1
|
Yang Y, Yuan F, Zhou H, Quan J, Liu C, Wang Y, Xiao F, Liu Q, Liu J, Zhang Y, Yu X. Potential roles of heparanase in cancer therapy: Current trends and future direction. J Cell Physiol 2023; 238:896-917. [PMID: 36924082 DOI: 10.1002/jcp.30995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
Heparanase (HPSE; heparanase-1) is an endo-β-glucuronidase capable of degrading the carbohydrate moiety of heparan sulfate proteoglycans, thus modulating and facilitating the remodeling of the extracellular matrix and basement membrane. HPSE activity is strongly associated with major human pathological complications, including but not limited to tumor progress and angiogenesis. Several lines of literature have shown that overexpression of HPSE leads to enhanced tumor growth and metastatic transmission, as well as poor prognosis. Gene silencing of HPSE or treatment of tumor with compounds that block HPSE activity are shown to remarkably attenuate tumor progression. Therefore, targeting HPSE is considered as a potential therapeutical strategy for the treatment of cancer. Intriguingly, recent findings disclose that heparanase-2 (HPSE-2), a close homolog of HPSE but lacking enzymatic activity, can also regulate antitumor mechanisms. Given the pleiotropic roles of HPSE, further investigation is in demand to determine the precise mechanism of regulating action of HPSE in different cancer settings. In this review, we first summarize the current understanding of HPSE, such as its structure, subcellular localization, and tissue distribution. Furthermore, we systematically review the pro- and antitumorigenic roles and mechanisms of HPSE in cancer progress. In addition, we delineate HPSE inhibitors that have entered clinical trials and their therapeutic potential.
Collapse
Affiliation(s)
- Yiyuan Yang
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Fengyan Yuan
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Huiqin Zhou
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jing Quan
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Chongyang Liu
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Yi Wang
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Fen Xiao
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Qiao Liu
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jie Liu
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Yujing Zhang
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xing Yu
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| |
Collapse
|
2
|
Purushothaman A, Mohajeri M, Lele TP. The role of glycans in the mechanobiology of cancer. J Biol Chem 2023; 299:102935. [PMID: 36693448 PMCID: PMC9930169 DOI: 10.1016/j.jbc.2023.102935] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Although cancer is a genetic disease, physical changes such as stiffening of the extracellular matrix also commonly occur in cancer. Cancer cells sense and respond to extracellular matrix stiffening through the process of mechanotransduction. Cancer cell mechanotransduction can enhance cancer-promoting cell behaviors such as survival signaling, proliferation, and migration. Glycans, carbohydrate-based polymers, have recently emerged as important mediators and/or modulators of cancer cell mechanotransduction. Stiffer tumors are characterized by increased glycan content on cancer cells and their associated extracellular matrix. Here we review the role of cancer-associated glycans in coupled mechanical and biochemical alterations during cancer progression. We discuss the recent evidence on how increased expression of different glycans, in the form of glycoproteins and proteoglycans, contributes to both mechanical changes in tumors and corresponding cancer cell responses. We conclude with a summary of emerging tools that can be used to modify glycans for future studies in cancer mechanobiology.
Collapse
Affiliation(s)
- Anurag Purushothaman
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA.
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Tanmay P Lele
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA; Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA; Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA; Department of Translational Medical Sciences, Texas A&M University, Houston, Texas, USA.
| |
Collapse
|
3
|
Zhu Y, Cheung ALM. Proteoglycans and their functions in esophageal squamous cell carcinoma. World J Clin Oncol 2021; 12:507-521. [PMID: 34367925 PMCID: PMC8317653 DOI: 10.5306/wjco.v12.i7.507] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/13/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly malignant disease that has a poor prognosis. Its high lethality is mainly due to the lack of symptoms at early stages, which culminates in diagnosis at a late stage when the tumor has already metastasized. Unfortunately, the common cancer biomarkers have low sensitivity and specificity in esophageal cancer. Therefore, a better understanding of the molecular mechanisms underlying ESCC progression is needed to identify novel diagnostic markers and therapeutic targets for intervention. The invasion of cancer cells into the surrounding tissue is a crucial step for metastasis. During metastasis, tumor cells can interact with extracellular components and secrete proteolytic enzymes to remodel the surrounding tumor microenvironment. Proteoglycans are one of the major components of extracellular matrix. They are involved in multiple processes of cancer cell invasion and metastasis by interacting with soluble bioactive molecules, surrounding matrix, cell surface receptors, and enzymes. Apart from having diverse functions in tumor cells and their surrounding microenvironment, proteoglycans also have diagnostic and prognostic significance in cancer patients. However, the functional significance and underlying mechanisms of proteoglycans in ESCC are not well understood. This review summarizes the proteoglycans that have been studied in ESCC in order to provide a comprehensive view of the role of proteoglycans in the progression of this cancer type. A long term goal would be to exploit these molecules to provide new strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Yun Zhu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
4
|
Faria-Ramos I, Poças J, Marques C, Santos-Antunes J, Macedo G, Reis CA, Magalhães A. Heparan Sulfate Glycosaminoglycans: (Un)Expected Allies in Cancer Clinical Management. Biomolecules 2021; 11:136. [PMID: 33494442 PMCID: PMC7911160 DOI: 10.3390/biom11020136] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
In an era when cancer glycobiology research is exponentially growing, we are witnessing a progressive translation of the major scientific findings to the clinical practice with the overarching aim of improving cancer patients' management. Many mechanistic cell biology studies have demonstrated that heparan sulfate (HS) glycosaminoglycans are key molecules responsible for several molecular and biochemical processes, impacting extracellular matrix properties and cellular functions. HS can interact with a myriad of different ligands, and therefore, hold a pleiotropic role in regulating the activity of important cellular receptors and downstream signalling pathways. The aberrant expression of HS glycan chains in tumours determines main malignant features, such as cancer cell proliferation, angiogenesis, invasion and metastasis. In this review, we devote particular attention to HS biological activities, its expression profile and modulation in cancer. Moreover, we highlight HS clinical potential to improve both diagnosis and prognosis of cancer, either as HS-based biomarkers or as therapeutic targets.
Collapse
Affiliation(s)
- Isabel Faria-Ramos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Juliana Poças
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Catarina Marques
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - João Santos-Antunes
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
- Gastroenterology Department, Centro Hospitalar S. João, 4200-319 Porto, Portugal
| | - Guilherme Macedo
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
- Gastroenterology Department, Centro Hospitalar S. João, 4200-319 Porto, Portugal
| | - Celso A. Reis
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
| |
Collapse
|
5
|
Katakam SK, Pelucchi P, Cocola C, Reinbold R, Vlodavsky I, Greve B, Götte M. Syndecan-1-Dependent Regulation of Heparanase Affects Invasiveness, Stem Cell Properties, and Therapeutic Resistance of Caco2 Colon Cancer Cells. Front Oncol 2020; 10:774. [PMID: 32477959 PMCID: PMC7240066 DOI: 10.3389/fonc.2020.00774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022] Open
Abstract
The heparan sulfate proteoglycan Syndecan-1 binds cytokines, morphogens and extracellular matrix components, regulating cancer stem cell properties and invasiveness. Syndecan-1 is modulated by the heparan sulfate-degrading enzyme heparanase, but the underlying regulatory mechanisms are only poorly understood. In colon cancer pathogenesis, complex changes occur in the expression pattern of Syndecan-1 and heparanase during progression from well-differentiated to undifferentiated tumors. Loss of Syndecan-1 and increased expression of heparanase are associated with a change in phenotypic plasticity and an increase in invasiveness, metastasis and dedifferentiation. Here we investigated the regulatory and functional interplay of Syndecan-1 and heparanase employing siRNA-mediated silencing and plasmid-based overexpression approaches in the human colon cancer cell line Caco2. Heparanase expression and activity were upregulated in Syndecan-1 depleted cells. This increase was linked to an upregulation of the transcription factor Egr1, which regulates heparanase at the promoter level. Inhibitor experiments demonstrated an impact of focal adhesion kinase, Wnt and ROCK-dependent signaling on this process. siRNA-depletion of Syndecan-1, and upregulation of heparanase increased the colon cancer stem cell phenotype based on sphere formation assays and phenotypic marker analysis (Side-population, NANOG, KLF4, NOTCH, Wnt, and TCF4 expression). Syndecan-1 depletion increased invasiveness of Caco2 cells in vitro in a heparanase-dependent manner. Finally, upregulated expression of heparanase resulted in increased resistance to radiotherapy, whereas high expression of enzymatically inactive heparanase promoted chemoresistance to paclitaxel and cisplatin. Our findings provide a new avenue to target a stemness-associated signaling axis as a therapeutic strategy to reduce metastatic spread and cancer recurrence.
Collapse
Affiliation(s)
- Sampath Kumar Katakam
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Paride Pelucchi
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Cinzia Cocola
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Rolland Reinbold
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Israel Vlodavsky
- The Rappaport Faculty of Medicine, Technion Integrated Cancer Center (TICC), Haifa, Israel
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
6
|
Prevalence of Syndecan-1 (CD138) Expression in Different Kinds of Human Tumors and Normal Tissues. DISEASE MARKERS 2019; 2019:4928315. [PMID: 31976021 PMCID: PMC6954471 DOI: 10.1155/2019/4928315] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/07/2019] [Indexed: 12/28/2022]
Abstract
Syndecan-1 (CD138) is a transmembrane proteoglycan known to be expressed in various normal and malignant tissues. It is of interest because of a possible prognostic role of differential expression in tumors and its role as a target for indatuximab, a monoclonal antibody coupled with a cytotoxic agent. To comprehensively analyze CD138 in normal and neoplastic tissues, we used tissue microarrays (TMAs) for analyzing immunohistochemically detectable CD138 expression in 2,518 tissue samples from 85 different tumor entities and 76 different normal tissue types. The data showed that CD138 expression is abundant in tumors. At least an occasional weak CD138 immunostaining could be detected in 71 of 82 (87%) different tumor types, and 58 entities (71%) had at least one tumor with a strong positivity. In normal tissues, a particularly strong expression was found in normal squamous epithelium of various organs, goblet and columnar cells of the gastrointestinal tract, and in hepatocytes. The highly standardized analysis of most human cancer types resulted in a ranking order of tumors according to the frequency and levels of CD138 expression. CD138 immunostaining was highest in squamous cell carcinomas such as from the esophagus (100%), cervix uteri (79.5%), lung (85.7%), vagina (89.7%) or vulva (73.3%), and in invasive urothelial cancer (76.2%). In adenocarcinomas, CD138 was also high in lung (82.9%) and colorectal cancer (85.3%) but often lower in pancreas (73.3%), stomach (54.2% in intestinal type), or prostate carcinomas (16.3%). CD138 expression was usually low or absent in germ cell tumors, sarcomas, endocrine tumors including thyroid cancer, and neuroendocrine tumors. In summary, the preferential expression in squamous cell carcinomas of various sites makes these cancers prime targets for anti-CD138 treatments once these might become available. Abundant expression in many different normal tissues might pose obstacles to exploiting CD138 as a therapeutic target, however.
