their levels may be affected by H. pylori infection. The aim of this study was to investigate the association of the presence of antibodies to H. pylori and cytotoxin-associated gene A (CagA) with plasma levels of PGs and suPAR in a cohort of Guatemalan GC patients and controls. To this end, levels of suPAR, Pepsinogens I and II (PGI and PGII), and antibodies to H. pylori and CagA toxin were determined by ELISA in plasma samples from 67 GC patients and 136 matched healthy controls. Seropositivity for CagA was significantly higher in patients with GC than in controls. Pepsinogens II and suPAR levels were higher and PGI/PGII ratios were lower in GC patients than in controls. There was a significant association of H. pylori seropositivity status with increased levels of PGII and lower PGI/PGII ratios, particularly in the control (non-GC) population. The levels of suPAR were not significantly affected by H. pylori or CagA seropositivity status. These results suggest that the seropositivity status for H. pylori and CagA need to be taken into account during the GC diagnostic process.
Collapse
Affiliation(s)
- Rafael Fernandez-Botran
- Department of Pathology & Laboratory Medicine, University of Louisville, Louisville, Kentucky
| | - Irmgardt Alicia Wellmann
- Centro de Investigaciones Biomédicas, Facultad de Ciencias Médicas, Universidad de San Carlos de Guatemala, Guatemala City, Guatemala
| | - Clas Une
- Instituto de Investigación en Salud (INISA), Universidad de Costa Rica, San José, Costa Rica
| | - Ericka Méndez-Chacón
- Instituto de Investigación en Salud (INISA), Universidad de Costa Rica, San José, Costa Rica
| | - Elisa Hernández de Rodas
- Centro de Investigaciones Biomédicas, Facultad de Ciencias Médicas, Universidad de San Carlos de Guatemala, Guatemala City, Guatemala
| | - Bikash Bhandari
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky
| | - Carmen I Villagrán de Tercero
- Centro de Investigaciones Biomédicas, Facultad de Ciencias Médicas, Universidad de San Carlos de Guatemala, Guatemala City, Guatemala
| |
Collapse
|
7
|
Molina-Castro S, Ramírez-Mayorga V, Alpízar-Alpízar W. Priming the seed: Helicobacter pylori alters epithelial cell invasiveness in early gastric carcinogenesis. World J Gastrointest Oncol 2018; 10:231-243. [PMID: 30254719 PMCID: PMC6147766 DOI: 10.4251/wjgo.v10.i9.231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 02/05/2023] Open
Abstract
Helicobacter pylori (H. pylori) infection is a well-established risk factor for the development of gastric cancer (GC), one of the most common and deadliest neoplasms worldwide. H. pylori infection induces chronic inflammation in the gastric mucosa that, in the absence of treatment, may progress through a series of steps to GC. GC is only one of several clinical outcomes associated with this bacterial infection, which may be at least partially attributed to the high genetic variability of H. pylori. The biological mechanisms underlying how and under what circumstances H. pylori alters normal physiological processes remain enigmatic. A key aspect of carcinogenesis is the acquisition of traits that equip preneoplastic cells with the ability to invade. Accumulating evidence implicates H. pylori in the manipulation of cellular and molecular programs that are crucial for conferring cells with invasive capabilities. We present here an overview of the main findings about the involvement of H. pylori in the acquisition of cell invasive behavior, specifically focusing on the epithelial-to-mesenchymal transition, changes in cell polarity, and deregulation of molecules that control extracellular matrix remodeling.
Collapse
Affiliation(s)
- Silvia Molina-Castro
- Cancer Epidemiology Research Program, Health Research Institute, University of Costa Rica, San José 2060, Costa Rica
- Clinical Department, School of Medicine, University of Costa Rica, San José 2060, Costa Rica
| | - Vanessa Ramírez-Mayorga
- Cancer Epidemiology Research Program, Health Research Institute, University of Costa Rica, San José 2060, Costa Rica
- Public Nutrition Section, School of Nutrition, University of Costa Rica, San José 2060, Costa Rica
| | - Warner Alpízar-Alpízar
- Center for Research in Microscopic Structures, University of Costa Rica, San José 2060, Costa Rica
- Department of Biochemistry, School of Medicine, University of Costa Rica, San José 2060, Costa Rica
| |
Collapse
|
8
|
Mahmood N, Mihalcioiu C, Rabbani SA. Multifaceted Role of the Urokinase-Type Plasminogen Activator (uPA) and Its Receptor (uPAR): Diagnostic, Prognostic, and Therapeutic Applications. Front Oncol 2018; 8:24. [PMID: 29484286 PMCID: PMC5816037 DOI: 10.3389/fonc.2018.00024] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/24/2018] [Indexed: 01/01/2023] Open
Abstract
The plasminogen activator (PA) system is an extracellular proteolytic enzyme system associated with various physiological and pathophysiological processes. A large body of evidence support that among the various components of the PA system, urokinase-type plasminogen activator (uPA), its receptor (uPAR), and plasminogen activator inhibitor-1 and -2 (PAI-1 and PAI-2) play a major role in tumor progression and metastasis. The binding of uPA with uPAR is instrumental for the activation of plasminogen to plasmin, which in turn initiates a series of proteolytic cascade to degrade the components of the extracellular matrix, and thereby, cause tumor cell migration from the primary site of origin to a distant secondary organ. The components of the PA system show altered expression patterns in several common malignancies, which have identified them as ideal diagnostic, prognostic, and therapeutic targets to reduce cancer-associated morbidity and mortality. This review summarizes the various components of the PA system and focuses on the role of uPA-uPAR in different biological processes especially in the context of malignancy. We also discuss the current state of knowledge of uPA-uPAR-targeted diagnostic and therapeutic strategies for various malignancies.
Collapse
Affiliation(s)
- Niaz Mahmood
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Catalin Mihalcioiu
- Department of Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Shafaat A. Rabbani
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Department of Oncology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
9
|
Brungs D, Chen J, Aghmesheh M, Vine KL, Becker TM, Carolan MG, Ranson M. The urokinase plasminogen activation system in gastroesophageal cancer: A systematic review and meta-analysis. Oncotarget 2018; 8:23099-23109. [PMID: 28416743 PMCID: PMC5410288 DOI: 10.18632/oncotarget.15485] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/07/2017] [Indexed: 12/19/2022] Open
Abstract
Background The urokinase plasminogen activation (uPA) system is a crucial pathway for tumour invasion and establishment of metastasis. Although there is good evidence that uPA system expression is a clinically relevant biomarker in some solid tumours, its role in gastroesophageal cancer is uncertain. Results We identified 22 studies encompassing 1966 patients which fulfilled the inclusion criteria. uPA, uPAR, or PAI-1 expression is significantly associated with high risk clinicopathological features. High uPA expression is associated with a shorter RFS (HR 1.90 95% 1.16–3.11, p = 0.01) and OS (HR 2.21 95% CI 1.74–2.80, p < 0.0001). High uPAR expression is associated with poorer OS (HR 2.21 95%CI 1.82–2.69, p < 0.0001). High PAI-1 expression is associated with shorter RFS (HR 1.96 96% CI 1.07–3.58, p = 0.03) and OS (HR 1.84 95%CI 1.28–2.64, p < 0.0001). There was no significant association between PAI-2 expression and OS (HR 0.97 95%CI 0.48–1.94, p < 0.92) although data was limited. Materials and Methods We undertook a systematic review evaluating expression of uPA, urokinase plasminogen activator receptor (uPAR), plasminogen activator inhibitor-1 (PAI-1/SerpinE1) and plasminogen activator inhibitor-2 (PAI-2/SerpinB2) on primary oesophageal, gastro-oesophageal junction, and gastric adenocarcinomas. We performed a meta-analysis of clinicopathological associations, overall survival (OS) and recurrence free survival (RFS). Conclusions We conclude that the uPA system is a clinically relevant biomarker in primary gastroesophageal cancer, with higher expression of uPA, uPAR and PAI-1 associated with higher risk disease and poorer prognosis. This also highlights the potential utility of the uPA system as a therapeutic target for improved treatment strategies.
Collapse
Affiliation(s)
- Daniel Brungs
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia.,School of Biological Sciences, University of Wollongong, Wollongong, Australia.,Illawarra Cancer Centre, Wollongong Hospital, Wollongong, Australia.,CONCERT-Translational Cancer Research Centre, New South Wales, Australia
| | - Julia Chen
- St George Cancer Centre, St George Hospital, Sydney, Australia
| | - Morteza Aghmesheh
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia.,Illawarra Cancer Centre, Wollongong Hospital, Wollongong, Australia.,CONCERT-Translational Cancer Research Centre, New South Wales, Australia
| | - Kara L Vine
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia.,School of Biological Sciences, University of Wollongong, Wollongong, Australia.,CONCERT-Translational Cancer Research Centre, New South Wales, Australia
| | - Therese M Becker
- CONCERT-Translational Cancer Research Centre, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool Hospital, Australia.,School of Medicine, University of Western Sydney, Liverpool, Australia.,South Western Medical School, University of New South Wales, Liverpool, Australia
| | - Martin G Carolan
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia.,Illawarra Cancer Centre, Wollongong Hospital, Wollongong, Australia.,CONCERT-Translational Cancer Research Centre, New South Wales, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia.,School of Biological Sciences, University of Wollongong, Wollongong, Australia.,CONCERT-Translational Cancer Research Centre, New South Wales, Australia
| |
Collapse
|
10
|
Kugaevskaya E, Gureeva T, Timoshenko O, Solovyeva N. The urokinase-type plasminogen activator system and its role in tumor progression. ACTA ACUST UNITED AC 2018; 64:472-486. [DOI: 10.18097/pbmc20186406472] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the multistage process of carcinogenesis, the key link in the growth and progression of the tumor is the invasion of malignant cells into normal tissue and their distribution and the degree of destruction of tissues. The most important role in the development of these processes is played by the system of urokinase-type plasminogen activator (uPA system), which consists of several components: serine proteinase – uPA, its receptor – uPAR and its two endogenous inhibitors – PAI-1 and PAI-2. The components of the uPA system are expressed by cancer cells to a greater extent than normal tissue cells. uPA converts plasminogen into broad spectrum, polyfunctional protease plasmin, which, in addition to the regulation of fibrinolysis, can hydrolyze a number of components of the connective tissue matrix (СTM), as well as activate the zymogens of secreted matrix metalloproteinases (MMР) – pro-MMР. MMРs together can hydrolyze all the main components of the СTM, and thus play a key role in the development of invasive processes, as well as to perform regulatory functions by activating and releasing from STM a number of biologically active molecules that are involved in the regulation of the main processes of carcinogenesis. The uPA system promotes tumor progression not only through the proteolytic cascade, but also through uPAR, PAI-1 and PAI-2, which are involved in both the regulation of uPA/uPAR activity and are involved in proliferation, apoptosis, chemotaxis, adhesion, migration and activation of epithelial-mesenchymal transition pathways. All of the above processes are aimed at regulating invasion, metastasis and angiogenesis. The components of the uPA system are used as prognostic and diagnostic markers of many cancers, as well as serve as targets for anticancer therapy.
