1
|
Dolskii A, dos Santos SAA, Andrake M, Franco-Barraza J, Dunbrack RL, Cukierman E. Exploring the potential role of palladin in modulating human CAF/ECM functional units. Cytoskeleton (Hoboken) 2025; 82:175-185. [PMID: 39239855 PMCID: PMC11882928 DOI: 10.1002/cm.21926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Fibroblasts, crucial for maintaining tissue homeostasis, significantly shape the tumor microenvironment (TME). In pancreatic cancer, a highly aggressive malignancy, cancer-associated fibroblast (CAF)/extracellular matrix (ECM) units dominate the TME, influencing tumor initiation, progression, and treatment responses. Palladin, an actin-associated protein, is vital for fibroblast structural integrity and activation, playing a key role in CAF/ECM functionality. Palladin interacts with cytoskeletal proteins such as alpha-actinin (α-Act) and can therefore regulate other proteins like syndecans, modulating cytoskeletal features, cell adhesion, integrin recycling, and signaling. In this review, we propose that targeting the palladin/α-Act/syndecan interaction network could modulate CAF/ECM units, potentially shifting the TME from a tumor-promoting to a tumor-suppressive state. In silico data and reported studies to suggest that stabilizing palladin-α-Act interactions, via excess palladin, influences syndecan functions; potentially modulating integrin endocytosis via syndecan engagement with protein kinase C alpha as opposed to syndecan binding to α-Act. This mechanism can then affect the distribution of active α5β1-integrin between the plasma membrane and known intracellular vesicular compartments, thereby influencing the tumor-suppressive versus tumor-promoting functions of CAF/ECM units. Understanding these interactions offers likely future therapeutic avenues for stroma normalization in pancreatic and other cancers, aiming to inhibit tumor progression and improve future treatment outcomes.
Collapse
Affiliation(s)
| | | | - Mark Andrake
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| | - Janusz Franco-Barraza
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| | - Roland L. Dunbrack
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| |
Collapse
|
2
|
Reszegi A, Tátrai P, Regős E, Kovalszky I, Baghy K. Syndecan-1 in liver pathophysiology. Am J Physiol Cell Physiol 2022; 323:C289-C294. [PMID: 35704700 DOI: 10.1152/ajpcell.00039.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Syndecan-1 is a heparan sulfate/chondroitin sulfate proteoglycan (PG) of the cell surface and the extracellular matrix, which regulates a broad spectrum of physiological and pathological processes such as cell proliferation, migration, inflammation, matrix remodeling, wound healing, or tumorigenesis. Syndecan-1 represents the major PG of the liver, expressed by hepatocytes and cholangiocytes, and its elevated expression is a characteristic feature of liver diseases. The highest syndecan-1 expression is found in liver cirrhosis and in hepatocellular carcinoma (HCC) developed in cirrhotic livers. In addition, as being a hepatitis C receptor, hepatitis C virus (HCV) infected livers produce extremely large amounts of syndecan-1. The serum levels of the cleaved (shedded) extracellular domain has clinical significance, as its increased concentration reflects on poor prognosis in cirrhosis as well as in cancer. In vivo experiments confirmed that syndecan-1 protects against early stages of fibrogenesis mainly by enhanced clearance of transforming growth factor beta (TGFβ1) and thrombospondin-1 via circulation, and against hepatocarcinogenesis by interfering with several signaling pathways and enhancing cell cycle blockade. In addition, syndecan-1 is capable to hinder lipid metabolism and ribosomal biogenesis in induced cancer models.. These observations together with its participation in the uptake of viruses (e.g. HCV, SARS-CoV-2) indicate that syndecan-1 is a central player in liver pathologies.