Collapse
|
7
|
Alaeddini M, Yazdani F, Etemad-Moghadam S. Stromal and epithelial syndecan-1 expression in benign and malignant salivary gland tumors: which is more reflective of behavior? Braz J Otorhinolaryngol 2019; 87:171-177. [PMID: 31540870 PMCID: PMC9422431 DOI: 10.1016/j.bjorl.2019.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/31/2019] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Salivary gland tumors are a diverse group of lesions, with various origins and extremely different behaviors, leading to a variety of outcomes for patients. Therefore, the need to discover novel markers with the ability to predict the behavior of benign and malignant salivary gland neoplasms is crucial. Syndecan-1 is a cell-surface protein with significant roles in various aspects of tumor function. Its expression in salivary gland neoplasms, especially their stromal component, has not been investigated. OBJECTIVES We aimed to assess the immunopositivity of syndecan-1 in epithelial and stromal components of salivary gland neoplasms and to compare it between benign and malignant subtypes in addition to evaluating its correlation with clinicopathologic parameters. METHODS 133 salivary gland tumors were immunohistochemically stained with syndecan-1 and the intensity and percentage of this protein was determined, compared between the tumors and correlated with clinicopathologic factors. RESULTS Statistical analysis of lesions with a sufficient sample size showed significant differences in percentage and intensity between both epithelial and stromal components of all tumors (p<0.05). Pairwise-comparisons demonstrated significantly higher staining-percentage of epithelial cells (p=0.02) in Warthin's tumor compared to pleomorphic adenoma and adenoid cystic carcinoma. Similarly, significantly higher staining intensities and/or percentages was observed in mucoepidermoid carcinoma and adenoid cystic carcinoma compared to pleomorphic adenoma and Warthin's tumor (p<0.05). Of the clinicopathologic factors, there was only a significant negative correlation between stromal percentage of mucoepidermoid carcinoma and age and a significant difference between stromal intensity+percentage of adenoid cystic carcinoma and gender (p<0.05). CONCLUSIONS According to our findings we postulate that stromal syndecan-1 correlates with the behavior of salivary gland tumors, with malignant neoplasms demonstrating a higher expression, indicating a role for syndecan-1 in invasion and metastasis.
Collapse
Affiliation(s)
- Mojgan Alaeddini
- Tehran University of Medical Sciences, Dentistry Research Institute, Dental Research Center, Tehran, Iran
| | - Farzad Yazdani
- Tehran University of Medical Sciences, Amiralam Hospital, Department of Pathology, Tehran, Iran
| | - Shahroo Etemad-Moghadam
- Tehran University of Medical Sciences, Dentistry Research Institute, Dental Research Center, Tehran, Iran.
| |
Collapse
|
8
|
Kang H, Wu Q, Sun A, Liu X, Fan Y, Deng X. Cancer Cell Glycocalyx and Its Significance in Cancer Progression. Int J Mol Sci 2018; 19:ijms19092484. [PMID: 30135409 PMCID: PMC6163906 DOI: 10.3390/ijms19092484] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/11/2018] [Accepted: 08/13/2018] [Indexed: 12/31/2022] Open
Abstract
Cancer is a malignant tumor that threatens the health of human beings, and has become the leading cause of death in urban and rural residents in China. The glycocalyx is a layer of multifunctional glycans that covers the surfaces of a variety of cells, including vascular endothelial cells, smooth muscle cells, stem cells, epithelial, osteocytes, as well as cancer cells. The glycosylation and syndecan of cancer cell glycocalyx are unique. However, heparan sulfate (HS), hyaluronic acid (HA), and syndecan are all closely associated with the processes of cancer progression, including cell migration and metastasis, tumor cell adhesion, tumorigenesis, and tumor growth. The possible underlying mechanisms may be the interruption of its barrier function, its radical role in growth factor storage, signaling, and mechanotransduction. In the later sections, we discuss glycocalyx targeting therapeutic approaches reported in animal and clinical experiments. The study concludes that cancer cells’ glycocalyx and its role in cancer progression are beginning to be known by more groups, and future studies should pay more attention to its mechanotransduction of interstitial flow-induced shear stress, seeking promising therapeutic targets with less toxicity but more specificity.
Collapse
Affiliation(s)
- Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 102402, China.
| | - Qiuhong Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 102402, China.
| | - Anqiang Sun
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 102402, China.
| | - Xiao Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 102402, China.
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 102402, China.
- National Research Center for Rehabilitation Technical Aids, Beijing 100176, China.
| | - Xiaoyan Deng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 102402, China.
| |
Collapse
|
9
|
Agere SA, Kim EY, Akhtar N, Ahmed S. Syndecans in chronic inflammatory and autoimmune diseases: Pathological insights and therapeutic opportunities. J Cell Physiol 2018; 233:6346-6358. [PMID: 29226950 DOI: 10.1002/jcp.26388] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
Syndecans (SDCs) are a family of heparan sulfate proteoglycans (HSPGs) glycoproteins ubiquitously expressed on the cell surfaces and extracellular matrix of all mammalian tissues. There are four mammalian syndecans, SDC-1 thorough 4, which play a critical role in cell adhesion, migration, proliferation, differentiation, and angiogenesis through independent and growth factor mediated signaling. An altered expression of SDCs is often observed in autoimmune disorders, cancer, HIV infection, and many other pathological conditions. SDCs modulate disease progression by interacting with a diverse array of ligands, receptors, and other proteins, including extracellular matrix, glycoproteins, integrins, morphogens, and various growth factors and chemokines, along with their receptors and kinases. Specifically, SDCs present on cell surface can bind directly to chemokines to enhance their binding to receptors, downstream signaling, and migration. Alternatively, SDCs can be cleaved and shed to mediate negative regulation of chemokine and growth factor signaling pathways and ligand sequestration. Importantly, SDC shedding may be a biomarker of inflammation, especially in chronic inflammatory diseases. While the current therapies for cancer and several autoimmune disorders have revolutionized treatment outcomes, understanding the pathophysiological role of SDCs and the use of HSPG mimetic or antagonists on cytokine signaling networks may uncover potentially novel targeted therapeutic approaches. This review mainly summarizes the current findings on the role of individual SDCs in disease processes, mechanisms through which SDCs mediate their biological functions, and the possibility of targeting SDCs as future potential therapeutic approaches.
Collapse
Affiliation(s)
- Solomon A Agere
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington
| | - Eugene Y Kim
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington
| | - Nahid Akhtar
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington
| | - Salahuddin Ahmed
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington.,Division of Rheumatology, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
10
|
Jin H, Zhou S, Yang S, Cao HM. Heparanase overexpression down-regulates syndecan-1 expression in a gallbladder carcinoma cell line. J Int Med Res 2017; 45:662-672. [PMID: 28351285 PMCID: PMC5536678 DOI: 10.1177/0300060517700323] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Objective To discuss the relevance of heparanase and syndecan-1 and regulation of the heparanase-syndecan1 axis in the invasiveness of gallbladder carcinoma cells. Methods 1. Generation of a gallbladder cancer cell line overexpressing a heparanase (GBD-SD) transgene. 2. Western blot analysis of syndecan-1 levels of GBD-SD and control gallbladder carcinoma (GBC-SD) cells. 3. RT-PCR analysis of syndecan-1 mRNA levels of GBD-SD and GBC-SD. 4. Evaluation of invasion and migration of GBD-SD and GBC-SD cells. Results 1. Heparanase expression in GBD-SD cells was significantly increased. 2. The syndecan-1 mRNA level of GBD-SD cells was significantly lower compared with that of GBC-SD cells. 3. The syndecan-1 DNA copy number in GBD-SD cells was significantly lower compared with that of GBC-SD. 4. The invasiveness and migration of GBD-SD cells were significantly higher compared with GBC-SD cells. Conclusions 1. The expression of heparanase negatively correlated with that of syndecan-1 in a gallbladder carcinoma cell line. 2. The expression of heparanase and syndecan-1 in gallbladder carcinomas negatively correlated, similar to other tumours. 3. The heparanase/syndecan1 axis in gallbladder carcinoma plays an important role in the invasion and metastasis, thus providing a new therapeutic target. 4. Further research is required to identify the detailed mechanisms.
Collapse
Affiliation(s)
- Hao Jin
- 1 Zhuhai People's Hospital, Zhuhai, China
| | | | - Song Yang
- 2 The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | | |
Collapse
|
11
|
Abstract
Heparanase is an endo-β-D-glucuronidase that is capable of cleaving heparan sulfate side chains of heparan sulfate proteoglycans on cell surfaces and the extracellular matrix. This activity is strongly implicated in tumor metastasis and angiogenesis. We have earlier demonstrated that apart of its well characterized enzymatic activity, heparanase may also affect the hemostatic system in a non-enzymatic manner. We showed that heparanase up-regulated the expression of the blood coagulation initiator-tissue factor (TF) and interacted with the tissue factor pathway inhibitor (TFPI) on the cell surface membrane of endothelial and tumor cells, leading to dissociation of TFPI and resulting in increased cell surface coagulation activity. Moreover, we demonstrated that heparanase directly enhanced TF activity, which led to increased factor Xa production and subsequent activation of the coagulation system. In patients with cancer, increased heparanase procoagulant activity appeared to be a potential predictor of survival. We have also shown that JAK-2 is involved in heparanase up-regulation via the erythropoietin receptor, a finding that may point to a new mechanism of thrombosis in JAK-2 positive patents with essential thrombocytosis. Recently, we found that the solvent accessible surface of TFPI-2 first Kunitz domain had a role in TF/heparanase complex inhibition. Peptides derived from TFPI-2 inhibitory site were shown to reduce coagulation activation induced by heparanase and to attenuate sepsis severity and tumor growth in a mouse model, without predisposing to significant bleeding tendency. These data imply that inhibition of heparanase procoagulant domain is potentially a good target for sepsis and cancer therapy.