Collapse
Affiliation(s)
| | - T.A. Gureeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | |
Collapse
|
11
|
Nitta H, Shimose T, Emi Y, Imamura T, Ohnishi K, Kusumoto T, Yamamoto M, Fukuzawa K, Takahashi I, Higashi H, Tsuji A, Akagi Y, Oki E, Maehara Y, Baba H. Expression of the anaphylatoxin C5a receptor in gastric cancer: implications for vascular invasion and patient outcomes. Med Oncol 2016; 33:118. [DOI: 10.1007/s12032-016-0834-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/24/2016] [Indexed: 02/06/2023]
|
12
|
Fossmark R, Rao S, Mjønes P, Munkvold B, Flatberg A, Varro A, Thommesen L, Nørsett KG. PAI-1 deficiency increases the trophic effects of hypergastrinemia in the gastric corpus mucosa. Peptides 2016; 79:83-94. [PMID: 27038741 DOI: 10.1016/j.peptides.2016.03.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/18/2016] [Accepted: 03/29/2016] [Indexed: 12/16/2022]
Abstract
The gastric hormone gastrin plays a role in organizing the gastric mucosa. Gastrin also regulates the expression of genes that have important actions in extracellular matrix modelling, including plasminogen activator inhibitor (PAI)-1 which is part of the urokinase plasminogen activator (uPA) system. The uPA system (including PAI-1) is associated with cancer progression, fibrosis and thrombosis. Its biological role in the stomach and molecular mechanisms of action are not well understood. The aim of this study was to examine the effect of PAI-1 on the trophic changes observed in gastric corpus mucosa in hypergastrinemia using PAI-1 and/or HK-ATPase beta subunit knockout (KO) mice. HK-ATPase beta subunit KO mice were used as a model of hypergastrinemia. In 12 month old female mice, intragastric acidity and plasma gastrin were measured. The stomachs were examined for macroscopic and histological changes. In mice null for both PAI-1 and HK-ATPase beta (double KO), there was exaggerated hypergastrinemia, increased stomach weight and corpus mucosal thickness, and more pronounced trophic and architectural changes in the corpus compared with HK-ATPase beta KO mice. Genome-wide microarray expression data for the gastric corpus mucosa showed a distinct gene expression profile for the HK-ATPase beta KO mice; moreover, enrichment analysis revealed changes in expression of genes regulating intracellular processes including cytoskeleton remodelling, cell adhesion, signal transduction and epithelial-to-mesenchymal transition (EMT). Genes differentially expressed in the double KO compared with HK-ATPase beta KO mice included the transcription factor Barx2 and the chromatin remodeler gene Tet2, which may be involved in both normal gastric physiology and development of gastric cancer. Based on the present data, we suggest that PAI-1 plays a role in maintaining gastric mucosal organization in hypergastrinemia.
Collapse
Affiliation(s)
- Reidar Fossmark
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway; Department of Gastroenterology and Hepatology, St. Olav's University Hospital, Trondheim, Norway.
| | - Shalini Rao
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway.
| | - Patricia Mjønes
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway; Department of Pathology, St. Olav's University Hospital, Trondheim, Norway.
| | - Bjørn Munkvold
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway.
| | - Arnar Flatberg
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway.
| | - Andrea Varro
- Department of Cell and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.
| | - Liv Thommesen
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway.
| | - Kristin G Nørsett
- Department of Cancer Research and Molecular Medicine, NTNU, Trondheim, Norway; The Central Norway Regional Health Authority, Trondheim, Norway.
| |
Collapse
|
13
|
Hale MD, Gotoda T, Hayden JD, Grabsch HI. Endoscopic biopsies from gastrointestinal carcinomas and their suitability for molecular analysis: a review of the literature and recommendations for clinical practice and research. Histopathology 2015; 67:147-57. [PMID: 25431371 DOI: 10.1111/his.12626] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Takuji Gotoda
- Department of Gastroenterology and Hepatology; Tokyo Medical University; Tokyo Japan
| | - Jeremy David Hayden
- Department of Upper Gastrointestinal Surgery; St James's Institute of Oncology; Leeds Teaching Hospitals NHS Trust; Leeds UK
| | - Heike Irmgard Grabsch
- Leeds Institute of Cancer and Pathology; University of Leeds; Leeds UK
- Department of Pathology; Maastricht University Medical Center; Maastricht The Netherlands
| |
Collapse
|
14
|
From plasminogen to plasmin: role of plasminogen receptors in human cancer. Int J Mol Sci 2014; 15:21229-52. [PMID: 25407528 PMCID: PMC4264222 DOI: 10.3390/ijms151121229] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/04/2014] [Accepted: 11/12/2014] [Indexed: 12/02/2022] Open
Abstract
Cell surface-associated proteolysis mediated by plasmin (PLA) is an essential feature of wound healing, angiogenesis and cell invasion, processes that are dysregulated in cancer development, progression and systemic spread. The generation of PLA, initiated by the binding of its precursor plasminogen (PLG) to the cell surface, is regulated by an array of activators, inhibitors and receptors. In this review, we will highlight the importance of the best-characterized components of the PLG/PLA cascade in the pathogenesis of cancer focusing on the role of the cell surface-PLG receptors (PLG-R). PLG-R overexpression has been associated with poor prognosis of cancer patients and resistance to chemotherapy. We will also discuss recent findings on the molecular mechanisms regulating cell surface expression and distribution of PLG-R.
Collapse
|
15
|
Herszényi L, Barabás L, Hritz I, István G, Tulassay Z. Impact of proteolytic enzymes in colorectal cancer development and progression. World J Gastroenterol 2014; 20:13246-13257. [PMID: 25309062 PMCID: PMC4188883 DOI: 10.3748/wjg.v20.i37.13246] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 01/26/2014] [Accepted: 05/26/2014] [Indexed: 02/06/2023] Open
Abstract
Tumor invasion and metastasis is a highly complicated, multi-step phenomenon. In the complex event of tumor progression, tumor cells interact with basement membrane and extracellular matrix components. Proteolytic enzymes (proteinases) are involved in the degradation of extracellular matrix, but also in cancer invasion and metastasis. The four categories of proteinases (cysteine-, serine-, aspartic-, and metalloproteinases) are named and classified according to the essential catalytic component in their active site. We and others have shown that proteolytic enzymes play a major role not only in colorectal cancer (CRC) invasion and metastasis, but also in malignant transformation of precancerous lesions into cancer. Tissue and serum-plasma antigen concentrations of proteinases might be of great value in identifying patients with poor prognosis in CRC. Our results, in concordance with others indicate the potential tumor marker impact of proteinases for the early diagnosis of CRC. In addition, proteinases may also serve as potential target molecules for therapeutic agents.
Collapse
|
16
|
Reverse-phase protein array analysis to identify biomarker proteins in human pancreatic cancer. Dig Dis Sci 2014; 59:968-75. [PMID: 24248418 PMCID: PMC3995856 DOI: 10.1007/s10620-013-2938-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 10/28/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic cancer is the fourth leading cause of cancer death in the United States. The high mortality rate of patients with pancreatic cancer is primarily due to the difficulty of early diagnosis and a lack of effective therapies. There is an urgent need to discover novel molecular targets for early diagnosis and new therapeutic approaches to improve the clinical outcome of this deadly disease. AIM We utilized the reverse-phase protein assay (RPPA) to identify differentially expressed biomarker proteins in tumors and matched adjacent, normal-appearing tissue samples from 15 pancreatic cancer patients. METHODS The antibody panel used for the RPPA included 130 key proteins involved in various cancer-related pathways. The paired t test was used to determine the significant differences between matched pairs, and the false discovery rate-adjusted p values were calculated to take into account the effect of multiple comparisons. RESULTS After correcting for multiple comparisons, we found 19 proteins that had statistically significant differences in expression between matched pairs. However, only four (AKT, β-catenin, GAB2, and PAI-1) of them met the conservative criteria (both a q value <0.05 and a fold-change of ≥3/2 or ≤2/3) to be considered differentially expressed. Overexpression of AKT, β-catenin, and GAB2 in pancreatic cancer tissues identified by RPPA has also been further confirmed by western blot analysis. Further analysis identified several significantly associated canonical pathways and overrepresented network functions. CONCLUSION GAB2, a newly identified protein in pancreatic cancer, may provide additional insight into this cancer's pathogenesis. Future studies in a larger population are warranted to further confirm our results.