Collapse
Affiliation(s)
- Andrea Reszegi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | | | - Eszter Regős
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornelia Baghy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
3
|
Yablecovitch D, Ben-Horin S, Picard O, Yavzori M, Fudim E, Nadler M, Levy I, Sakhnini E, Lang A, Engel T, Lahav M, Saker T, Neuman S, Selinger L, Dvir R, Raitses-Gurevich M, Golan T, Laish I. Serum Syndecan-1: A Novel Biomarker for Pancreatic Ductal Adenocarcinoma. Clin Transl Gastroenterol 2022; 13:e00473. [PMID: 35297817 PMCID: PMC9132524 DOI: 10.14309/ctg.0000000000000473] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/01/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Syndecan-1 (SDC1) has multiple functions in tumorigenesis in general and specifically in pancreatic cancer. We aimed to evaluate SDC1 as a diagnostic and prognostic biomarker in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS In this case-control study, patients newly diagnosed with a biopsy-proven PDAC were enrolled alongside healthy individuals in a derivation-validation cohort design. Serum SDC1 was measured by enzyme-linked immunoassay. The diagnostic accuracy of SDC1 levels for diagnosing PDAC was computed. A unified cohort enriched with additional early-stage patients with PDAC was used to evaluate the association of SDC1 with survival outcomes and patient characteristics. RESULTS In the derivation cohort, serum SDC1 levels were significantly higher in patients with PDAC (n = 39) compared with healthy controls (n = 20) (40.1 ng/mL, interquartile range 29.8-95.3 vs 25.6 ng/mL, interquartile range 17.1-29.8, respectively; P < 0.001). The receiver operating characteristic analysis area under the curve was 0.847 (95% confidence interval 0.747-0.947, P < 0.001). These results were replicated in a separate age-matched validation cohort (n = 38 PDAC, n = 38 controls; area under the curve 0.844, 95% confidence interval 0.757-0.932, P < 0.001). In the combined-enriched PDAC cohort (n = 110), using a cutoff of 35 ng/mL, the median overall 5-year survival between patients below and above this cutoff was not significantly different, although a trend for better survival after 1 year was found in the lower level group (P = 0.06). There were 12 of the 110 patients with PDAC (11%) who had normal CA 19-9 in the presence of elevated SDC1. DISCUSSION These findings suggest serum SDC1 as a promising novel biomarker for early blood-based diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Doron Yablecovitch
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Shomron Ben-Horin
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Orit Picard
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Miri Yavzori
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Ella Fudim
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Moshe Nadler
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Idan Levy
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Emad Sakhnini
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Alon Lang
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Tal Engel
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Maor Lahav
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Talia Saker
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Shalvata Mental Health Center, Hod Hasharon, Israel;
| | - Sandra Neuman
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Limor Selinger
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Revital Dvir
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Maria Raitses-Gurevich
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel.
| | - Talia Golan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel.
| | - Ido Laish
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| |
Collapse
|
4
|
Alghandour R, Ebrahim MA, Ghazy H, Shamaa S, Emarah Z, Al-Gayyar MM. Evaluation of the Diagnostic and Prognostic Value of Syndecan-1 in Acute Leukemia Patients. Cureus 2020; 12:e10594. [PMID: 32983743 PMCID: PMC7511075 DOI: 10.7759/cureus.10594] [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: 08/29/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Syndecan-1 (also known as SDC-1 or CD138) is a transmembrane proteoglycan that is expressed in many hematological and solid tumors and affects the prognosis of those cancers. We conducted this study to investigate the prognostic role of syndecan-1 in acute leukemia. Forty cases of de novo acute leukemia patients, 24 with acute myeloid leukemia (AML) and 16 with acute lymphoblastic leukemia (ALL), presented at the Oncology Center of Mansoura University, Mansoura, Egypt, with a follow-up period of 26 months. Syndecan-1 was determined in serum and leukocytes by enzyme-linked immunosorbent assay (ELISA). The results from acute leukemia patients were compared with those of 15 healthy subjects. We observed that soluble syndecan-1 was higher in AML (median, 160.60 ng/ml) compared with ALL (median, 76.10 ng/ml) and healthy controls (median, 30.95 ng/ml). There was a significant correlation between syndecan-1 either in leukocytes or soluble form and response to treatment in patients with AML (p = 0.02 and p = 0.04, respectively), but these correlations were not statistically significant for ALL cases. Finally, there was a significant correlation between the soluble syndecan-1 level and overall survival in AML cases (p = 0.04), but the correlation was not significant for ALL cases. In conclusion, syndecan-1 is a useful biomarker for AML but not for ALL.