Collapse
Affiliation(s)
- Yona Nadir
- Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus, Haifa, Israel.
| | - Benjamin Brenner
- Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
12
|
Farnedi A, Rossi S, Bertani N, Gulli M, Silini EM, Mucignat MT, Poli T, Sesenna E, Lanfranco D, Montebugnoli L, Leonardi E, Marchetti C, Cocchi R, Ambrosini-Spaltro A, Foschini MP, Perris R. Proteoglycan-based diversification of disease outcome in head and neck cancer patients identifies NG2/CSPG4 and syndecan-2 as unique relapse and overall survival predicting factors. BMC Cancer 2015; 15:352. [PMID: 25935541 PMCID: PMC4429505 DOI: 10.1186/s12885-015-1336-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/22/2015] [Indexed: 01/07/2023] Open
Abstract
Background Tumour relapse is recognized to be the prime fatal burden in patients affected by head and neck squamous cell carcinoma (HNSCC), but no discrete molecular trait has yet been identified to make reliable early predictions of tumour recurrence. Expression of cell surface proteoglycans (PGs) is frequently altered in carcinomas and several of them are gradually emerging as key prognostic factors. Methods A PG expression analysis at both mRNA and protein level, was pursued on primary lesions derived from 173 HNSCC patients from whom full clinical history and 2 years post-surgical follow-up was accessible. Gene and protein expression data were correlated with clinical traits and previously proposed tumour relapse markers to stratify high-risk patient subgroups. Results HNSCC lesions were indeed found to exhibit a widely aberrant PG expression pattern characterized by a variable expression of all PGs and a characteristic de novo transcription/translation of GPC2, GPC5 and NG2/CSPG4 respectively in 36%, 72% and 71% on 119 cases. Importantly, expression of NG2/CSPG4, on neoplastic cells and in the intralesional stroma (Hazard Ratio [HR], 6.76, p = 0.017) was strongly associated with loco-regional relapse, whereas stromal enrichment of SDC2 (HR, 7.652, p = 0.007) was independently tied to lymphnodal infiltration and disease-related death. Conversely, down-regulated SDC1 transcript (HR, 0.232, p = 0.013) uniquely correlated with formation of distant metastases. Altered expression of PGs significantly correlated with the above disease outcomes when either considered alone or in association with well-established predictors of poor prognosis (i.e. T classification, previous occurrence of precancerous lesions and lymphnodal metastasis). Combined alteration of all three PGs was found to be a reliable predictor of shorter survival. Conclusions An unprecedented PG-based prognostic portrait is unveiled that incisively diversifies disease course in HNSCC patients beyond the currently known clinical and molecular biomarkers. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1336-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Farnedi
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology, University of Bologna, Bellaria Hospital, Bologna, Italy.
| | - Silvia Rossi
- COMT - Centre for Molecular Translational Oncology & Department of Life Sciences, University of Parma, Parma, Italy.
| | - Nicoletta Bertani
- COMT - Centre for Molecular Translational Oncology & Department of Life Sciences, University of Parma, Parma, Italy.
| | - Mariolina Gulli
- Department of Life Sciences, Division of Genetics and Environmental Biotechnology, University of Parma, Parma, Italy.
| | - Enrico Maria Silini
- COMT - Centre for Molecular Translational Oncology & Department of Life Sciences, University of Parma, Parma, Italy. .,Department of Pathology and Laboratory Medicine, University of Parma, Parma, Italy.
| | - Maria Teresa Mucignat
- S.O.C. of Experimental Oncology 2, The National Tumour Institute Aviano - CRO-IRCCS, Aviano, Pordenone, Italy.
| | - Tito Poli
- Maxillofacial Surgery Section, Head and Neck Department, University of Parma, Parma, Italy.
| | - Enrico Sesenna
- Maxillofacial Surgery Section, Head and Neck Department, University of Parma, Parma, Italy.
| | - Davide Lanfranco
- Maxillofacial Surgery Section, Head and Neck Department, University of Parma, Parma, Italy.
| | - Lucio Montebugnoli
- Unit of Maxillo-Facial Surgery, Department of Oral Sciences, University of Bologna, Bellaria Hospital, Bologna, Italy.
| | - Elisa Leonardi
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology, University of Bologna, Bellaria Hospital, Bologna, Italy.
| | - Claudio Marchetti
- Department of Biomedical and Neuromotor Sciences, Unit of Maxillo-Facial Surgery, University of Bologna, S. Orsola Hospital, Bologna, Italy.
| | - Renato Cocchi
- Unit of Maxillo-facial Surgery at Bellaria Hospital, Bologna, Italy. .,Unit of Maxillo-facial Surgery, "Casa Sollievo della Sofferenza", San Giovanni in Rotondo, Italy.
| | - Andrea Ambrosini-Spaltro
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology, University of Bologna, Bellaria Hospital, Bologna, Italy.
| | - Maria Pia Foschini
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology, University of Bologna, Bellaria Hospital, Bologna, Italy.
| | - Roberto Perris
- COMT - Centre for Molecular Translational Oncology & Department of Life Sciences, University of Parma, Parma, Italy. .,S.O.C. of Experimental Oncology 2, The National Tumour Institute Aviano - CRO-IRCCS, Aviano, Pordenone, Italy.
| |
Collapse
|
13
|
Abstract
Heparanase is an endo-β-D-glucuronidase that is capable of cleaving heparan sulfate side chains of heparan sulfate proteoglycans on cell surfaces and the extracellular matrix, activity that is strongly implicated in tumor metastasis and angiogenesis. Apart of its well characterized enzymatic activity, heparanase was noted to exert also enzymatic-independent functions. Among these are the up-regulation of vascular endothelial growth factor (VEGF)-A, VEGF-C and activation of intra-cellular signaling involved in cell survival and proliferation. We had earlier demonstrated that heparanase may also affect the hemostatic system in a non-enzymatic manner. We had shown that heparanase up-regulated the expression of the blood coagulation initiator- tissue factor (TF) and interacted with the tissue factor pathway inhibitor (TFPI) on the cell surface membrane of endothelial and tumor cells, leading to dissociation of TFPI and resulting in increased cell surface coagulation activity. Moreover, we have demonstrated that heparanase directly enhanced TF activity which led to increased factor Xa production and subsequent activation of the coagulation system. Taking into account the prometastatic, pro-angiogenic and pro-coagulant functions of heparanase, over-expression in human malignancies and abundance in platelets, implies that heparanase is potentially a good target for cancer therapy.
Collapse
Affiliation(s)
- Yona Nadir
- Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus, Haifa, Israel.
| | - Benjamin Brenner
- Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
14
|
Simmons SC, Jämsä H, Silva D, Cortez CM, McKenzie EA, Bitu CC, Salo S, Nurmenniemi S, Nyberg P, Risteli J, deAlmeida CEB, Brenchley PEC, Salo T, Missailidis S. Anti-heparanase aptamers as potential diagnostic and therapeutic agents for oral cancer. PLoS One 2014; 9:e96846. [PMID: 25295847 PMCID: PMC4189786 DOI: 10.1371/journal.pone.0096846] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 04/11/2014] [Indexed: 12/13/2022] Open
Abstract
Heparanase is an endoglycosidase enzyme present in activated leucocytes, mast cells, placental tissue, neutrophils and macrophages, and is involved in tumour metastasis and tissue invasion. It presents a potential target for cancer therapies and various molecules have been developed in an attempt to inhibit the enzymatic action of heparanase. In an attempt to develop a novel therapeutic with an associated diagnostic assay, we have previously described high affinity aptamers selected against heparanase. In this work, we demonstrated that these anti-heparanase aptamers are capable of inhibiting tissue invasion of tumour cells associated with oral cancer and verified that such inhibition is due to inhibition of the enzyme and not due to other potentially cytotoxic effects of the aptamers. Furthermore, we have identified a short 30 bases aptamer as a potential candidate for further studies, as this showed a higher ability to inhibit tissue invasion than its longer counterpart, as well as a reduced potential for complex formation with other non-specific serum proteins. Finally, the aptamer was found to be stable and therefore suitable for use in human models, as it showed no degradation in the presence of human serum, making it a potential candidate for both diagnostic and therapeutic use.
Collapse
Affiliation(s)
- Suzanne C. Simmons
- Department of Chemistry and Analytical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Hannaleena Jämsä
- Department of Diagnostics and Oral Medicine, Institute of Dentistry, University of Oulu, Oulu, Finland
- Medical Research Center and Oulu University Hospital, Oulu, Finland
| | - Dilson Silva
- Institute of Mathematics and Statistics, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Celia M. Cortez
- Institute of Mathematics and Statistics, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Edward A. McKenzie
- Manchester Institute of Biotechnology, University of Manchester, Manchester, United Kingdom
| | - Carolina C. Bitu
- Department of Diagnostics and Oral Medicine, Institute of Dentistry, University of Oulu, Oulu, Finland
- Medical Research Center and Oulu University Hospital, Oulu, Finland
| | - Sirpa Salo
- Department of Diagnostics and Oral Medicine, Institute of Dentistry, University of Oulu, Oulu, Finland
| | - Sini Nurmenniemi
- Department of Diagnostics and Oral Medicine, Institute of Dentistry, University of Oulu, Oulu, Finland
| | - Pia Nyberg
- Department of Diagnostics and Oral Medicine, Institute of Dentistry, University of Oulu, Oulu, Finland
- Medical Research Center and Oulu University Hospital, Oulu, Finland
| | - Juha Risteli
- Institute of Diagnostics, Department of Clinical Chemistry, University of Oulu, Oulu, Finland
| | - Carlos E. B. deAlmeida
- Department of Chemistry and Analytical Sciences, The Open University, Milton Keynes, United Kingdom
- Laboratório de Radiobiologia, Instituto de Radioproteção e Dosimetria, Rio de Janeiro, Brazil
| | | | - Tuula Salo
- Department of Diagnostics and Oral Medicine, Institute of Dentistry, University of Oulu, Oulu, Finland
- Medical Research Center and Oulu University Hospital, Oulu, Finland
- Graduate Program in Estomatopatologia, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
- Institute of Dentistry, University of Helsinki, Helsinki, Finland
- * E-mail: (SM); (TS)
| | - Sotiris Missailidis
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (SM); (TS)
| |
Collapse
|
15
|
Abstract
Heparanase, a β-D-endoglucuronidase abundant in platelets that was discovered 30 years ago, is an enzyme that cleaves heparan sulfate side chains on the cell surface and in the extracellular matrix. It was later recognized as being a pro-inflammatory and pro-metastatic protein. We had earlier demonstrated that heparanase may also affect the hemostatic system in a non-enzymatic manner. We had shown that heparanase up-regulated the expression of the blood coagulation initiator tissue factor (TF) and interacted with the tissue factor pathway inhibitor (TFPI) on the cell surface membrane of endothelial and tumor cells, leading to dissociation of TFPI and resulting in increased cell surface coagulation activity. Moreover, we have demonstrated that heparanase directly enhanced TF activity which led to increased factor Xa production and subsequent activation of the coagulation system. Recently, heparanase inhibitory peptides derived of TFPI-2 were demonstrated by us to inhibit heparanase procoagulant activity and attenuate sepsis in mouse models.
Collapse
Affiliation(s)
- Yona Nadir
- Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
16
|
Boeddeker SJ, Baston-Buest DM, Altergot-Ahmad O, Kruessel JS, Hess AP. Syndecan-1 knockdown in endometrial epithelial cells alters their apoptotic protein profile and enhances the inducibility of apoptosis. Mol Hum Reprod 2014; 20:567-78. [DOI: 10.1093/molehr/gau009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
17
|
Huining L, Yi Z, Dihong T, Yifeng P, Man X, Ting Y, Jingting C. Inhibition of choriocarcinoma by Fe3O4-dextran-anti-β-human chorionic gonadotropin nanoparticles containing antisense oligodeoxynucleotide of heparanase. Int J Nanomedicine 2013; 8:4371-8. [PMID: 24235832 PMCID: PMC3826771 DOI: 10.2147/ijn.s44739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To observe the influence of Fe3O4-dextran-anti-β-human chorionic gonadotropin (HCG) carrying heparanase (Hpa) antisense oligodeoxynucleotide (ASODN), via the invasion, proliferation, and Hpa expression of JEG-3 cell lines and inhibitory effect of transplanted choriocarcinoma tumor growth. METHODS The different abilities of invasion and proliferation between transfected JEG-3 and untransfected JEG-3 were measured by Matrigel invasion assay and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay in vitro. The effect of Hpa ASODN transfection on the expression of Hpa mRNA and protein was measured by reverse-transcription polymerase chain reaction and Western blot. The transplanted choriocarcinoma tumors were taken out to calculate the inhibitory effect on tumor growth of Hpa ASODN. RESULTS IN THIS STUDY, WE FOUND THAT: (1) the invasive ability of JEG-3 cells was inhibited sufficiently (P < 0.05) after JEG-3 cells were transfected by Fe3O4-dextran-anti-βHCG carrying Hpa ASODN; (2) after JEG-3 cells were transfected by Fe3O4-dextran-anti-βHCG carrying Hpa ASODN at 48 and 72 hours, the proliferative ability of JEG-3 cells was inhibited sufficiently (P < 0.05); (3) the expression of Hpa mRNA and protein in JEG-3 cells was inhibited efficiently after JEG-3 cells were transfected by Fe3O4-dextran-anti-βHCG carrying Hpa ASODN (P < 0.05); and (4) Fe3O4-dextran-anti-βHCG carrying Hpa ASODN had an inhibitory effect on the transplanted choriocarcinoma tumor growth (P < 0.05) and was harmless on nude mice. CONCLUSION Fe3O4-dextran-anti-βHCG carrying Hpa ASODN weakened the invasive and proliferative ability of choriocarcinoma, with a significant inhibitory effect on the transplanted choriocarcinoma tumor. Therefore, Fe3O4-dextran-anti-βHCG carrying Hpa ASODN is an effective gene therapy, and Fe3O4-dextran-anti-βHCG nanoparticles are a harmless and effective gene vector.