Collapse
|
17
|
Mekkawy AH, Pourgholami MH, Morris DL. Involvement of urokinase-type plasminogen activator system in cancer: an overview. Med Res Rev 2014; 34:918-56. [PMID: 24549574 DOI: 10.1002/med.21308] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Currently, there are several studies supporting the role of urokinase-type plasminogen activator (uPA) system in cancer. The association of uPA to its receptor triggers the conversion of plasminogen into plasmin. This process is regulated by the uPA inhibitors (PAI-1 and PAI-2). Plasmin promotes degradation of basement membrane and extracellular matrix (ECM) components as well as activation of ECM latent matrix metalloproteases. Degradation and remodeling of the surrounding tissues is crucial in the early steps of tumor progression by facilitating expansion of the tumor mass, release of tumor growth factors, activation of cytokines as well as induction of tumor cell proliferation, migration, and invasion. Hence, many tumors showed a correlation between uPA system component levels and tumor aggressiveness and survival. Therefore, this review summarizes the structure of the uPA system, its contribution to cancer progression, and the clinical relevance of uPA family members in cancer diagnosis. In addition, the review evaluates the significance of uPA system in the development of cancer-targeted therapies.
Collapse
Affiliation(s)
- Ahmed H Mekkawy
- Department of Surgery, Cancer Research Laboratories, St. George Hospital, University of New South Wales, Sydney, NSW 2217, Australia
| | | | | |
Collapse
|
18
|
Mengele K, Napieralski R, Magdolen V, Reuning U, Gkazepis A, Sweep F, Brünner N, Foekens J, Harbeck N, Schmitt M. Characteristics of the level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn 2014; 10:947-62. [DOI: 10.1586/erm.10.73] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Hsu LS, Wu PR, Yeh KT, Yeh CM, Shen KH, Chen CJ, Soon MS. Positive nuclear expression of KLF8 might be correlated with shorter survival in gastric adenocarcinoma. Ann Diagn Pathol 2013; 18:74-7. [PMID: 24461703 DOI: 10.1016/j.anndiagpath.2013.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/13/2013] [Indexed: 12/28/2022]
Abstract
Krűppel-like factor 8 (KLF8) is important in cell proliferation, epithelial-to-mesenchymal transition, cell migration, and invasion. Gastric adenocarcinoma is among the leading causes of cancer-related death in the world. In this study, the clinicopathologic correlation of KLF8 expression with gastric adenocarcinoma in Taiwan was investigated. The nuclear localization of KLF8 was correlated with advanced stage (P = .008) and 3-year survival rate (P = .043). The nuclear expression of KLF8 was significantly higher in the diffused type of gastric adenocarcinoma compared with the intestinal type (P = .036). Kaplan-Meier analysis results showed that patients with positive nuclear KLF8 had significantly lower overall survival rate compared with those with negative nuclear KLF8 (P = .011). Univariate analysis results indicated that positive nuclear KLF8 expression, advanced stage, and lymph node metastasis are correlated with lower overall survival. Positive nuclear KLF8 might be correlated with lower survival in gastric adenocarcinoma patients and might be an oncogene property in gastric adenocarcinoma carcinogenesis.
Collapse
Affiliation(s)
- Li-Sung Hsu
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Ru Wu
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Ken-Tu Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chung-Min Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Ko-Hung Shen
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Jun Chen
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan; Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Maw-Soan Soon
- Department of Gastroenterology, Changhua Christian Hospital, Changhua, Taiwan.
| |
Collapse
|
20
|
Noh H, Hong S, Huang S. Role of urokinase receptor in tumor progression and development. Am J Cancer Res 2013; 3:487-95. [PMID: 23843896 PMCID: PMC3706692 DOI: 10.7150/thno.4218] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 08/15/2012] [Indexed: 12/21/2022] Open
Abstract
Elevated level of urokinase receptor (uPAR) is detected in various aggressive cancer types and is closely associated with poor prognosis of cancers. Binding of uPA to uPAR triggers the conversion of plasminogen to plasmin and the subsequent activation of metalloproteinases. These events confer tumor cells with the capability to degrade the components of the surrounding extracellular matrix, thus contributing to tumor cell invasion and metastasis. uPA-uPAR interaction also elicits signals that stimulate cell proliferation/survival and the expression of tumor-promoting genes, thus assisting tumor development. In addition to its interaction with uPA, uPAR also interacts with vitronectin and this interaction promotes cancer metastasis by activating Rac and stimulating cell migration. Although underlying mechanisms are yet to be fully elucidated, uPAR has been shown to facilitate epithelial-mesenchymal transition (EMT) and induce cancer stem cell-like properties in breast cancer cells. The fact that uPAR lacks intracellular domain suggests that its signaling must be mediated through its co-receptors. Indeed, uPAR interacts with diverse transmembrane proteins including integrins, ENDO180, G protein-coupled receptors and growth factor receptors in cancer cells and these interactions are proven to be critical for the role of uPAR in tumorigenesis. Inhibitory peptide that prevents uPA-uPAR interaction has shown the promise to prolong patients' survival in the early stage of clinical trial. The importance of uPAR's co-receptor in uPAR's tumor-promoting effects implicate that anti-cancer therapeutic agents may also be developed by disrupting the interactions between uPAR and its functional partners.
Collapse
|
21
|
Kenny S, Steele I, Lyons S, Moore AR, Murugesan SV, Tiszlavicz L, Dimaline R, Pritchard DM, Varro A, Dockray GJ. The role of plasminogen activator inhibitor-1 in gastric mucosal protection. Am J Physiol Gastrointest Liver Physiol 2013; 304:G814-22. [PMID: 23494120 PMCID: PMC3652002 DOI: 10.1152/ajpgi.00017.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric mucosal health is maintained in response to potentially damaging luminal factors. Aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs) disrupt protective mechanisms leading to bleeding and ulceration. The plasminogen activator system has been implicated in fibrinolysis following gastric ulceration, and an inhibitor of this system, plasminogen activator inhibitor (PAI)-1, is expressed in gastric epithelial cells. In Helicobacter pylori-negative patients with normal gastric histology taking aspirin or NSAIDs, we found elevated gastric PAI-1 mRNA abundance compared with controls; the increase in patients on aspirin was independent of whether they were also taking proton pump inhibitors. In the same patients, aspirin tended to lower urokinase plasminogen activator mRNA. Immunohistochemistry indicated PAI-1 localization to epithelial cells. In a model system using MKN45 or AGS-GR cells transfected with a PAI-1 promoter-luciferase reporter construct, we found no evidence for upregulation of PAI-1 expression by indomethacin, and, in fact, cyclooxygenase products such as PGE2 and PGI2 weakly stimulated expression. Increased gastric PAI-1 mRNA was also found in mice following gavage with ethanol or indomethacin, but plasma PAI-1 was unaffected. In PAI-1(-/-) mice, gastric hemorrhagic lesions in response to ethanol or indomethacin were increased compared with C57BL/6 mice. In contrast, in PAI-1-H/Kβ mice in which PAI-1 is overexpressed in parietal cells, there were decreased lesions in response to ethanol and indomethacin. Thus, PAI-1 expression is increased in gastric epithelial cells in response to mucosal irritants such as aspirin and NSAIDs probably via an indirect mechanism, and PAI-1 acts as a local autoregulator to minimize mucosal damage.
Collapse
Affiliation(s)
- Susan Kenny
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - Islay Steele
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - Suzanne Lyons
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - Andrew R. Moore
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - Senthil V. Murugesan
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | | | - Rod Dimaline
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - D. Mark Pritchard
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - Andrea Varro
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| | - Graham J. Dockray
- 1Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; and
| |
Collapse
|
22
|
Ma YY, Tao HQ. Role of urokinase plasminogen activator receptor in gastric cancer: a potential therapeutic target. Cancer Biother Radiopharm 2012; 27:285-90. [PMID: 22702495 DOI: 10.1089/cbr.2012.1232] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advancements in cancer research have led to major breakthroughs; however, the impact on overall cancer-related death rate remains unacceptable. Thus, further insights into tumor markers and subsequent development of targeted therapies are urgently needed. For decades the urokinase plasminogen activator (uPA) system has been thought to drive tumor progression by mediating directed extracellular proteolysis on the surface of migrating or invading cells. Intervention with this proteolysis by targeting of uPA receptor (uPAR) has been proposed to represent a novel approach for inhibiting tumor progression. Recent data have provided new insights into the role of uPAR in gastric cancer progression. In addition to mediating proteolysis, this receptor also appears to mediate cell signaling, proliferation, and survival, and these observations have revealed novel ways to target uPAR. In this review, we discuss uPAR expression in gastric cancer, the relationship between uPAR and Helicobacter pylori, and recent insights into uPAR-signaling mechanisms. The role of uPAR as a cancer target in gastric cancer is also summarized.