Collapse
Affiliation(s)
- Reham Alghandour
- Medical Oncology, Internal Medicine Department, Mansoura University Faculty of Medicine, Mansoura, EGY
| | - Mohamed A Ebrahim
- Medical Oncology, Internal Medicine Department, Mansoura University Faculty of Medicine, Mansoura, EGY
- Medical Oncology Unit, Oncology Center, Mansoura University, Mansoura, EGY
| | - Hayam Ghazy
- Medical Oncology, Internal Medicine Department, Mansoura University Faculty of Medicine, Mansoura, EGY
| | - Sameh Shamaa
- Medical Oncology, Internal Medicine Department, Mansoura University Faculty of Medicine, Mansoura, EGY
| | - Ziad Emarah
- Medical Oncology Unit, Oncology Center, Mansoura University, Mansoura, EGY
- Medical Oncology, Internal Medicine Department, Mansoura University Faculty of Medicine, Mansoura, EGY
| | - Mohammed M Al-Gayyar
- Department of Pharmaceutical Chemistry, University of Tabuk Faculty of Pharmacy, Tabuk, SAU
- Department of Biochemistry, Mansoura University Faculty of Pharmacy, Mansoura, EGY
| |
Collapse
|
5
|
Yang W, Eken Y, Zhang J, Cole LE, Ramadan S, Xu Y, Zhang Z, Liu J, Wilson AK, Huang X. Chemical synthesis of human syndecan-4 glycopeptide bearing O-, N-sulfation and multiple aspartic acids for probing impacts of the glycan chain and the core peptide on biological functions. Chem Sci 2020; 11:6393-6404. [PMID: 34094105 PMCID: PMC8159385 DOI: 10.1039/d0sc01140a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Proteoglycans are a family of complex glycoproteins with glycosaminoglycan chains such as heparan sulfate (HS) attached to the core protein backbone. Due to the high structural heterogeneity of HS in nature, it is challenging to decipher the respective roles of the HS chain and the core protein on proteoglycan functions. While the sulfation patterns of HS dictate many activities, the core protein can potentially impact HS functions. In order to decipher this, homogeneous proteoglycan glycopeptides are needed. Herein, we report the first successful synthesis of proteoglycan glycopeptides bearing multiple aspartic acids in the core peptide and O- and N-sulfations in the glycan chain, as exemplified by the syndecan-4 glycopeptides. To overcome the high acid sensitivities of sulfates and base sensitivities of the glycopeptide during synthesis, a new synthetic approach has been developed to produce a sulfated glycan chain on a peptide sequence prone to the formation of aspartimide side products. The availability of the structurally well-defined synthetic glycopeptide enabled the investigation of their biological functions including cytokine, growth factor binding and heparanase inhibition. Interestingly, the glycopeptide exhibited context dependent enhancement or decrease of biological activities compared to the peptide or the glycan alone. The results presented herein suggest that besides varying the sulfation patterns of HS, linking the HS chain to core proteins as in proteoglycans may be an additional approach to modulate biological functions of HS in nature.
Collapse
Affiliation(s)
- Weizhun Yang
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA
| | - Yigitcan Eken
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA
| | - Jicheng Zhang
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA
| | - Logan Emerson Cole
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA
| | - Sherif Ramadan
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA.,Chemistry Department, Faculty of Science, Benha University Benha Qaliobiya 13518 Egypt
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina Chapel Hill NC 27599 USA
| | - Zeren Zhang
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina Chapel Hill NC 27599 USA
| | - Angela K Wilson
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University 578 South Shaw Lane East Lansing MI 48824 USA.,Department of Biomedical Engineering, Michigan State University East Lansing MI 48824 USA.,Institute for Quantitative Health Science and Engineering, Michigan State University East Lansing MI 48824 USA
| |
Collapse
|
6
|
Benediktsson S, Kander T, Ostrowski SR, Johansson PI, Thomas OD, Schött U. Platelet increment is not associated with endothelial damage in haematological patients: a prospective observational study. Scandinavian Journal of Clinical and Laboratory Investigation 2019; 79:395-403. [DOI: 10.1080/00365513.2019.1636403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- S. Benediktsson
- Medical Faculty, Institution of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Perioperative and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - T. Kander
- Medical Faculty, Institution of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Perioperative and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - S. R. Ostrowski