Collapse
Affiliation(s)
- Liu Huining
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
18
|
Nguyen TL, Grizzle WE, Zhang K, Hameed O, Siegal GP, Wei S. Syndecan-1 overexpression is associated with nonluminal subtypes and poor prognosis in advanced breast cancer. Am J Clin Pathol 2013; 140:468-74. [PMID: 24045542 DOI: 10.1309/ajcpz1d8calhdxcj] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Syndecan-1 expression is decreased in diverse tumor types but remains controversial in breast carcinomas. The goal of the study was to examine syndecan-1 expression in breast carcinoma and its prognostic significance. METHODS The epithelial expression of syndecan-1 was examined in tissue microarrays constructed from 62 consecutive breast carcinoma cases diagnosed between 1997 and 2004 with distant organ metastasis and 10 consecutive control cases (breast carcinoma with no distant metastasis after at least 8 years of follow-up). The prognostic significance of syndecan-1 was estimated by utilizing a Cox proportional hazards regression model. RESULTS Among tumors with distant metastasis, syndecan-1 expression was significantly associated with a higher histologic grade and inversely related to hormonal receptor status. The HER2 subtype and triple-negative carcinomas exhibited markedly higher syndecan-1 levels than those of luminal subtypes, while the latter remained significantly higher than nonmetastatic control cases. Furthermore, high syndecan-1 expression had a negative impact on both overall and disease-free survival rates. CONCLUSIONS These findings suggest that syndecan-1 may regulate breast cancer cell behavior and thus deserves further investigation to ascertain its potential as a therapeutic target, especially in metastatic, triple-negative carcinomas.
Collapse
Affiliation(s)
- Thuy L. Nguyen
- Department of Pathology, School of Medicine, University of Alabama at Birmingham
| | - William E. Grizzle
- Department of Pathology, School of Medicine, University of Alabama at Birmingham
| | - Kui Zhang
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham
| | - Omar Hameed
- Department of Pathology, School of Medicine, University of Alabama at Birmingham
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Gene P. Siegal
- Department of Pathology, School of Medicine, University of Alabama at Birmingham
| | - Shi Wei
- Department of Pathology, School of Medicine, University of Alabama at Birmingham
| |
Collapse
|
19
|
Mishra M, Chandavarkar V, Naik VV, Kale AD. An immunohistochemical study of basement membrane heparan sulfate proteoglycan (perlecan) in oral epithelial dysplasia and squamous cell carcinoma. J Oral Maxillofac Pathol 2013; 17:31-5. [PMID: 23798826 PMCID: PMC3687184 DOI: 10.4103/0973-029x.110704] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Basement membrane heparan sulfate proteoglycan (perlecan) has been demonstrated in precancer lesions and carcinomas of oral cavity. It helps in malignant transformation of epithelial cells. The aim of our study was to understand the immuno-localization of perlecan in oral dysplastic epithelium and oral carcinomas. MATERIALS AND METHODS A total of 50 cases comprising 10 normal mucosa, 20 dysplastic mucosa, and 20 oral squamous cell carcinomas (OSCC) were included in the retrospective study. They were examined for the presence of perlecan protein core by immunohistochemistry using monoclonal antibody. Interpretation of the pattern of staining was done, and majority of the observations were taken for statistical analysis. RESULTS In normal epithelium, perlecan was found to be present in basal layer at the cell border. In dysplastic epithelium, it was present in suprabasal layers also. With the increase in severity of dysplasia, its expression was more in suprabasal layers, and the immuno-localization was found to be at cell border and cytoplasm. In OSCC cases, perlecan was present in stroma and tumor islands. CONCLUSION It was deduced from the above results that perlecan helps potentially in dysplastic changes of epithelial cells. It gets accumulated within the cell and intercellular spaces and serves as a reservoir for various growth factors. In OSCC, it breaks down and releases growth factors, which help in tumor progression, angiogenesis, and metastasis of the carcinoma.
Collapse
Affiliation(s)
- Mithilesh Mishra
- Department of Oral Pathology, ITS Dental College and Hospital, Greater Noida, Delhi-NCR, India
| | | | | | | |
Collapse
|
20
|
Batista LTEA, Matos LL, Machado LR, Suarez ER, Theodoro TR, Martins JRM, Nader HB, Pompeo ACL, Pinhal MADS. Heparanase expression and glycosaminoglycans profile in renal cell carcinoma. Int J Urol 2012; 19:1036-40. [PMID: 22738382 DOI: 10.1111/j.1442-2042.2012.03086.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A better understanding of the molecular mechanisms of renal cell carcinogenesis could contribute to a decrease in the mortality rate of this disease. The aim of this study was to evaluate the glycosaminoglycans profile and heparanase expression in renal cell carcinoma. The study included 24 patients submitted to nephrectomy with confirmed pathological diagnosis of renal cell carcinoma. The majority of the samples (87.5%) were classified in the initial stage of renal cell carcinoma (clinical stages I and II). Heparanase messenger ribonucleic acid expression was evaluated by quantitative real-time reverse transcription polymerase chain reaction, and sulfated glycosaminoglycans were identified and quantified by agarose gel electrophoresis of renal cell carcinoma samples or non-neoplastic tissues obtained from the same patients (control group). The sulfated glycosaminoglycans and hyaluronic acid were analyzed in urine samples of the patients before and after surgery. The data showed a significant statistical increase in chondroitin sulfate, and a decrease in heparan sulfate and dermatan sulfate present in neoplastic tissues compared with non-neoplastic tissues. Higher heparanase messenger ribonucleic acid expression in the neoplastic tissues was also shown, compared with the non-neoplastic tissues. The urine glycosaminoglycans profile showed no significant difference between renal cell carcinoma and control samples. Extracellular matrix changes observed in the present study clarify that heparanase is possibly involved with heparan sulfate turnover, and that heparanase and the glycosaminoglycans can modulate initial events of renal cell carcinoma development.
Collapse
|
21
|
Yuan L, Hu J, Luo Y, Liu Q, Li T, Parish CR, Freeman C, Zhu X, Ma W, Hu X, Yu H, Tang S. Upregulation of heparanase in high-glucose-treated endothelial cells promotes endothelial cell migration and proliferation and correlates with Akt and extracellular-signal-regulated kinase phosphorylation. Mol Vis 2012; 18:1684-95. [PMID: 22773906 PMCID: PMC3388982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 06/17/2012] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The objectives of this study were to determine whether high-glucose-induced upregulation of heparanase (HPSE) expression and differential heparanase expression in human retinal vascular endothelial cells (HRECs) can alter HREC migration and proliferation. We also aimed to determine whether HREC migration and proliferation correlate with the levels of protein kinase B (Akt) and extracellular-signal-regulated kinase (ERK) phosphorylation and activation. METHODS HRECs were treated with either 5 mM glucose (Glu5) or high (30 mM) glucose (Glu30) for 48 h. Untransfected HRECs were grown in human endothelial serum-free medium (HE-SFM) in the presence of 5 mM glucose and supplemented with 30 mM mannitol for 48 h as an osmotic control (mannitol). HRECs were also infected with a heparanase small interfering RNA recombinant lentiviral vector (HPSE-LV) or a control vector (Con-LV) at a multiplicity of infection (MOI) of 60 for three days. Then the con-LV and HPSE-LV-infected cells were treated with 30 mM glucose for 48 h (Con-LV-Glu30 and HSPE-LV-Glu30, respectively). The expression levels of heparanase mRNA and protein and HREC proliferation and migration were examined using quantitative real-time polymerase chain reaction (qRT-PCR), western blot analysis, 3-(4,5-dimethylthiahiazol-2-y1)-2,5-diphenyltetrazolium bromide assay, bromodeoxyuridine histochemical staining, and the Boyden chamber assay. The expression level of paxillin was examined using immunofluorescent staining. Akt and ERK phosphorylation was evaluated using western blot analysis. RESULTS We successfully transfected the HPSE RNAi lentiviral vector into HRECs and demonstrated that it can suppress the expression of the heparanase gene in these cells. Western blot and qRT-PCR analyses showed that HRECs treated with a high concentration of glucose exhibited increased heparanase protein and mRNA levels, while the levels were decreased in HRECs that had been infected with HPSE-LV before treatment with high glucose (HPSE-LV-Glu30; p<0.05). The observed increase or decrease in the levels of heparanase correlated with increased or decreased HREC migration and proliferation, respectively (p<0.05). HREC proliferation and migration were found to correlate with Akt and ERK phosphorylation levels (p<0.5). CONCLUSIONS Our results indicate that heparanase plays a significant role in mediating retinal vascular endothelial cell proliferation and migration after the HRECs are exposed to high levels of glucose. Signaling inducing heparanase-stimulated HREC proliferation and migration appears to be related to the activation of Akt and ERK via their phosphorylation.
Collapse
Affiliation(s)
- Ling Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China,The First Affilliated Hospital of Kunming Medical College, Kunming, Yunnan, China
| | - Jie Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - QingYun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - Tao Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - Christopher R. Parish
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Craig Freeman
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - XiaoBo Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - Wei Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - XuTing Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - HongHua Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| | - ShiBo Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic center of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
Development of novel single-stranded nucleic acid aptamers against the pro-angiogenic and metastatic enzyme heparanase (HPSE1). PLoS One 2012; 7:e37938. [PMID: 22719856 PMCID: PMC3376095 DOI: 10.1371/journal.pone.0037938] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 04/26/2012] [Indexed: 01/07/2023] Open
Abstract
Heparanase is an enzyme involved in extracellular matrix remodelling and heparan sulphate proteoglycan catabolism. It is secreted by metastatic tumour cells, allowing them to penetrate the endothelial cell layer and basement membrane to invade target organs. The release of growth factors at the site of cleaved heparan sulphate chains further enhance the potential of the tumour by encouraging the process of angiogenesis. This leads to increased survival and further proliferation of the tumour. Aptamers are single or double stranded oligonucleotides that recognise specific small molecules, peptides, proteins, or even cells or tissues and have shown great potential over the years as diagnostic and therapeutic agents in anticancer treatment. For the first time, single stranded DNA aptamers were successfully generated against the active heterodimer form of heparanase using a modified SELEX protocol, and eluted based on increasing affinity for the target. Sandwich ELISA assays showed recognition of heparanase by the aptamers at a site distinct from that of a polyclonal HPSE1 antibody. The binding affinities of aptamer to immobilised enzyme were high (7×107 to 8×107 M−1) as measured by fluorescence spectroscopy. Immunohistochemistry and immunofluorescence studies demonstrated that the aptamers were able to recognise heparanase with staining comparable or in some cases superior to that of the HPSE1 antibody control. Finally, matrigel assay demonstrated that aptamers were able to inhibit heparanase. This study provides clear proof of principle concept that nucleic acid aptamers can be generated against heparanase. These reagents may serve as useful tools to explore the functional role of the enzyme and in the future development of diagnostic assays or therapeutic reagents.