Collapse
Affiliation(s)
- Ying-Yu Ma
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, Zhejiang, China
| | | |
Collapse
|
23
|
Mazzoccoli G, Pazienza V, Panza A, Valvano MR, Benegiamo G, Vinciguerra M, Andriulli A, Piepoli A. ARNTL2 and SERPINE1: potential biomarkers for tumor aggressiveness in colorectal cancer. J Cancer Res Clin Oncol 2012; 138:501-11. [PMID: 22198637 DOI: 10.1007/s00432-011-1126-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/12/2011] [Indexed: 12/24/2022]
Abstract
PURPOSE Cathepsin and plasmin may favor cancer cell invasion degrading extracellular matrix. Plasmin formation from plasminogen is regulated by plasminogen activator inhibitor type-1 (PAI-1). ARNTL2 activates the promoters of the PAI-1 gene, officially called SERPINE1, driving the circadian variation in circulating PAI-1 levels. METHODS We evaluated ARNTL2 and SERPINE1 expression in 50 colorectal cancer specimens and adjacent normal tissue and in colon cancer cell lines. RESULTS We found up-regulation of ARNTL2 (P = 0.004) and SERPINE1 (P = 0.002) in tumor tissue. A statistically significant association was found between high ARNTL2 mRNA levels and vascular invasion (P < 0.0001), and between high SERPINE1 mRNA levels and microsatellite instability (MSI-H and MSI-L, P = 0.025). Sorting the subjects into quartile groups, a statistically significant association was found between high ARNTL2 expression and lymph node involvement (P < 0.001), between high SERPINE1 expression and grading (P < 0.001) and between high SERPINE1 expression and MSI H-L (P < 0.0001). In SW480 cells, a more proliferative model compared to CaCo2 cells, there were higher mRNA levels of ARNTL2 (P < 0.001) and SERPINE1 (P = 0.001). CONCLUSION ARNTL2 and SERPINE1 expression is increased in colorectal cancer and in a highly proliferative colon cancer cell line and is related to tumor invasiveness and aggressiveness.
Collapse
Affiliation(s)
- Gianluigi Mazzoccoli
- Division of Internal Medicine and Chronobiology Unit, IRCCS Casa Sollievo della Sofferenza, Research Hospital, San Giovanni Rotondo, FG, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Ottini L, Falchetti M, Nesi G. Gene Signatures in Gastric Cancer. DIAGNOSTIC, PROGNOSTIC AND THERAPEUTIC VALUE OF GENE SIGNATURES 2012:95-113. [DOI: 10.1007/978-1-61779-358-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
25
|
Alpízar-Alpízar W, Christensen IJ, Santoni-Rugiu E, Skarstein A, Ovrebo K, Illemann M, Laerum OD. Urokinase plasminogen activator receptor on invasive cancer cells: A prognostic factor in distal gastric adenocarcinoma. Int J Cancer 2011; 131:E329-36. [DOI: 10.1002/ijc.26417] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 08/23/2011] [Indexed: 12/27/2022]
|
26
|
Nørsett KG, Steele I, Duval C, Sammut SJ, Murugesan SVM, Kenny S, Rainbow L, Dimaline R, Dockray GJ, Pritchard DM, Varro A. Gastrin stimulates expression of plasminogen activator inhibitor-1 in gastric epithelial cells. Am J Physiol Gastrointest Liver Physiol 2011; 301:G446-53. [PMID: 21193525 PMCID: PMC3174540 DOI: 10.1152/ajpgi.00527.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Plasminogen activator inhibitor (PAI)-1 is associated with cancer progression, fibrosis and thrombosis. It is expressed in the stomach but the mechanisms controlling its expression there, and its biological role, are uncertain. We sought to define the role of gastrin in regulating PAI-1 expression and to determine the relevance for gastrin-stimulated cell migration and invasion. In gastric biopsies from subjects with elevated plasma gastrin, the abundances of PAI-1, urokinase plasminogen activator (uPA), and uPA receptor (uPAR) mRNAs measured by quantitative PCR were increased compared with subjects with plasma concentrations in the reference range. In patients with hypergastrinemia due to autoimmune chronic atrophic gastritis, there was increased abundance of PAI-1, uPA, and uPAR mRNAs that was reduced by octreotide or antrectomy. Immunohistochemistry revealed localization of PAI-1 to parietal cells and enterochromaffin-like cells in micronodular neuroendocrine tumors in hypergastrinemic subjects. Transcriptional mechanisms were studied by using a PAI-1-luciferase promoter-reporter construct transfected into AGS-G(R) cells. There was time- and concentration-dependent increase of PAI-1-luciferase expression in response to gastrin that was reversed by inhibitors of the PKC and MAPK pathways. In Boyden chamber assays, recombinant PAI-1 inhibited gastrin-stimulated AGS-G(R) cell migration and invasion, and small interfering RNA treatment increased responses to gastrin. We conclude that elevated plasma gastrin concentrations are associated with increased expression of gastric PAI-1, which may act to restrain gastrin-stimulated cell migration and invasion.
Collapse
Affiliation(s)
| | | | | | | | - Senthil V. M. Murugesan
- 1Physiological Laboratory and ,2Department of Gastroenterology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | - D. Mark Pritchard
- 2Department of Gastroenterology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
27
|
Schmitt M, Mengele K, Napieralski R, Magdolen V, Reuning U, Gkazepis A, Sweep F, Brünner N, Foekens J, Harbeck N. Clinical utility of level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn 2011; 10:1051-67. [PMID: 21080821 DOI: 10.1586/erm.10.71] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The prognostic and/or predictive value of the cancer biomarkers, urokinase-type plasminogen activator (uPA) and its inhibitor (plasminogen activator inhibitor [PAI]-1), determined by ELISA in tumor-tissue extracts, was demonstrated for several cancer types in numerous clinically relevant retrospective or prospective studies, including a multicenter breast cancer therapy trial (Chemo-N0). Consequently, for the first time ever for any cancer biomarker for breast cancer, uPA and PAI-1 have reached the highest level of evidence, level-of-evidence-1. At present, two other breast cancer therapy trials, NNBC-3 and Plan B, also incorporating uPA and PAI-1 as treatment-assignment tools are in effect. Furthermore, small synthetic molecules targeting uPA are currently in Phase II clinical trials in patients afflicted with advanced cancer of the ovary, breast or pancreas.
Collapse
Affiliation(s)
- Manfred Schmitt
- Frauenklinik der Technischen Universitaet Muenchen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nobili S, Bruno L, Landini I, Napoli C, Bechi P, Tonelli F, Rubio CA, Mini E, Nesi G. Genomic and genetic alterations influence the progression of gastric cancer. World J Gastroenterol 2011; 17:290-9. [PMID: 21253387 PMCID: PMC3022288 DOI: 10.3748/wjg.v17.i3.290] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 08/09/2010] [Accepted: 08/16/2010] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the leading causes of cancer-related deaths worldwide, although the incidence has gradually decreased in many Western countries. Two main gastric cancer histotypes, intestinal and diffuse, are recognised. Although most of the described genetic alterations have been observed in both types, different genetic pathways have been hypothesized. Genetic and epigenetic events, including 1q loss of heterozygosity (LOH), microsatellite instability and hypermethylation, have mostly been reported in intestinal-type gastric carcinoma and its precursor lesions, whereas 17p LOH, mutation or loss of E-cadherin are more often implicated in the development of diffuse-type gastric cancer. In this review, we summarize the sometimes contradictory findings regarding those markers which influence the progression of gastric adenocarcinoma.
Collapse
|
29
|
Liu D, Overbey D, Watkinson L, Giblin MF. Synthesis and characterization of an (111)In-labeled peptide for the in vivo localization of human cancers expressing the urokinase-type plasminogen activator receptor (uPAR). Bioconjug Chem 2010; 20:888-94. [PMID: 19354275 DOI: 10.1021/bc800433y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study describes the synthesis and preliminary biologic evaluation of an (111)In-labeled peptide antagonist of the urokinase-type plasminogen activator receptor (uPAR) as a potential probe for assessing metastatic potential of human breast cancer in vivo. The peptide (NAc-dD-CHA-F-dS-dR-Y-L-W-S-betaAla)(2)-K-K(DOTA)-NH(2) was synthesized and conjugated with the DOTA chelating moiety via conventional solid-phase peptide synthesis (SPPS), purified by reversed-phase HPLC, and characterized by MALDI-TOF MS and receptor binding assay. In vitro receptor binding studies demonstrated an IC(50) of 240 +/- 125 nM for the peptide, compared with IC(50) values of 0.44 +/- 0.02 and 0.75 +/- 0.01 nM for the amino terminal fragment (ATF) of the urokinase-type plasminogen activator (uPA) and full-length uPA, respectively. In vivo biodistribution studies were carried out using SCID mice bearing MDA-MB-231 human breast cancer xenografts. Biodistribution data was collected at 1, 4, and 24 h postinjection of (111)In-DOTA-peptide, and compared with data obtained using a scrambled control peptide as well as with data obtained using wild-type ATF radiolabeled with I-125. Biodistribution studies showed rapid elimination of the (111)-labeled peptide from the blood pool, with 0.12 +/- 0.06% ID/g remaining in blood at 4 h pi. Elimination was seen primarily via the renal/urinary route, with 83.9 +/- 2.2% ID in the urine at the same time point. Tumor uptake at this time was 0.53 +/- 0.11% ID/g, resulting in tumor/blood and tumor/muscle ratios of 4.2 and 9.4, respectively. Uptake in tumor was significantly higher than that obtained using a scrambled control peptide that showed no specific binding to uPAR (p < 0.05). In vitro and ex vivo results both suggested that the magnitude of tumor-specific binding was reduced in this model by endogenous expression of uPA. The results indicate that radiolabeled peptide uPAR antagonists may find application in the imaging and therapy of uPAR-expressing breast cancers in vivo.
Collapse
Affiliation(s)
- Dijie Liu
- Research Service, Harry S. Truman Memorial Veterans' Administration Hospital, Columbia, MO 65201, USA
| | | | | | | |
Collapse
|
30
|
Körner A, Mudduluru G, Manegold C, Allgayer H. Enzastaurin inhibits invasion and metastasis in lung cancer by diverse molecules. Br J Cancer 2010; 103:802-11. [PMID: 20736951 PMCID: PMC2966618 DOI: 10.1038/sj.bjc.6605818] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/22/2010] [Accepted: 06/28/2010] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Enzastaurin (Enz) is a serine/threonine kinase inhibitor blocking protein kinase C (PKC)beta/AKT pathway. However, an ability of this compound to inhibit cancer invasion and metastasis is not yet clearly elucidated. METHODS The ability of Enz to inhibit invasion and metastasis, and to target molecules was investigated in non-small cell lung cancer (NSCLC) by RT-PCR validated microarray, Matrigel, and in vivo chorionallantoic membrane (CAM) assays. RESULTS Enzastaurin significantly reduced migration, invasion, and in vivo metastasis to lungs and liver (CAM assay) of diverse NSCLC cell lines. Genes promoting cancer progression (u-PAR, VEGFC, and HIF1alpha) and tumour suppression (VHL, RASSF1, and FHIT) of NSCLC were significantly (P<0.05) down- or upregulated after Enz treatment in H460, A549, and H1299 cells, respectively. Luciferase/chromatin immunoprecipitation analysis showed that Enz transcriptionally controls urokinase-type plasminogen activator receptor (u-PAR) expression by promoter inhibition through Sp1, Sp3, and c-Jun(AP-1). Moreover, siRNA knockdown of u-PAR re-sensitised Enz-resistant cells and induced apoptosis, suggesting u-PAR as a marker of Enz resistance. CONCLUSION This study shows that Enz inhibits migration, invasion, and in vivo metastasis by targeting u-PAR, besides further targeting progression-related and tumour-suppressor genes in NSCLC. Together with u-PAR being a novel putative marker of Enz response, these data encourage molecularly tailored clinical studies on Enz in NSCLC therapy.
Collapse
Affiliation(s)
- A Körner
- Department of Experimental Surgery Mannheim/Molecular Oncology of Solid Tumors (German Cancer Research Center-DKFZ-Heidelberg), Mannheim Medical Faculty, Ruprecht-Karls-University Heidelberg, Mannheim 68167, Germany
| | - G Mudduluru
- Department of Experimental Surgery Mannheim/Molecular Oncology of Solid Tumors (German Cancer Research Center-DKFZ-Heidelberg), Mannheim Medical Faculty, Ruprecht-Karls-University Heidelberg, Mannheim 68167, Germany
| | - C Manegold
- Interdisciplinary Thoracic Oncology, Department of Surgery, Medical Faculty Mannheim, University Heidelberg, Mannheim 68167, Germany
| | - H Allgayer
- Department of Experimental Surgery Mannheim/Molecular Oncology of Solid Tumors (German Cancer Research Center-DKFZ-Heidelberg), Mannheim Medical Faculty, Ruprecht-Karls-University Heidelberg, Mannheim 68167, Germany
| |
Collapse
|
31
|
Abstract
The urokinase receptor (u-PAR) is one of the most critical molecules in migration, invasion, intravasation, and metastasis and is also a key regulator between tumour cell proliferation and dormancy. It is overexpressed in most human solid cancer types, which has led to increasing translational and clinical research on this molecule. The current review discusses in particular the in vivo, translational, and putative clinical relevance of u-PAR in the context of this latest development. It outlines how u-PAR is already being used and might increasingly be applied as a diagnostic tool, for example, in distinguishing benign from malignant neoplasms, as a molecular marker for predicting clinical response to chemotherapy or novel targeted therapy, and finally as a promising tool for the development of novel cancer therapeutics.
Collapse
|
32
|
Alpízar-Alpízar W, Nielsen BS, Sierra R, Illemann M, Ramírez JA, Arias A, Durán S, Skarstein A, Ovrebo K, Lund LR, Laerum OD. Urokinase plasminogen activator receptor is expressed in invasive cells in gastric carcinomas from high- and low-risk countries. Int J Cancer 2010; 126:405-15. [PMID: 19609941 DOI: 10.1002/ijc.24755] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gastric cancer is the second cancer causing death worldwide. Both incidence and mortality rates vary according to geographical regions. The receptor for urokinase plasminogen activator (uPAR) is involved in extracellular matrix degradation by mediating cell surface associated plasminogen activation, and its presence on gastric cancer cells is linked to micro-metastasis and poor prognosis. Immunohistochemical analyses of a set of 44 gastric cancer lesions from Costa Rica showed expression of uPAR in cancer cells in both intestinal subtype (14 of 27) and diffuse subtype (10 of 17). We compared the expression pattern of uPAR in gastric cancers from a high-risk country (Costa Rica) with a low-risk country (Norway). We found uPAR on gastric cancer cells in 24 of 44 cases (54%) from Costa Rica and in 13 of 23 cases (56%) from Norway. uPAR was seen in macrophages and neutrophils in all cases. We also examined the nonneoplastic mucosa and found that uPAR was more frequently seen in epithelial cells located at the luminal edge of the crypts in cases with Helicobacter pylori infection than in similar epithelial cells in noninfected mucosa (p = 0.033; chi(2) = 4.54). In conclusion, the expression of uPAR in cancer cells in more than half of the gastric cancer cases suggests that their uPAR-positivity do not contribute to explain the different mortality rates between the 2 countries, however, the actual prevalence of uPAR-positive cancer cells in the gastric cancers may still provide prognostic information.
Collapse
Affiliation(s)
- Warner Alpízar-Alpízar
- The Gade Institute, University of Bergen and Department of Pathology, Haukeland University Hospital, Norway.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Palwai NR, Zang XP, Harrison RG, Benbrook D, Pento JT. Selective growth inhibition of cancer cells by L-methioninase-containing fusion protein targeted to the urokinase receptor. Pharmacology 2009; 84:271-5. [PMID: 19797936 DOI: 10.1159/000242997] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 07/13/2009] [Indexed: 11/19/2022]
Abstract
BACKGROUND We have reported the development of a novel fusion protein (FP) consisting of an amino-terminal fragment of urokinase linked to the amino terminus of the enzyme L-methioninase (L-M). The present study compared the effect of this novel FP on the proliferation of human ovarian, skin, breast endometrial and pancreatic cancer cell lines. METHODS The FP, L-M and a mutated FP, with reduced L-M activity, were produced by recombinant methods. The effect of treatment with FP, L-M and mutated FP on the proliferation of the cancer cells was measured in vitro using an MTS assay. RESULTS The inhibitory effect of the FP was found to be significantly greater than that of L-M alone or the mutated FP. In addition, the FP produced a greater inhibitory effect on an ovarian cancer cell line than on comparable normal, non-cancerous cells. Further, the FP produced a dose-dependent inhibition of the proliferation of pancreatic cancer cell lines. CONCLUSION These results suggest that this FP is a potent and selective inhibitor of the proliferation of various cancer cell lines and has potential as a therapeutic agent for the treatment of various methionine-dependent cancers.
Collapse
Affiliation(s)
- Naveen R Palwai
- Bioengineering Center and School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma 73117, USA
| | | | | | | | | |
Collapse
|
34
|
Cancer-specific MALDI-TOF profiles of blood serum and plasma: biological meaning and perspectives. J Proteomics 2009; 73:537-51. [PMID: 19782778 DOI: 10.1016/j.jprot.2009.09.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 09/14/2009] [Accepted: 09/16/2009] [Indexed: 12/13/2022]
Abstract
MALDI-TOF mass-spectrometry has become a popular tool of cancer research during the last decade. High throughput and relative simplicity of this technology have made it attractive for biomarker discovery and validation across various platforms in blood serum/plasma. Many technical approaches have been developed for plasma/serum profiling including protein-chip based SELDI-TOF mass-spectrometry, purification of serum on magnetic beads, analysis of carrier-associated fraction and mass-spectrometric immunoassays. Extensive data about the identity of differential features detected on mass-spectra up to now makes it possible to draw conclusions about potency and perspectives of MALDI-TOF mass-spectrometry in this field. A great majority of identified differentially expressed proteins are either house-keeping or inflammatory proteins as well as their modifications or fragments. Discriminating ability of mass-spectra is likely to be based on differential modification and fragmentation patterns of abundant serum proteins reflecting activity of enzymes including proteases and their inhibitors.
Collapse
|
35
|
Huang WC, Hung MC. Induction of Akt activity by chemotherapy confers acquired resistance. J Formos Med Assoc 2009; 108:180-94. [PMID: 19293033 DOI: 10.1016/s0929-6646(09)60051-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Resistance to chemotherapy is a major cause of treatment failure in human cancer. Accumulating evidence has indicated that the acquisition of resistance to chemotherapeutic drugs involves the activation of the PI3K/Akt pathway. Modulating Akt activity in response to chemotherapy has been observed often in chemoresistant cancers. The potential molecular mechanisms by which chemotherapeutic agents activate the PI3K/Akt pathway are emerging. Activation of this pathway evades the cytotoxic effects of chemotherapeutic agents via regulation of essential cellular functions such as protein synthesis, antiapoptosis, survival and proliferation in cancer. How chemotherapeutic agents induce Akt activation and how activated Akt confers chemoresistance through regulation of signaling networks are discussed in this review. Combining PI3K/Akt inhibitors with standard chemotherapy has been successful in increasing the efficacy of chemotherapeutic agents both in vivo and in vitro. Several small molecules have been developed to specifically target PI3K/Akt and other components of this pathway, which in combination with chemotherapy may be a valid approach to overcome therapeutic resistance. We propose several feedback and feedforward regulatory mechanisms of signaling networks for maintenance of the Akt activity for cell survival. These regulatory mechanisms may limit the efficacy of PI3K/Akt-targeted therapy; therefore, disruption of these mechanisms may be an effective strategy for development of novel anti-cancer therapies.