Collapse
|
23
|
Abstract
Heparanase that was cloned from and is abundant in the placenta is implicated in cell invasion, tumor metastasis, and angiogenesis. Recently we have demonstrated that heparanase may also affect the hemostatic system in a non-enzymatic manner. Heparanase was shown to up-regulate tissue factor (TF) expression and interact with tissue factor pathway inhibitor (TFPI) on the cell surface, leading to dissociation of TFPI from the cell membrane of endothelial and tumor cells, resulting in increased cell surface coagulation activity. More recently, we have shown that heparanase directly enhances TF activity, resulting in increased factor Xa production and activation of the coagulation system. Data indicate increased levels and possible involvement of heparanase in vascular complications in pregnancy. Taking into account the prometastatic and proangiogenic functions of heparanase, overexpression in human malignancies, and abundance in platelets and placenta, its involvement in the coagulation machinery is an intriguing novel arena for further research.
Collapse
Affiliation(s)
- Yona Nadir
- To whom correspondence should be addressed. E-mail:
| | | |
Collapse
|
24
|
Sun H, Hu Y, Gu Z, Owens RT, Chen YQ, Edwards IJ. Omega-3 fatty acids induce apoptosis in human breast cancer cells and mouse mammary tissue through syndecan-1 inhibition of the MEK-Erk pathway. Carcinogenesis 2011; 32:1518-24. [PMID: 21771724 DOI: 10.1093/carcin/bgr132] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human epidemiological studies have shown that diets enriched in n-3 polyunsaturated fatty acids (n-3 PUFA) are associated with a lower incidence of cancers including breast cancer. Our previous studies showed that the n-3 PUFA, docosahexaenoic acid (DHA), upregulated syndecan-1 (SDC-1) expression to induce apoptosis in the human breast cancer cell line MCF-7. We now present evidence of a signaling pathway that is impacted by SDC-1 in these cells and in mouse mammary tissues to result in apoptosis. In MCF-7 cells and SK-BR-3 cells, DHA and a SDC-1 ectodomain impaired signaling of the p44/42 mitogen-activated protein kinase (MAPK) pathway by inhibiting the phosphorylation of MAPK/Erk (MEK)/extracellular signal-regulated kinase (Erk) and Bad to induce apoptosis. SDC-1 siRNA significantly enhanced phosphorylation of these signal molecules and blocked the inhibitory effects of DHA on their phosphorylation. SDC-1 siRNA diminished apoptosis of MCF-7 cells, an effect that was markedly blocked by MEK inhibitor, PD98059. In vivo studies used (i) Fat-1 mice, a genetic model able to convert n-6 to n-3 PUFA to result in higher SDC-1 levels in Fat-1 mammary tissue compared with that of wild-type (wt) mice. Phosphorylation of MEK, Erk and Bad was lower in the Fat-1 versus wt tissue and (ii) SDC-1(-/-) mice that demonstrated markedly higher levels of phosphorylated MEK, Erk and Bad in mammary gland tissue compared with those of SDC(+/+) mice. These data elucidate a pathway whereby SDC-1, upregulated by DHA, induces apoptosis in breast cancer cells through inhibition of MEK/Erk/Bad signaling.
Collapse
Affiliation(s)
- Haiguo Sun
- Department of Pathology, Wake ForestUniversity School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
25
|
Sun H, Hu Y, Gu Z, Wilson MD, Chen YQ, Rudel LL, Willingham MC, Edwards IJ. Endogenous synthesis of n-3 polyunsaturated fatty acids in Fat-1 mice is associated with increased mammary gland and liver syndecan-1. PLoS One 2011; 6:e20502. [PMID: 21655218 PMCID: PMC3105073 DOI: 10.1371/journal.pone.0020502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 05/02/2011] [Indexed: 01/12/2023] Open
Abstract
Long chain n-3 PUFA have been shown to have chemopreventive properties against breast cancer through various mechanisms. One pathway, studied in human breast cancer cell lines, involves upregulation of the proteoglycan, syndecan-1 (SDC-1) by n-3 PUFA-enriched LDL. Using Fat-1 mice that are able to convert n-6 to n-3 PUFA, we tested whether SDC-1 level in vivo is elevated in mammary glands due to endogenously synthesized rather than LDL-derived n-3 PUFA. Female Fat-1 and wild type (wt) mice were fed an n-6 PUFA- enriched diet for 7 weeks. Fatty acid analysis of plasma lipoproteins showed that total n-6 PUFA reflected dietary intake similarly in both genotypes (VLDL, 36.2±2.2 and 40.9±3.9; LDL, 49.0±3.3 and 48.1±2.0; HDL, 54.6±1.2 and 58.2±1.3, mean ± SEM percent of total fatty acids for Fat-1 and wt animals respectively). Lipoprotein percent n-3 PUFA was also similar between groups. However, phospholipids and triglycerides extracted from mammary and liver tissues demonstrated significantly higher n-3 PUFA and a corresponding decrease in the ratio n-6/n-3 PUFA in Fat-1 compared to wt mice. This was accompanied by higher SDC-1 in mammary glands and livers of Fat-1 mice, thus demonstrating that endogenously synthesized n-3 PUFA may upregulate SDC-1 in the presence of high dietary n-6 PUFA.
Collapse
Affiliation(s)
- Haiguo Sun
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Yunping Hu
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Zhennan Gu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Martha D. Wilson
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Yong Q. Chen
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Lawrence L. Rudel
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Mark C. Willingham
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Iris J. Edwards
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail: E-mail:
| |
Collapse
|
26
|
Sonoda R, Naomoto Y, Shirakawa Y, Fujiwara Y, Yamatsuji T, Noma K, Tanabe S, Takaoka M, Gunduz M, Tsujigiwa H, Nagatsuka H, Ohara N, Yoshino T, Takubo K, Vieth M, Tanaka N. Preferential up-regulation of heparanase and cyclooxygenase-2 in carcinogenesis of Barrett's oesophagus and intestinal-type gastric carcinoma. Histopathology 2010; 57:90-100. [PMID: 20653782 DOI: 10.1111/j.1365-2559.2010.03594.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Metaplastic changes secondary to chronic inflammation at the gastro-oesophageal junction and at the pyloric antrum are recognized as the premalignant conditions of Barrett's oesophageal adenocarcinoma and intestinal-type gastric carcinoma (GC), respectively. Heparanase (HPSE) and cyclooxygenase (COX)-2 have been proved to play critical roles in inflammation as well as in cancer. The aim was to examine the meaning of their expression in inflammation-related carcinogenesis. METHODS AND RESULTS First, expression of HPSE and COX-2 in 78 clinical tissues of Barrett's oesophagus was examined by immunohistochemistry and in situ hybridization. Their expression was increased during the metaplasia-dysplasia sequence with increased neovascularization. Successively, their expression in Barrett's dysplasia was compared with that of GC (22 cases of diffuse-type and 10 of intestinal-type). Interestingly, the expression pattern in Barrett's dysplasia was similar to that in intestinal-type GC, which mainly arises from chronic inflammation. Furthermore, cultured cell lines isolated from differentiated GC tissues, which are often found to be of intestinal-type, revealed up-regulated mRNA expression of HPSE and COX-2. CONCLUSIONS HPSE and COX-2 are preferentially up-regulated in Barrett's oesophagus and intestinal-type GC. These molecules may play an important role during the development of inflammation-related adenocarcinoma of the upper gastrointestinal tract.
Collapse
Affiliation(s)
- Ryotaro Sonoda
- Department of Gastroenterological Surgery, Transplant, and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang X, Zhou K, Huang L, Yan Y. Induction of anti-tumor immunity by dendritic cells pulsed with an endoplasmic reticulum retrieval signal modifies heparanase epitope in mice. Cytotherapy 2010; 12:735-42. [DOI: 10.3109/14653241003615156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
28
|
Abstract
Heparanase is an endo-beta-D-glucuronidase capable of cleaving heparan sulfate (HS) side chains of heparan sulfate proteoglycans (HSPG) on cell surfaces and the extracellular matrix, activity that is strongly implicated in tumor metastasis and angiogenesis. Evidence was provided that heparanase over-expression in cancer cells results in a marked increase in tissue factor (TF) levels. Likewise, TF was induced by exogenous addition of recombinant heparanase to tumor cells and primary endothelial cells, induction that was mediated by p38 phosphorylation and correlated with enhanced procoagulant activity. TF induction was further confirmed in heparanase over-expressing transgenic mice and correlated with heparanase expression levels in leukemia patients. Heparanase was also found to be involved in the regulation of tissue factor pathway inhibitor (TFPI). A physical interaction between heparanase and TFPI was demonstrated, suggesting a mechanism by which secreted heparanase interacts with TFPI on the cell surface, leading to dissociation of TFPI from the cell membrane and increased coagulation activity, thus further supporting the local pro-thrombotic function of heparanase. Data indicate a possible involvement of heparanase in early miscarriages and point to a regulatory effect on TFPI and TFPI-2 in trophoblasts. As heparins are strong inhibitor of heparanase, in view of the effect of heparanase on TF, the role of heparins anticoagulant-activity may potentially be expanded. Taking into account the pro-metastatic and pro-angiogenic functions of heparanase, its over-expression in human malignancies and abundance in platelets, its involvement in the coagulation machinery is an intriguing novel arena for further research.
Collapse
|
29
|
Expression of syndecan-1 and cathepsins D and K in advanced esophageal squamous cell carcinoma. Folia Histochem Cytobiol 2010; 47:571-8. [PMID: 20430722 DOI: 10.2478/v10042-008-0012-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The key features of malignant neoplasms are their local invasiveness and metastatic potential. Syndecan-1 - integral membrane heparan sulfate proteoglycan and cathepsins D and K - lysosomal proteases are important factors influencing different aspects of these processes. The study was undertaken to determine their expression in esophageal squamous cell carcinoma, and analyze relationship to selected clinicopathological features as well as to survival. Formalin-fixed, paraffin-embedded sections from 39 advanced esophageal squamous cell carcinoma were used for immunohistochemical staining. The epithelial and stromal staining were evaluated separately and compared to conventional clinicopathological features and one-year survival. Positive epithelial immunostaining for syndecan-1, cathepsin D and K were observed in 82.05%, 56.41% and 30.77% of tumors, respectively. However, stromal staining was noted in 51.28%, 51.28% and 46.15% ones, respectively. Epithelial syndecan-1-positive cases were significantly more frequent in well- and moderately differentiated carcinomas. Stromal cathepsin D expression predominated in tumors with infiltrative growth pattern. However, there were no statistically significant differences between any marker-positive and -negative groups with respect to other clinicopathological features studied. The only factors significantly influencing one-year survival were epithelial cathepsin D staining and distant metastasis. In a group of patients who survived one year post surgery, the percentage of cases with negative epithelial cathepsin D staining and without features of distant metastasis were higher. The results may suggest a relationship between syndecan-1 and cathepsins D and K with growth and invasiveness of esophageal squamous cell carcinoma, but such thesis requires further study on a larger and more heterogeneous population.