Collapse
Affiliation(s)
- Wei-Chien Huang
- Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University and Hospital, and Department of Biotechnology, Asia University, Taichung, Taiwan.
| | | |
Collapse
|
36
|
Buergy D, Weber T, Maurer GD, Mudduluru G, Medved F, Leupold JH, Brauckhoff M, Post S, Dralle H, Allgayer H. Urokinase receptor, MMP-1 and MMP-9 are markers to differentiate prognosis, adenoma and carcinoma in thyroid malignancies. Int J Cancer 2009; 125:894-901. [PMID: 19480010 DOI: 10.1002/ijc.24462] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The identification of high-risk patients with thyroid cancer and the preoperative differentiation between follicular adenoma and carcinoma remain clinically challenging. Our study was conducted to analyze whether the quantification of matrix metalloproteinases (MMPs) and urokinase-type plasminogen activator receptor (u-PAR) and transcription factor binding to the u-PAR promoter improve prognostic predictability and differential diagnosis of thyroid tumors. Tumor/normal tissue was collected from 69 prospectively followed patients with thyroid carcinomas (papillary, medullary, follicular and anaplastic, PTC, MTC, FTC and ATC) or follicular adenomas. U-PAR, MMP-1, MMP-7 and MMP-9 amounts were determined by ELISA, and transcription factor binding was determined by electrophoretic mobility shift assay. Binding of transcription factors to the u-PAR promoter was observed, but not associated with u-PAR expression. Carcinomas except MTC expressed significantly more u-PAR/MMPs than adenomas/normal tissues, this being associated with advanced pT- or M-stages. MMP-1 and MMP-9 were significantly higher in follicular carcinomas than in adenomas. In carcinomas, high u-PAR-gene expression correlated significantly with high MMP-9, the latter being associated with MMP-7 in normal tissues. Poor survival in differentiated tumors was associated in trend (p = 0.07); poor survival of all patients (p = 0.043) and especially of patients with carcinomas of follicular origin (including ATC), but not medullary carcinomas, were significantly associated with high u-PAR-protein (p = 0.015). Quantification of u-PAR is of prognostic relevance in thyroid carcinomas of non-c-cell origin, and u-PAR in part may be regulated nontranscriptionally in thyroid cancers. This is the first study to suggest MMP-1/-9 as significant differentiation markers between follicular adenoma and follicular carcinoma.
Collapse
Affiliation(s)
- Daniel Buergy
- Department of Experimental Surgery and Molecular Oncology of Solid Tumors, Mannheim Faculty, University of Heidelberg, and DKFZ Heidelberg, Mannheim, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Nikolova DA, Asangani IA, Nelson LD, Hughes DPM, Siwak DR, Mills GB, Harms A, Buchholz E, Pilz LR, Manegold C, Allgayer H. Cetuximab attenuates metastasis and u-PAR expression in non-small cell lung cancer: u-PAR and E-cadherin are novel biomarkers of cetuximab sensitivity. Cancer Res 2009; 69:2461-70. [PMID: 19276367 DOI: 10.1158/0008-5472.can-08-3236] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cetuximab, which blocks ligand binding to epidermal growth factor receptor (EGFR), is currently being studied as a novel treatment for non-small cell lung cancer (NSCLC). However, its mechanisms of action toward metastasis, and markers of drug sensitivity, have not been fully elucidated. This study was conducted to (a) determine the effect of Cetuximab on invasion and NSCLC-metastasis; (b) investigate urokinase-type plasminogen activator receptor (u-PAR), a major molecule promoting invasion and metastasis, as a target molecule; (c) delineate molecular mediators of Cetuximab-induced metastasis inhibition; and (d) identify biomarkers of drug sensitivity in NSCLC. Cetuximab treatment resulted in reduced growth and Matrigel invasion of H1395 and A549 NSCLC cell lines, in parallel with reduced u-PAR mRNA and protein. u-PAR down-regulation was brought about by suppressing the binding of JunD and c-Jun to u-PAR promoter motif -190/-171 in vivo, and an inhibition of MAP/ERK kinase signaling. Furthermore, Cetuximab inhibited NSCLC proliferation and metastasis to distant organs in vivo as indicated by the chicken embryo metastasis assay. Low E-cadherin and high u-PAR, but not EGFR, was associated with resistance to Cetuximab in seven NSCLC cell lines. Furthermore, siRNA knockdown of u-PAR led to a resensitization to Cetuximab. Moreover, low E-cadherin and high u-PAR was found in 63% of resected tumor tissues of NSCLC patients progressing under Cetuximab therapy. This is the first study to show u-PAR as a target and marker of sensitivity to Cetuximab, and to delineate novel mechanisms leading to metastasis suppression of NSCLC by Cetuximab.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/pharmacology
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Cadherins/biosynthesis
- Cadherins/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cetuximab
- Chick Embryo
- Epidermal Growth Factor/antagonists & inhibitors
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/metabolism
- Gene Expression
- Humans
- JNK Mitogen-Activated Protein Kinases/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- MAP Kinase Signaling System
- Neoplasm Metastasis
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-jun/metabolism
- Receptors, Urokinase Plasminogen Activator/biosynthesis
- Receptors, Urokinase Plasminogen Activator/genetics
Collapse
Affiliation(s)
- Dessislava A Nikolova
- Department of Experimental Surgery and Molecular Oncology of Solid Tumors, Medical Faculty Mannheim, University Heidelberg and DKFZ (German Cancer Research Center), Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Components of the plasminogen-plasmin system participate in a wide variety of physiologic and pathologic processes, including tumor growth, invasion and metastasis, through their effect on angiogenesis and cell migration. These components are found in most tumors and their expression not only signifies their function but also carries a prognostic value. Their expression is in turn modulated by cytokines and growth factors, many of which are up-regulated in cancer. Though both tPA and uPA are expressed in tumor cells, uPA with its receptor (uPAR) is mostly involved in cellular functions, while tPA with its receptor Annexin II on endothelial surface, regulates intravascular fibrin deposition. Among the inhibitors of fibrinolysis, PAI-1 is a major player in the pathogenesis of many vascular diseases as well as in cancer. Therapeutic interventions, either using plasminogen activators or experimental inhibitor agents against PAI-1, have shown encouraging results in experimental tumors but not been verified clinically.
Collapse
Affiliation(s)
- Hau C Kwaan
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | | |
Collapse
|
39
|
Plasminogen activator inhibitor 1 protects fibrosarcoma cells from etoposide-induced apoptosis through activation of the PI3K/Akt cell survival pathway. Neoplasia 2008; 10:1083-91. [PMID: 18813358 DOI: 10.1593/neo.08486] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 06/26/2008] [Accepted: 06/27/2008] [Indexed: 11/18/2022] Open
Abstract
High levels of plasminogen activator inhibitor (PAI-1) in tumors are associated with poor prognosis in several cancer types, and the reason for this association is not fully understood. Plasminogen activator inhibitor 1 has been suggested to contribute to tumor growth by protecting cancer cells from apoptosis, and we have previously shown that wild type murine fibrosarcoma cells are significantly more resistant to apoptosis induced by chemotherapy than PAI-1-deficient fibrosarcoma cells. Here, we further investigated the molecular mechanisms underlying the antiapoptotic function of PAI-1 focusing on the phosphatidylinositol 3-phosphate kinase (PI3K)/Akt cell survival pathway. We demonstrate that the activation level of the Akt cell survival pathway is reduced in PAI-1-deficient cells. Inhibition of either PI3K or Akt by synthetic inhibitors sensitized the wild type but not the PAI-1-deficient cells to etoposide-induced cell death. More importantly, reintroduction of PAI-1 expression in PAI-1-deficient cells induced an increase in Akt activity and protection against etoposide-induced apoptosis. Concordantly, silencing of PAI-1 by RNA interference in wild type fibrosarcoma cells decreased the level of active Akt, and this was accompanied by a sensitization of the cells to etoposide-induced cell death. Altogether, our data suggest that PAI-1 influences sensitivity to etoposide-induced apoptosis through the PI3K/Akt cell survival pathway by acting upstream of PI3K and Akt. This points to PAI-1 as a possible therapeutic target in cancer diseases where PAI-1 inhibits chemotherapy-induced apoptosis.
Collapse
|
40
|
Serum cathepsin B and plasma urokinase-type plasminogen activator levels in gastrointestinal tract cancers. Eur J Cancer Prev 2008; 17:438-45. [PMID: 18714186 DOI: 10.1097/cej.0b013e328305a130] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cathepsin B (CATB) and urokinase-type plasminogen activator (UPA) play an important part in cancer invasion and metastasis. The behavior of CATB and UPA has not been evaluated in the same experimental setting in different gastrointestinal tumors and in precancerous lesions. Serum CATB and plasma UPA levels were determined by enzyme-linked immunoadsorbent assay and their sensitivity, specificity, and diagnostic accuracy have been calculated in patients with colorectal (n=72), gastric (n=30), hepatocellular (n=28), and pancreatic cancer (n=15) as well as in gastric epithelial dysplasia (n=25), colorectal adenomas (n=30), and tumor-free control patients (n=44). Serum CATB and plasma UPA antigen concentrations were significantly higher in patients with cancer than in controls. When all tumors were considered, the sensitivity, specificity, and diagnostic accuracy of CATB (89, 86, and 89%) were higher than that of UPA (76, 70, and 74%). CATB demonstrated in all types of tumors a better diagnostic accuracy than UPA. The positive predictive values of CATB (95%) and UPA (89%) may suggest their use in the evaluation of patients with a suspicion of malignancy. CATB and UPA were significantly higher in patients with gastric epithelial dysplasia and colorectal adenomas than in controls. Antigen levels of CATB and UPA were significantly correlated in both cancers and precancerous lesions. At the time of clinical presentation, serum CATB and plasma UPA antigen levels are sensitive indicators of gastrointestinal malignancies. Determination of serum CATB and plasma UPA levels may be useful to identify patients at a higher risk for progression to cancer, who could be subjected to a more strict follow-up protocol.
Collapse
|
41
|
Allgayer H, Aguirre-Ghiso JA. The urokinase receptor (u-PAR)--a link between tumor cell dormancy and minimal residual disease in bone marrow? APMIS 2008; 116:602-14. [PMID: 18834405 DOI: 10.1111/j.1600-0463.2008.00997.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Minimal residual disease (MRD) is hypothesized to be the major cause of tumor recurrence and metastasis even years and decades after primary cancer diagnosis and curative solid tumor resection. In these patients disseminated tumor cells reflecting MRD can be detected in the bone marrow years after treatment. It is to be assumed that genetic determinants and a complex interplay between the disseminated tumor cells and their microenvironment in the bone marrow are responsible for tumor cell dormancy and the final reactivation towards metastasis. The urokinase receptor (u-PAR), a critical regulator of invasion, intravasation, and metastasis, is found to be a key player in regulating the shift between single cell tumor dormancy and proliferation. This has mainly been attributed to a regulation by u-PAR of integrins, and the ability of the latter to propagate signals from fibronectin through the EGF-receptor, ERK, and p38 signaling. Interestingly, u-PAR is found in disseminated tumor cells in the bone marrow of solid cancer patients, and is associated with the expansion of these cells and clinical prognosis. Here we summarize and discuss findings on disseminated tumor cells in the bone marrow, MRD and the role of u-PAR in tumor biology, especially focusing on its specific role in providing a switch between tumor cell proliferation and dormancy. Finally, we discuss the hypothesis that u-PAR might be an essential molecule in bone marrow disseminated tumor cells for long-term survival during dormancy, and/or reactivation of their proliferation years after primary treatment.
Collapse
Affiliation(s)
- Heike Allgayer
- Department of Experimental Surgery and Molecular Oncology of Solid Tumors, Medical Faculty Mannheim, University of Heidelberg, and DKFZ German Cancer Research Center Heidelberg, Germany.
| | | |
Collapse
|
42
|
Deshet N, Lupu‐Meiri M, Espinoza I, Fili O, Shapira Y, Lupu R, Gershengorn MC, Oron Y. Plasminogen‐induced aggregation of PANC‐1 cells requires conversion to plasmin and is inhibited by endogenous plasminogen activator inhibitor‐1. J Cell Physiol 2008; 216:632-9. [DOI: 10.1002/jcp.21441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Kenny S, Duval C, Sammut SJ, Steele I, Pritchard DM, Atherton JC, Argent RH, Dimaline R, Dockray GJ, Varro A. Increased expression of the urokinase plasminogen activator system by Helicobacter pylori in gastric epithelial cells. Am J Physiol Gastrointest Liver Physiol 2008; 295:G431-41. [PMID: 18599586 PMCID: PMC2536790 DOI: 10.1152/ajpgi.90283.2008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The gastric pathogen Helicobacter pylori (H. pylori) is linked to peptic ulcer and gastric cancer, but the relevant pathophysiological mechanisms are unclear. We now report that H. pylori stimulates the expression of plasminogen activator inhibitor (PAI)-1, urokinase plasminogen activator (uPA), and its receptor (uPAR) in gastric epithelial cells and the consequences for epithelial cell proliferation. Real-time PCR of biopsies from gastric corpus, but not antrum, showed significantly increased PAI-1, uPA, and uPAR in H. pylori-positive patients. Transfection of primary human gastric epithelial cells with uPA, PAI-1, or uPAR promoters in luciferase reporter constructs revealed expression of all three in H+/K+ATPase- and vesicular monoamine transporter 2-expressing cells; uPA was also expressed in pepsinogen- and uPAR-containing trefoil peptide-1-expressing cells. In each case expression was increased in response to H. pylori and for uPA, but not PAI-1 or uPAR, required the virulence factor CagE. H. pylori also stimulated soluble and cell surface-bound uPA activity, and both were further increased by PAI-1 knockdown, consistent with PAI-1 inhibition of endogenous uPA. H. pylori stimulated epithelial cell proliferation, which was inhibited by uPA immunoneutralization and uPAR knockdown; exogenous uPA also stimulated proliferation that was further increased after PAI-1 knockdown. The proliferative effects of uPA were inhibited by immunoneutralization of the EGF receptor and of heparin-binding EGF (HB-EGF) by the mutant diphtheria toxin CRM197 and an EGF receptor tyrosine kinase inhibitor. H. pylori induction of uPA therefore leads to epithelial proliferation through activation of HB-EGF and is normally inhibited by concomitant induction of PAI-1; treatments directed at inhibition of uPA may slow the progression to gastric cancer.
Collapse
Affiliation(s)
- Susan Kenny
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Cedric Duval
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Stephen J. Sammut
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Islay Steele
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - D. Mark Pritchard
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - John C. Atherton
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Richard H. Argent
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Rod Dimaline
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Graham J. Dockray
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| | - Andrea Varro
- Physiological Laboratory, School of Biomedical Sciences, Division of Gastroenterology, School of Clinical Sciences, University of Liverpool, Liverpool, United Kingdom; and Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, United Kingdom
| |
Collapse
|
44
|
Herszényi L, Farinati F, Cardin R, István G, Molnár LD, Hritz I, De Paoli M, Plebani M, Tulassay Z. Tumor marker utility and prognostic relevance of cathepsin B, cathepsin L, urokinase-type plasminogen activator, plasminogen activator inhibitor type-1, CEA and CA 19-9 in colorectal cancer. BMC Cancer 2008; 8:194. [PMID: 18616803 PMCID: PMC2474636 DOI: 10.1186/1471-2407-8-194] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Accepted: 07/10/2008] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cathepsin B and L (CATB, CATL), urokinase-type plasminogen activator (uPA) and its inhibitor PAI-1 play an important role in colorectal cancer invasion. The tumor marker utility and prognostic relevance of these proteases have not been evaluated in the same experimental setting and compared with that of CEA and CA-19-9. METHODS Protease, CEA and CA 19-9 serum or plasma levels were determined in 56 patients with colorectal cancer, 25 patients with ulcerative colitis, 26 patients with colorectal adenomas and 35 tumor-free control patients. Protease, CEA, CA 19-9 levels have been determined by ELISA and electrochemiluminescence immunoassay, respectively; their sensitivity, specificity, diagnostic accuracy have been calculated and correlated with clinicopathological staging. RESULTS The protease antigen levels were significantly higher in colorectal cancer compared with other groups. Sensitivity of PAI-1 (94%), CATB (82%), uPA (69%), CATL (41%) were higher than those of CEA or CA 19-9 (30% and 18%, respectively). PAI-1, CATB and uPA demonstrated a better accuracy than CEA or CA 19-9. A combination of PAI-1 with CATB or uPA exhibited the highest sensitivity value (98%). High CATB, PAI-1, CEA and CA 19-9 levels correlated with advanced Dukes stages. CATB (P = 0.0004), CATL (P = 0.02), PAI-1 (P = 0.01) and CA 19-9 (P = 0.004) had a significant prognostic impact. PAI-1 (P = 0.001), CATB (P = 0.04) and CA 19-9 (P = 0.02) proved as independent prognostic variables. CONCLUSION At the time of clinical detection proteases are more sensitive indicators for colorectal cancer than the commonly used tumor markers. Determinations of CATB, CATL and PAI-1 have a major prognostic impact in patients with colorectal cancer.
Collapse
Affiliation(s)
- László Herszényi
- 2nd Department of Medicine, Semmelweis University, Budapest Hungarian Academy of Science, Clinical Gastroenterology Research Unit, Budapest, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Helicobacter pylori infection stimulates plasminogen activator inhibitor 1 production by gastric epithelial cells. Infect Immun 2008; 76:3992-9. [PMID: 18519558 DOI: 10.1128/iai.00584-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic infection with the gastric pathogen Helicobacter pylori significantly increases the risk of developing atrophic gastritis, peptic ulcer disease, and gastric adenocarcinoma. H. pylori strains that possess the cag pathogenicity island, which translocates CagA into the host cells, augment these risks. The aim of this study was to determine the molecular mechanisms through which H. pylori upregulates the expression of plasminogen activator inhibitor 1 (PAI-1), a member of the urokinase activator system that is involved in tumor metastasis and angiogenesis. Levels of PAI-1 mRNA and protein were examined in tissues from H. pylori-infected patients and in vitro using AGS gastric epithelial cells. In vitro, cells were infected with toxigenic cag-positive or nontoxigenic cag-negative strains of H. pylori or isogenic mutants. The amount of PAI-1 secretion was measured by enzyme-linked immunosorbent assay, and mRNA levels were determined using real-time PCR. The regulation of PAI-1 was examined using the extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor and small interfering RNA. Analysis of human biopsy samples revealed an increase in both PAI-1 mRNA and protein levels in patients with H. pylori gastritis compared to those of uninfected controls. Infection of AGS cells with H. pylori significantly increased PAI-1 mRNA expression and the secretion of PAI-1 protein. Moreover, PAI-1 mRNA and protein production was more pronounced when AGS cells were infected by H. pylori strains carrying a functional cag secretion system than when cells were infected by strains lacking this system. PAI-1 secretion was also reduced when cells were infected with either cagE-negative or cagA-negative mutants. The ectopic overexpression of CagA significantly increased the levels of PAI-1 mRNA and protein, whereas blockade of the ERK1/2 pathway inhibited H. pylori-mediated PAI-1 upregulation. These findings suggest that the upregulation of PAI-1 in H. pylori-infected gastric epithelial cells may contribute to the carcinogenic process.