Collapse
|
30
|
Kim H, Choi DS, Chang SJ, Han JH, Min CK, Chang KH, Ryu HS. The expression of syndecan-1 is related to the risk of endometrial hyperplasia progressing to endometrial carcinoma. J Gynecol Oncol 2010; 21:50-5. [PMID: 20379448 DOI: 10.3802/jgo.2010.21.1.50] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 01/17/2010] [Accepted: 01/18/2010] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Aberrant expression of the cell surface proteoglycan, syndecan-1, is found in many malignancies. The current study describes the immunohistochemical study of syndecan-1 expression in normal, hyperplastic, and malignant endometrial tissues for evaluation of application as a parameter of cancer progression in patients with endometrial hyperplasia. METHODS Immunohistochemical staining of syndecan-1 was performed in 101 formalin fixed, paraffin embedded sections of normal, hyperplastic, and malignant endometrial tissues. We analyzed specimens from patients with normal endometrium (NE, N=10) as controls, and those of simple hyperplasia (SH, N=20), complex hyperplasia without atypia (CH, N=20), atypical hyperplasia (AH, N=20), and endometrial cancer (EC, N=31). RESULTS The mean rank of expression scores based on the frequency of syndecan-1 staining were 31.6, 20.5, 52.9, 72.1, and 62.1 for NE, SH, CH, AH and EC, respectively (p<0.001). Syndecan-1 expression was significantly greater in CH (p<0.001) or AH (p<0.001) than in SH, and significantly greater in AH compared to CH (p=0.028). Syndecan-1 is more frequently expressed in CH (p=0.042), AH (p<0.001), or EC (p=0.002) than in NE. Syndecan-1 expression did not differ significantly between NE and SH (p=0.248). CONCLUSION Syndecan-1 expression appears to be useful as a predictive indicator in endometrial hyperplasia.
Collapse
Affiliation(s)
- Hyunjin Kim
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Varchalama E, Rodolakis A, Strati A, Papageorgiou T, Valavanis C, Vorgias G, Lianidou E, Antsaklis A. Quantitative analysis of heparanase gene expression in normal cervical, cervical intraepithelial neoplastic, and cervical carcinoma tissues. Int J Gynecol Cancer 2010; 19:1614-9. [PMID: 19955948 DOI: 10.1111/igc.0b013e3181ae3f40] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Heparanase is an endoglycosidase that specifically cleaves heparan sulfate side chains of heparan sulfate proteoglycans, the major proteoglycans in the extracellular matrix and cell surfaces. Traditionally, heparanase activity was implicated in cellular invasion associated with angiogenesis, inflammation, and cancer metastasis. More recently, heparanase up-regulation was documented in an increasing number of primary human tumors. Iotan this study, we sought to investigate the expression of heparanase messenger RNA (mRNA) in normal cervical tissue and intraepithelial cervical lesion and its clinicopathologic importance in invasive cervical cancer. Gene expression of heparanase was assessed by quantitative real-time reverse transcriptase polymerase chain reaction in 28 normal cervical, 26 intraepithelial neoplastic, and 48 cervical cancer tissue samples. Heparanase mRNA expression was different between the 3 groups and lower in normal cervical specimens in relationship with intraepithelial cervical lesions and invasive cervical cancer tissue samples (P = 0.048). Gradually increasing expression of heparanase was evident as the cells progressed from low-grade to high-grade squamous intraepithelial lesions (P = 0.002). In invasive cervical cancer cases, there was a direct correlation between heparanase expression and tumor size (P = 0.002). In cases treated with radical hysterectomy and pelvic lymphadenectomy, the heparanase mRNA expression was significantly higher in tumors exhibiting lymph vascular space invasion (P = 0.044) and in cases with big tumor size (P = 0.005). In our study, we did not find any significant correlation between disease-free and overall survival rates and expression of heparanase (P = 0.396 and P = 0.712, respectively). The results of this study suggest that the gene expression of heparanase in cervical cancer enhances growth, invasion, and angiogenesis of the tumor and may have therapeutic applications.
Collapse
Affiliation(s)
- Eugene Varchalama
- 1st Department of Obstetrics and Gynecology, University of Athens, Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Peterson SB, Liu J. Unraveling the specificity of heparanase utilizing synthetic substrates. J Biol Chem 2010; 285:14504-13. [PMID: 20181948 DOI: 10.1074/jbc.m110.104166] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparanase is a promising anticancer target because of its involvement in cancer invasion and metastasis. Heparanase cleaves heparan sulfate (HS), a sulfated polysaccharide, and activates a series of HS-mediated cell proliferation and angiogenesis processes. Understanding the substrate specificity of heparanase will aid the discovery of heparanase inhibitors. Here, we sought to determine the specificity of heparanase using synthetic polysaccharide substrates. The substrates were prepared using purified HS biosynthetic enzymes. Using these substrates, we were able to dissect the structural moieties required for heparanase. Our data suggest that heparanase cleaves the linkage between a GlcA unit and an N-sulfo glucosamine unit carrying either a 3-O-sulfo or a 6-O-sulfo group. In addition, heparanase cleaves the linkage of a GlcA unit and N-sulfo glucosamine unit with a 2-O-sulfated GlcA residue, not a 2-O-sulfated IdoA residue, in proximity. We also discovered that the polysaccharide with repeating disaccharide units of IdoA2S-GlcNS inhibits the activity of heparanase. Our findings advance the understanding of the substrate specificity of heparanase and identify a lead compound for developing polysaccharide-based heparanase inhibitors.
Collapse
Affiliation(s)
- Sherket B Peterson
- Division of Medicinal Chemistry and Natural Products, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | |
Collapse
|
33
|
Bologna-Molina R, González-González R, Mosqueda-Taylor A, Molina-Frechero N, Damián-Matsumura P, Dominguez-Malagón H. Expression of syndecan-1 in papillary carcinoma of the thyroid with extracapsular invasion. Arch Med Res 2010; 41:33-7. [PMID: 20430252 DOI: 10.1016/j.arcmed.2009.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 10/23/2009] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Syndecan-1 (SDC-1) is a member of the family of transmembrane heparan sulfate proteoglycans, which are involved in cell-cell adhesion and the interaction of cells with the extracellular matrix. Evidence suggests that loss of SDC-1 expression in several benign and malignant epithelial neoplasms is an unfavorable prognostic indicator, but its expression profile in thyroid gland neoplasms remains to be elucidated. The aim of this study was to evaluate SDC-1 expression in papillary carcinomas of the thyroid (PCT) that were both larger and smaller (papillary microcarcinoma) than 10mm, with or without extracapsular extension (PCT-E and PCT-NE). METHODS The expression of SDC-1 was studied in 62 cases of PCT-E and PCT-NE using a tissue microarrays technique (TMA). SDC-1 positivity was predominantly observed in the cytoplasm of neoplastic epithelial cells and in the stroma of PCT. RESULTS SDC-1 is expressed in both neoplastic epithelial cells and the stroma. It is more frequently expressed in PCT-E than PCT-NE (p=0.002) and the stromal expression of SDC-1 is more intense in PCT-E that are >10 mm (p=0.026). CONCLUSIONS The epithelial and stromal expression of SDC-1 observed in this series of PCT suggests that the expression of this protein may be related to extracapsular invasion.
Collapse
Affiliation(s)
- Ronell Bologna-Molina
- Instituto de Investigación en Odontología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, Mexico.
| | | | | | | | | | | |
Collapse
|
34
|
Brimo F, Vollmer RT, Friszt M, Corcos J, Bismar TA. Syndecan-1 expression in prostate cancer and its value as biomarker for disease progression. BJU Int 2009; 106:418-23. [DOI: 10.1111/j.1464-410x.2009.09099.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
Brun R, Naroditsky I, Waterman M, Ben-Izhak O, Groisman G, Ilan N, Vlodavsky I. Heparanase expression by Barrett's epithelium and during esophageal carcinoma progression. Mod Pathol 2009; 22:1548-54. [PMID: 19749739 DOI: 10.1038/modpathol.2009.115] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Enzymatic activity responsible for the cleavage of heparan sulfate, commonly known as heparanase, is abundant in tumor-derived cells. Heparanase cleaves heparan sulfate side chains, presumably at sites of low sulfation, thus facilitating structural alterations of the extracellular matrix and basement membrane underlying epithelial and endothelial cells. Traditionally, heparanase activity was correlated with the metastatic potential of tumor-derived cells, attributed to enhanced cell dissemination as a consequence of heparan sulfate cleavage and remodeling of the extracellular matrix barrier. More recently, heparanase upregulation was documented in an increasing number of human carcinomas and hematological malignancies, correlating with increased tumor metastasis, vascular density, and shorter post-operative survival of cancer patients. Although heparanase upregulation and its pro-malignant features are well documented, the instance of its induction in the course of tumor development was less investigated. Here, we used immunohistochemical analysis to investigate heparanase expression in normal esophagus, Barrett's esophagus without dysplasia, Barrett's esophagus with low-grade dysplasia, Barrett's esophagus with high-grade dysplasia, and adenocarcinoma of the esophagus. We report that heparanase expression is already induced in Barrett's epithelium without dysplasia, and is further increased during progression through distinct pathological stages, namely, low-grade dysplasia, high-grade dysplasia, and adenocarcinoma. Notably, heparanase induction correlated with increased cell proliferation index revealed by Ki-67 staining. These findings suggest that heparanase function is not limited to the process of tumor metastasis, but rather is engaged at the early stages of esophagus carcinoma initiation and progression.
Collapse
Affiliation(s)
- Rita Brun
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Heparanase is an endo-beta-D-glucuronidase capable of cleaving heparan sulphate (HS) side chains of heparan sulphate proteoglycans on cell surfaces and the extracellular matrix; activity that is strongly implicated in tumour metastasis and angiogenesis. It has been shown that heparanase overexpression in human leukaemia, glioma and breast carcinoma cells results in a marked increase in tissue factor (TF) levels. In addition, TF was induced by exogenous addition of recombinant heparanase to tumour cells and primary endothelial cells; induction that was mediated by p38 phosphorylation and correlated with enhanced procoagulant activity. TF induction was further confirmed in transgenic mice overexpressing heparanase, and correlated with heparanase expression levels in leukaemia patients. Heparanase was also found to be involved in the regulation of tissue factor pathway inhibitor (TFPI). It has been shown that heparanase overexpression or exogenous addition induces a two- to three-fold increase in TFPI expression. Similarly, heparanase stimulated accumulation of TFPI in the cell culture medium. However, extracellular accumulation exceeded the observed increase in TFPI at the protein level, and appeared to be independent of HS and heparanase enzymatic activity. Instead, a physical interaction between heparanase and TFPI was demonstrated, suggesting a mechanism by which secreted heparanase interacts with TFPI on the cell surface, leading to dissociation of TFPI from the cell membrane and increased coagulation activity, thus further supporting the local prothrombotic function of heparanase. As heparins are strong inhibitors of heparanase, in view of the effect of heparanase on the TF/TFPI pathway, the role of anticoagulant activity of heparin may potentially be expanded. Taking into account the prometastatic and pro-angiogenic functions of heparanase, its overexpression in human malignancies and abundance in platelets, its involvement in the coagulation machinery is an intriguing novel arena for further research.