Collapse
|
46
|
Mudduluru G, Medved F, Grobholz R, Jost C, Gruber A, Leupold JH, Post S, Jansen A, Colburn NH, Allgayer H. Loss of programmed cell death 4 expression marks adenoma-carcinoma transition, correlates inversely with phosphorylated protein kinase B, and is an independent prognostic factor in resected colorectal cancer. Cancer 2007; 110:1697-707. [PMID: 17849461 DOI: 10.1002/cncr.22983] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Programmed cell death 4 (Pdcd4) inhibits malignant transformation, and initial studies of Pdcd4 suggested the regulation of Pdcd4 localization by protein kinase B (Akt). However, supporting patient tissue data are missing, and the diagnostic/prognostic potential of Pdcd4 rarely has been studied. The objectives of the current were 1) to determine Pdcd4 as a diagnostic marker in the adenoma-carcinoma sequence, 2) to support phosphorylated Akt (pAkt)-mediated Pdcd4 regulation in vivo, and 3) to obtain the first prognostic evidence of Pdcd4 in colorectal cancer. METHODS Tumor samples and normal tissues from 71 patients with colorectal cancer who were followed prospectively (median follow-up, 36 months) and 42 adenomas were analyzed for Pdcd4, Akt, and pAkt in immunohistochemical and Western blot analyses. RESULTS A significant reduction in Pdcd4 was observed between normal mucosa and adenomas and between adenomas and tumor samples (P < .01 and P < .01, respectively). Normal mucosa demonstrated strong nuclear Pdcd4, which was reduced significantly in adenomas (P < .01) and almost was lost in tumors (P < .01). pAkt was correlated inversely with Pdcd4 and with the transition of Pdcd4 from nucleus to cytoplasm (P < .01). Kaplan-Meier analysis (using the Mantel-Cox log-rank test) indicated a significant correlation between the loss of total and nuclear Pdcd4 in tumors and overall survival (P < .05 and P < .02, respectively) and disease-specific survival (P < .01 and P < .01, respectively). In multivariate analysis, loss of total or nuclear Pdcd4 was an independent predictor of disease-specific or overall survival. CONCLUSIONS To the authors' knowledge, this is the first study to demonstrate an independent prognostic impact of Pdcd4 and its expression pattern in colorectal cancer. Data from this study support the regulation of Pdcd4 localization by pAkt in vivo. Pdcd4 immunohistochemistry may be useful as a supportive diagnostic tool for the transition between normal, adenoma, and tumor tissues.
Collapse
Affiliation(s)
- Giridhar Mudduluru
- Department of Experimental Surgery Mannheim/Molecular Oncology of Solid Tumors, Deutsches Krebsforschungszentrum and University Heidelberg, Mannheim, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Gene expression profiling of liver metastases and tumour invasion in pancreatic cancer using an orthotopic SCID mouse model. Br J Cancer 2007; 97:1432-40. [PMID: 17940512 PMCID: PMC2360231 DOI: 10.1038/sj.bjc.6604031] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The prognosis of pancreatic adenocarcinoma is affected by early metastases and local tumour invasion beyond surgical margins. Gene expression profiling in pancreatic cancer tissue is complicated due to the high amount of RNAses being present in human tissue and that of suitable models. In order to demonstrate early metastases, the models should take into account the anatomical environment of the tumour. Using the orthotopic transplantation of pancreatic tumour cells in SCID (severe combined immunodeficiency) mice, these interactions are taken into consideration. In order to identify genes associated with local tumour invasion and metastases in ductal pancreatic cancer, we investigated a human pancreatic tumour cell line derived from an orthopic pancreatic tumour model in SCID mice. Differential gene expression was performed on the basis of microarray technique. The human MiaPaca-2 cell line was implanted orthotopically in SCID mice. Transcriptional profiling was performed on fresh frozen tissue derived from the primary tumour, the tumour invasion front and the liver metastases. Differentially expressed genes were identified using statistical analyses, and were validated with external databases and with immunohistochemistry. A total of 1066 of 14 500 genes were significantly differentially expressed. Comparing the primary tumour with the tumour invasion front, there were 614 statistically significant up- and 348 downregulated genes. Twenty-five statistically significant up- and 181 downregulated genes were identified comparing the liver metastases with the primary tumour. Eight genes (PAI-1, BNIP3l, VEGF, NSE, RGS4, HSP27, GADD45A, PTPN14) were chosen and validated in a semi-quantitative immunohistochemical analysis, which revealed a positive correlation to the array data. Overrepresentation analyses revealed a total of 66 significantly regulated pathways associated with cell proliferation, cell stress, cell communication metabolic and cytokine function. In conclusion, model marker genes for local invasion and liver metastases can be identified using transcriptional profiling in the SCID mouse. Overrepresentation analysis secures a good and fast overview about the significantly regulated genes and can assign genes to certain pathways. These marker genes can be related to the apoptotic cascade, angiogenesis and cell interaction.
Collapse
|
48
|
Moon EJ, Brizel DM, Chi JTA, Dewhirst MW. The potential role of intrinsic hypoxia markers as prognostic variables in cancer. Antioxid Redox Signal 2007; 9:1237-94. [PMID: 17571959 DOI: 10.1089/ars.2007.1623] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tumor hypoxia is related to tumor progression and therapy resistance, which leads to poor patient outcome. It has been suggested that measuring the hypoxic status of a tumor helps to predict patient outcome and to select more targeted treatment. However, current methods using needle electrodes or exogenous markers have limitations due to their invasiveness or necessity for preinjection. Recent studies showed that hypoxia-regulated genes could be alternatively used as endogenous hypoxia markers. This is a review of 15 hypoxia-regulated genes, including hypoxia-inducible factor-1 and its targets, and their correlation with tumor hypoxia and patient outcome from 213 studies. Though most of the studies showed significance of these genes in predicting prognosis, there was no definitive prognostic and hypoxia marker. In conclusion, this review suggests the need for further studies with standardized methods to examine gene expression, as well as the use of multiple gene expressions.
Collapse
Affiliation(s)
- Eui Jung Moon
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
49
|
Li ZW, Liu LY, Tian MM, You WC, Li JY. Expression of Sonic hedgehog gene, interleukin-1β, tumor necrosis factor-α in gastric epithelium of mongolian gerbils after H pylori inoculation. Shijie Huaren Xiaohua Zazhi 2007; 15:7-13. [DOI: 10.11569/wcjd.v15.i1.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of morphogene Sonic hedgehog (Shh), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α) in gastric epithelium of mongolian gerbils after H pylori inoculation and their correlations with the pathologic changes of Helicobacter pylori related gastritis.
METHODS: A total of 50 male mongolian gerbils were inoculated with H pylori and another 50 gerbils served as controls. The levels of Shh, IL-1β and TNF-α mRNA expression were detected by reverse transcription-polymerase chain reaction (RT-PCR), and the level of Shh protein expression was examined by immunohistochemistry 4, 12, 24, 36 and 48 weeks after inoculation.
RESULTS: As compared with those in the controls, both the expression levels of Shh mRNA and protein were decreased significantly at the 36th (t = 3.24, P < 0.05; Z = 4.84, P < 0.001) and 48th week (t = 3.01, P < 0.05; Z = 4.65, P < 0.001) after H pylori inoculation. Furthermore, the level of Shh mRNA expression was negatively correlated with the expression of IL-1β mRNA, TNF-α mRNA and the grade of gastritis, respectively (r = -0.372, P < 0.01; r = -0.301, P < 0.05; r = -0.397, P < 0.001). Similarly, the level of Shh protein expression was negatively correlated with the expression of IL-1β mRNA, TNF-α mRNA and the grade of gastritis (r = -0.321, P < 0.05; r = -0.313, P < 0.05; r = -0.371, P < 0.001) respectively.
CONCLUSION: After H pylori infection, the expression of IL-1β and TNF-α is increased and positively correlated with the grade of gastritis, and while the expression of Shh is decreased and negatively correlated with the expression of IL-1β, TNF-α and gastritis grades.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Although chronic Helicobacter pylori infection is the strongest known risk factor for development of gastric adenocarcinoma, only a small proportion of infected individuals will ever develop tumours. This article discusses various bacterial, host and environmental factors which may influence an individual's susceptibility. RECENT FINDINGS Recent research on bacterial virulence factors has focussed upon the cag pathogenicity island, particularly its roles in regulating epithelial growth and adhesion. Studies of host genetic factors have included several analyses of polymorphisms in inflammatory cytokines in human cohorts. Animal studies have recently clarified the roles of dysregulated epithelial apoptosis, proliferation and differentiation pathways during gastric carcinogenesis, and novel experiments involving H. felis infection of bone marrow transplanted irradiated mice have suggested that gastric cancer may originate from bone marrow-derived stem cells. Important roles for signalling between epithelial and mesenchymal cells, particularly myofibroblasts, are also emerging. Recent research on the importance of environmental factors has demonstrated how helminth coinfection may protect against atrophic gastritis and T helper type 1 responses. SUMMARY Complex interactions between several bacterial, host genetic and environmental factors determine whether H. pylori infected individuals develop gastric carcinoma. The importance of bone marrow stem cell engraftment during human gastric neoplasia is an area requiring urgent investigation.
Collapse
Affiliation(s)
- D Mark Pritchard
- Division of Gastroenterology, University of Liverpool, Liverpool, UK.
| | | |
Collapse
|