Collapse
|
37
|
Määttä JA, Olli K, Henttinen T, Tuittila MT, Elenius K, Salmivirta M. Removal of cell surface heparan sulfate increases TACE activity and cleavage of ErbB4 receptor. BMC Cell Biol 2009; 10:5. [PMID: 19171023 PMCID: PMC2636757 DOI: 10.1186/1471-2121-10-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 01/26/2009] [Indexed: 12/13/2022] Open
Abstract
Background Nuclear localization of proteolytically formed intracellular fragment of ErbB4 receptor tyrosine kinase has been shown to promote cell survival, and nuclear localization of ErbB4 receptor has been described in human breast cancer. Tumor necrosis factor alpha converting enzyme (TACE) initiates the proteolytic cascade leading to ErbB4 intracellular domain formation. Interactions between matrix metalloproteases and heparan sulfate have been described, but the effect of cell surface heparan sulfate on TACE activity has not been previously described. Results As indicated by immunodetection of increased ErbB4 intracellular domain formation and direct enzyme activity analysis, TACE activity was substantially amplified by enzymatic removal of cell surface heparan sulfate but not chondroitin sulfate. Conclusion In this communication, we suggest a novel role for cell surface heparan sulfate. Removal of cell surface heparan sulfate led to increased formation of ErbB4 intracellular domain. As ErbB4 intracellular domain has previously been shown to promote cell survival this finding may indicate a novel mechanism how HS degradation active in tumor tissue may favor cell survival.
Collapse
Affiliation(s)
- Jorma A Määttä
- Turku Center for Disease Modeling/Department of Cell Biology and Anatomy, University of Turku, Turku, Finland.
| | | | | | | | | | | |
Collapse
|
38
|
Ohashi M, Kusumi T, Sato F, Kudo Y, Jin H, Akasaka H, Miyamoto K, Toyoki Y, Hakamada K, Kijima H. Expression of syndecan-1 and E-cadherin is inversely correlated with poor patient's prognosis and recurrent status of extrahepatic bile duct carcinoma. Biomed Res 2009; 30:79-86. [DOI: 10.2220/biomedres.30.79] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
39
|
Chow LQM, Gustafson DL, O'Bryant CL, Gore L, Basche M, Holden SN, Morrow MC, Grolnic S, Creese BR, Roberts KL, Davis K, Addison R, Eckhardt SG. A phase I pharmacological and biological study of PI-88 and docetaxel in patients with advanced malignancies. Cancer Chemother Pharmacol 2008; 63:65-74. [PMID: 18320191 PMCID: PMC2813677 DOI: 10.1007/s00280-008-0712-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 02/17/2008] [Indexed: 12/30/2022]
Abstract
PURPOSE This study evaluated the safety, toxicity, pharmacological properties and biological activity of PI-88, a heparanase endoglycosidase enzyme inhibitor, with fixed weekly docetaxel in patients with advanced solid malignancies. EXPERIMENTAL DESIGN This was a phase I study to determine the maximal-tolerated dose of escalating doses of PI-88 administered subcutaneously for 4 days per week, along with docetaxel 30 mg/m(2) given on days 1, 8, 15 of a 28-day schedule. RESULTS Sixteen patients received a total of 42 courses of therapy. No dose-limiting toxicities were observed despite escalation to the highest planned dose level of PI-88 (250 mg/day). Frequent minor toxicities included fatigue (38%), dysgeusia (28.5%), thrombocytopenia (12%), diarrhea (14%), nausea (12%), and emesis (10%) in the 42 courses. No significant bleeding complications were observed. One patient developed a positive anti-heparin antibody test/serotonin releasing assay with positive anti-platelet factor 4/PI-88 antibodies and grade 1 thrombocytopenia in cycle 5, and was withdrawn from the study without any sequelae. PI-88 plasma concentrations (mirrored by APTT) and urinary elimination were linear and dose-proportional. Docetaxel did not alter the pharmacokinetic (PK) profile of PI-88, nor did PI-88 affect docetaxel PK. No significant relationship was determined between plasma or urine FGF-2, or plasma VEGF levels and PI-88 dose/response. Although no objective responses were observed; 9 of the 15 evaluable patients had stable disease for greater than two cycles of therapy. CONCLUSION PI-88 administered at 250 mg/day for 4 days each week for 3 weeks with docetaxel 30 mg/m(2) on days 1, 8 and 15, every 28 days, was determined to be the recommended dose level for phase II evaluation. This combination was well tolerated without severe toxicities or PK interactions.
Collapse
Affiliation(s)
- Laura Q M Chow
- University of Colorado Heath Sciences Center, Aurora, CO, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mikami S, Oya M, Shimoda M, Mizuno R, Ishida M, Kosaka T, Mukai M, Nakajima M, Okada Y. Expression of heparanase in renal cell carcinomas: implications for tumor invasion and prognosis. Clin Cancer Res 2008; 14:6055-61. [PMID: 18809970 DOI: 10.1158/1078-0432.ccr-08-0750] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Heparanase activity has been detected in many malignant tumors, showing a correlation with the metastatic potential. The present study was undertaken to investigate the expression of heparanase and its prognostic significance in renal cell carcinomas (RCC). EXPERIMENTAL DESIGN Nineteen RCCs and 6 nonneoplastic renal tissues were analyzed for heparanase mRNA expression by real-time PCR. Heparanase protein expression was semiquantitatively investigated by immunohistochemistry in 70 RCCs. Involvement of heparanase in the invasiveness of RCC cell lines, 786-O and Caki-2 cells, was examined by down-regulating the gene expression with small interfering RNA (siRNA) using the Matrigel invasion assay. RESULTS The expression level of heparanase mRNA was significantly higher in clear cell RCCs than in papillary RCCs, chromophobe RCCs, and nonneoplastic renal tissues. Heparanase was predominantly immunolocalized to cell surface and cytoplasm of clear cell RCCs and mean expression levels of heparanase were significantly higher in clear cell RCCs than in papillary and chromophobe RCCs. The protein expression levels were positively correlated with primary tumor stage, distant metastasis, and histologic grade. Targeting of heparanase mRNA expression in 786-O and Caki-2 cells with siRNA down-regulated the mRNA expression and inhibited the Matrigel invasion by these cells, whereas nonsilencing siRNA showed no effect. Multivariate Cox analysis revealed that elevated heparanase expression was a significant and an independent predictor of disease-specific survival (odds ratio, 8.814; P = 0.019). CONCLUSIONS These data suggest that heparanase plays an important role in invasion and metastasis and silencing of the gene might be a potential therapeutic target in clear cell RCCs.
Collapse
Affiliation(s)
- Shuji Mikami
- Division of Diagnostic Pathology, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
AIM To propose a quantitative method to detect heparanase-2 (HPA2) and syndecan-1 (Syn-1) using immunohistochemistry in colorectal (colon and rectal) carcinomas compared with nonneoplastic tissues and evaluate the possible role of these molecules in tumor development and extracellular remodeling. METHODS Cytoplasmic staining of HPA2 and Syn-1 was obtained by standard immunohistochemical reactions in 50 colorectal carcinoma and 20 nonneoplastic large bowels tissues. An image system was used to quantify the immunoexpression by digital computer-assisted method (Matos et al. 2006). The cutoff point for the immunohistochemistry variable was defined by sensibility and specificity curves. Statistical analysis was performed using SPSS version 13.0. RESULTS HPA2 was over-expressed in colorectal cancer (131.1+/-24.9 o.u./microm) when compared with nonneoplastic tissues (27.9+/-12.2 o.u./microm) (P<0.0001). However, an opposite correlation was observed between Syn-1 and tumor presence, where colorectal tissues expressed lower Syn-1 proteoglycan compared with nonneoplastic tissues, respectively (39.2+/-17.8 o.u./microm) and (102.2+/-25.2 o.u./microm) (P<0.0001). CONCLUSION A methodology with high sensitivity and specificity is proposed with a cutoff value for HPA2 and Syn-1 in the immunohistochemistry assay to define the presence of tumor. It was demonstrated for the first time in the literature that HPA2 is over-expressed in colorectal carcinoma tissues compared with nonneoplastic tissues. HPA2 over-expression could be possibly related to Syn-1 shedding despite the fact that HPA2 does not present enzymatic activity as HPA1.
Collapse
|
42
|
Lai NS, Simizu S, Morisaki D, Muroi M, Osada H. Requirement of the conserved, hydrophobic C-terminus region for the activation of heparanase. Exp Cell Res 2008; 314:2834-45. [PMID: 18662687 DOI: 10.1016/j.yexcr.2008.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Revised: 06/12/2008] [Accepted: 07/03/2008] [Indexed: 01/31/2023]
Abstract
Heparanase is an endo-beta-D-glucuronidase responsible for the cleavage of heparan sulfate, participating in extracellular matrix degradation and remodeling. Heparanase activity is well correlated with the potential for metastasis and angiogenesis in a large number of tumor-derived cell types, directly implicating the involvement of heparanase in tumor progression. Here, we provide the first evidence that the hydrophobic C-terminus region of heparanase has specific roles in intracellular trafficking, secretion, activation, and heparanase-mediated tumor cell migration. Furthermore, partial deletion of this hydrophobic C-terminus region, substitution within the hydrophobic C-terminus region to hydrophilic amino acids, and experiments of single amino acid mutations further point out the importance of the hydrophobic C-terminus region. Therefore, our findings suggest that the hydrophobic C-terminus region of heparanase is a determinant for its intracellular trafficking to the Golgi apparatus, followed by secretion, activation, and tumor cell migration.
Collapse
Affiliation(s)
- Ngit Shin Lai
- Antibiotics Laboratory, Advanced Science Institute, RIKEN, Saitama 351-0198, Japan
| | | | | | | | | |
Collapse
|
43
|
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant histological subtype of esophageal cancer in Asia, characterized by high incidence and mortality rate. Although significant progress has been made in surgery and adjuvant chemoradiotherapy, the prognosis of the patients with this cancer still remains poor. Investigation into protein alterations that occurred in tumors can provide clues to discover new biomarkers for improving diagnosis and guiding targeted therapy. Hundreds of papers have appeared over the past several decades concerning protein alterations in ESCC. This review summarizes all the dysregulated proteins investigated in the disease from 187 published papers and analyzes their contributions to tumor development and progression. We document protein alterations associated with tumor metastasis and the transition from normal esophageal epithelia to dysplasia in order to reveal the most useful markers for prediction of clinical outcome, early detection, and identification of high-risk patients for targeted therapies. In particular, we discuss the largest and most rigorous studies on prognostic implications of proteins in ESCC, in which cyclin D1, p53, E-cadherin and VEGF appeared to have the strongest evidence as independent predictors of patient outcome.
Collapse
Affiliation(s)
- D-C Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | |
Collapse
|
44
|
Sun H, Berquin IM, Owens RT, O'Flaherty JT, Edwards IJ. Peroxisome proliferator-activated receptor gamma-mediated up-regulation of syndecan-1 by n-3 fatty acids promotes apoptosis of human breast cancer cells. Cancer Res 2008; 68:2912-9. [PMID: 18413760 DOI: 10.1158/0008-5472.can-07-2305] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Diets enriched in n-3 polyunsaturated fatty acids (n-3 PUFA) may protect against breast cancer but biochemical mechanisms are unclear. Our studies showed that the n-3 fatty acid docosahexaenoic acid (DHA) up-regulated syndecan-1 (SDC-1) in human breast cancer cells, and we tested the hypothesis that DHA-mediated up-regulation of SDC-1 induces apoptosis. DHA was delivered to MCF-7 cells by n-3 PUFA-enriched low-density lipoproteins (LDL) or by albumin in the presence or absence of SDC-1 small interfering RNA. The n-3 PUFA induced apoptosis, which was blocked by SDC-1 silencing. We also confirmed that SDC-1 up-regulation and apoptosis promotion by n-3 PUFA was mediated by peroxisome proliferator-activated receptor gamma (PPAR gamma). Using a luciferase gene driven by either a PPAR response element or a DR-1 site present in the SDC-1 promoter, reporter activities were enhanced by n-3 LDL, DHA, and PPAR gamma agonist, whereas activity of a luciferase gene placed downstream of a mutant DR-1 site was unresponsive. Cotransfection with dominant-negative PPAR gamma DNA eliminated the increase in luciferase activity. These data provide strong evidence that SDC-1 is a molecular target of n-3 PUFA in human breast cancer cells through activation of PPAR gamma and that n-3 PUFA-induced apoptosis is mediated by SDC-1. This provides a novel mechanism for the chemopreventive effects of n-3 PUFA in breast cancer.
Collapse
Affiliation(s)
- Haiguo Sun
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
45
|
Tsuneki M, Cheng J, Maruyama S, Ida-Yonemochi H, Nakajima M, Saku T. Perlecan-rich epithelial linings as a background of proliferative potentials of keratocystic odontogenic tumor. J Oral Pathol Med 2008; 37:287-93. [DOI: 10.1111/j.1600-0714.2007.00620.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
46
|
Abstract
Heparan sulphate proteoglycans are ubiquitous macromolecules of cell surfaces and extracellular matrices. Numerous extracellular matrix proteins, growth factors, morphogens, cytokines, chemokines and coagulation factors are bound and regulated by heparan sulphate. Degradation of heparan sulphate thus potentially profoundly affects cell and tissue function. Although there is evidence that several heparan sulphate-degrading endoglucuronidases (heparanases) might exist, so far only one transcript encoding a functional heparanase has been identified: heparanase-1. In the first part of this review, we discuss the current knowledge about heparan sulphate proteoglycans and the functional importance of their versatile interactions. In the second part, we summarize recent findings that have contributed to the characterization of heparanase-1, focusing on the molecular properties, working mechanism, substrate specificity, expression pattern, cellular activation and localization of this enzyme. Additionally, we review data implicating heparanase-1 in several normal and pathological processes, focusing on tumour metastasis and angiogenesis, and on evidence for a potentially direct signalling function of the molecule. In that context, we also briefly discuss heparanase-2, an intriguing close homologue of heparanase-1, for which, so far, no heparan sulphate-degrading activity could be demonstrated.
Collapse
Affiliation(s)
- Veronique Vreys
- Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
- Laboratory for Glycobiology and Developmental Genetics, Department of Human Genetics, Catholic University of Leuven, Leuven, Belgium
- *Correspondence to: Guido DAVID Centre for Human Genetics, Campus Gasthuisberg, O&N1, Herestraat 49, 3000 Leuven, Belgium. Tel.: +32-16-345863; Fax: +32-16-347166; E-mail:
| | - Guido David
- Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
- Laboratory for Glycobiology and Developmental Genetics, Department of Human Genetics, Catholic University of Leuven, Leuven, Belgium
- *Correspondence to: Guido DAVID Centre for Human Genetics, Campus Gasthuisberg, O&N1, Herestraat 49, 3000 Leuven, Belgium. Tel.: +32-16-345863; Fax: +32-16-347166; E-mail:
| |
Collapse
|
47
|
Simizu S, Suzuki T, Muroi M, Lai NS, Takagi S, Dohmae N, Osada H. Involvement of disulfide bond formation in the activation of heparanase. Cancer Res 2007; 67:7841-9. [PMID: 17699790 DOI: 10.1158/0008-5472.can-07-1053] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Heparanase is overexpressed in many solid tumor cells and is capable of specifically cleaving heparan sulfate, and this activity is associated with the metastatic potential of tumor cells; however, the activation mechanism of heparanase has remained unknown. In this study, we investigated the link between disulfide bond formation and the activation of heparanase in human tumor cells. Mass spectrometry analysis of heparanase purified from a conditioned medium of human fibrosarcoma cells revealed two disulfide bonds, Cys127-Cys179 and Cys437-Cys542, and one S-cysteinylation at the Cys211 residue. It was shown that, although the formation of the Cys127-Cys179 bond and S-cysteinylation at Cys211 have little effect on heparanase function, the disulfide bond between Cys437 and Cys542 is necessary for the secretion and activation of heparanase. Thus, the present findings will provide a basis for the further refinement of heparanase structural studies and for the development of novel heparanase inhibitors.
Collapse
Affiliation(s)
- Siro Simizu
- Antibiotics Laboratory, Discovery Research Institute, RIKEN, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Katase N, Nagatsuka H, Tsujigiwa H, Gunduz M, Tamamura R, Pwint HP, Rivera RS, Nakajima M, Naomoto Y, Nagai N. Analysis of the neoplastic nature and biological potential of sporadic and nevoid basal cell carcinoma syndrome-associated keratocystic odontogenic tumor. J Oral Pathol Med 2007; 36:550-4. [PMID: 17850439 DOI: 10.1111/j.1600-0714.2007.00578.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Keratocystic odontogenic tumor (KCOT), also known as odontogenic keratocyst, is a benign cystic neoplasm, which may be associated with nevoid basal cell carcinoma syndrome (NBCCS) and if it does, will occur as multiple cystic lesions. KCOT is locally destructive despite its bland histological features. However, the neoplastic nature of KCOT is not well established. Heparanase is an endo-d-glucuronidase enzyme that specifically cleaves heparan sulfate (HS) and the increase of its level in tumors promotes invasion, angiogenesis, and metastasis. METHODS To investigate the neoplastic character of KCOT, we studied the localization patterns of heparanase in KCOT, focusing on the differences between sporadic and NBCCS-associated KCOTs, by immunohistochemistry and in situ hybridization. To compare the expression pattern of these cysts with non-tumorous odontogenic developmental cyst, dentigerous cyst was included. RESULTS All the odontogenic cysts showed positive immunoreaction for heparanase protein in various intensities. The expression pattern of heparanase gene corresponded to that of protein expression. Interestingly, intense gene and protein expressions were observed in KCOT associated with NBCCS compared with sporadic ones and dentigerous cyst. CONCLUSIONS The results implied that heparanase expression may be correlated with the neoplastic properties of KCOT, particularly in NBCCS-associated cases.
Collapse
Affiliation(s)
- Naoki Katase
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata-cho, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jackson LL, Wade Z, Hessler RB, Abdelsayed R, Rogers JB, Gourin CG. Quantitative analysis of syndecan-1 expression in dysplasia and squamous cell carcinoma of the oral cavity. Laryngoscope 2007; 117:868-71. [PMID: 17473685 DOI: 10.1097/mlg.0b013e318033c810] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Decreased expression of syndecan-1 has been reported in dysplasia and squamous cell carcinoma (SCCA) of the oral cavity and appears to correlate with decreasing histological differentiation and poor clinical outcome. Assays of syndecan-1 expression to date have utilized manual microscopic analysis with qualitative grading of immunohistochemical staining intensity, which may introduce observer bias. We evaluated syndecan-1 expression in dysplasia and squamous cell carcinoma (SCCA) of the oral cavity, using a novel automated cellular imaging system that incorporates both staining intensity as well as the percentage of positively stained cells to yield a quantitative value for syndecan-1 expression. MATERIALS AND METHODS We performed a quantitative immunohistochemical analysis of syndecan-1 expression using an automated cellular image analysis system. We analyzed specimens from cases of mild dysplasia (N = 55), moderate dysplasia (N = 38), severe dysplasia (N = 25), carcinoma in situ (CIS) (N = 43), and SCCA of the oral cavity (N = 45), using normal mucosal epithelium (N = 21) as a positive control. The SCCA specimens were further subdivided by degree of differentiation. We retrospectively reviewed patient charts to identify tumor stage at diagnosis, recurrence, and disease-specific survival. RESULTS Syndecan-1 expression was significantly greater in normal controls than in specimens of mild, moderate, or severe dysplasia, CIS, or invasive SCCA (P < .05). Syndecan-1 expression did not differ significantly among specimens of mild, moderate, or severe dysplasia, CIS or SCCA. There was no significant difference in syndecan-1 expression between specimens from patients with no evidence of disease at 3 years follow-up and patients with local, regional, or distant recurrence. CONCLUSIONS Syndecan-1 expression does not appear to be useful as a marker of differentiation or as a prognostic indicator in dysplasia and SCCA of the oral cavity. The search for a suitable and reliable marker of biological aggressiveness is ongoing.
Collapse
Affiliation(s)
- Lana L Jackson
- Departments of Otolaryngology-Head and Neck Surgery, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The remodelling of the extracellular matrix (ECM) has been shown to be highly upregulated in cancer and inflammation and is critically linked to the processes of invasion and metastasis. One of the key enzymes involved in specifically degrading the heparan sulphate (HS) component of the ECM is the endo-beta-glucuronidase enzyme heparanase. Processing of HS by heparanase releases both a host of bioactive growth factors anchored within the mesh of the ECM as well as defined fragments of HS capable of promoting cellular proliferation. The finding that heparanase is elevated in a wide variety of tumor types and is subsequently linked to the development of pathological processes has led to an explosion of therapeutic strategies to inhibit its enzyme activity. So far only one compound, the sulphated oligosaccharide PI88, which both inhibits heparanase activity and has effects on growth factor binding has reached clinical trials where it has shown to have promising efficacy. The scene has clearly been set however for a new generation of compounds, either specific to the enzyme or with dual roles, to emerge from the lab and enter the clinic. The aim of this review is to describe the current drug discovery status of small molecule, sugar and neutralising antibody inhibitors of heparanase enzyme activity. Potential strategies will also be discussed on the selection of suitable biomarker strategies for specific monitoring of in vivo heparanase inhibition which will be crucial for both animal model and clinical trial testing.
Collapse
Affiliation(s)
- E A McKenzie
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|