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - P. I. Johansson
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - O. D. Thomas
- Medical Faculty, Institution of Clinical Sciences Lund, Lund University, Lund, Sweden
- Paediatric Intensive Care Unit, SUS Skåne University Hospital Lund, Lund, Sweden
| | - U. Schött
- Medical Faculty, Institution of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Perioperative and Intensive Care, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
Gondelaud F, Ricard‐Blum S. Structures and interactions of syndecans. FEBS J 2019; 286:2994-3007. [DOI: 10.1111/febs.14828] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/04/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Frank Gondelaud
- ICBMS UMR 5246 CNRS – University Lyon 1 Univ Lyon Villeurbanne France
| | | |
Collapse
|
8
|
Metwaly HA, El-Gayar AM, El-Shishtawy MM. Inhibition of the signaling pathway of syndecan-1 by synstatin: A promising anti-integrin inhibitor of angiogenesis and proliferation in HCC in rats. Arch Biochem Biophys 2018; 652:50-58. [PMID: 29928859 DOI: 10.1016/j.abb.2018.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/18/2018] [Accepted: 06/16/2018] [Indexed: 12/25/2022]
Abstract
AIM OF WORK The study was conducted for evaluation of the antitumor activity of SSTN92-119 against HCC induced by thioacetamide in rats. METHODS Sixty male Sprague-Dawley rats were randomized into four equal groups: Control, SSTN92-119, HCC, and HCC + SSTN92-119. Liver function tests were measured in serum. Liver homogenate was used for determination of: i) integrinαѴβ3 (ITGαѴβ3), insulin like growth factor-1 receptor (IGF-1R), vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2) and alpha-fetoprotein (AFP) levels by ELISA, ii) syndecan-1 (CD-138), IGF-1R and VEGF genes expressions by qRT-PCR, iii) MDA, NO, GSH concentrations and SOD activity. Histopathological and immunohistochemical examination of liver tissue was performed. RESULTS SSTN92-119 decreased HCC-induced elevation in ALT, AST, ALP and GGT activities and reversed HCC-induced reduction in total protein and albumin concentrations significantly. SSTN92-119 significantly elevated hepatic SOD and GSH and reduced both NO and MDA levels. Protein levels of ITGαѴβ3, IGF-1R, VEGF, FGF-2 and AFP were decreased in HCC- SSTN92-119 group as well as gene expression of CD-138, IGF-1R and VEGF compared with HCC group. CONCLUSIONS SSTN92-119 down regulates ITGαѴβ3 receptor and subsequently reduces the activation of angiogenic growth factors VEGF and FGF-2. Therefore, SSTN92-119 is becoming a promising anti-integrin αѴβ3 that inhibits angiogenesis and proliferation in HCC.
Collapse
Affiliation(s)
- Heba A Metwaly
- Dept. of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Dept. of Biochemistry, Faculty of Pharmacy, Delta University for Science & Technology, International Costal Road, Gamasa City, Mansoura, Dakhliya, Egypt
| | - Amal M El-Gayar
- Dept. of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Mamdouh M El-Shishtawy
- Dept. of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
9
|
The transcriptional factor ZEB1 represses Syndecan 1 expression in prostate cancer. Sci Rep 2018; 8:11467. [PMID: 30065348 PMCID: PMC6068163 DOI: 10.1038/s41598-018-29829-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022] Open
Abstract
Syndecan 1 (SDC-1) is a cell surface proteoglycan with a significant role in cell adhesion, maintaining epithelial integrity. SDC1 expression is inversely related to aggressiveness in prostate cancer (PCa). During epithelial to mesenchymal transition (EMT), loss of epithelial markers is mediated by transcriptional repressors such as SNAIL, SLUG, or ZEB1/2 that bind to E-box promoter sequences of specific genes. The effect of these repressors on SDC-1 expression remains unknown. Here, we demonstrated that SNAIL, SLUG and ZEB1 expressions are increased in advanced PCa, contrarily to SDC-1. SNAIL, SLUG and ZEB1 also showed an inversion to SDC-1 in prostate cell lines. ZEB1, but not SNAIL or SLUG, represses SDC-1 as demonstrated by experiments of ectopic expression in epithelial prostate cell lines. Inversely, expression of ZEB1 shRNA in PCa cell line increased SDC-1 expression. The effect of ZEB1 is transcriptional since ectopic expression of this gene represses SDC-1 promoter activity and ZEB1 binds to the SDC-1 promoter as detected by ChIP assays. An epigenetic mark associated to transcription repression H3K27me3 was bound to the same sites that ZEB1. In conclusion, this study identifies ZEB1 as a key repressor of SDC-1 during PCa progression and point to ZEB1 as a potentially diagnostic marker for PCa.
Collapse
|
10
|
The heparanase/heparan sulfate proteoglycan axis: A potential new therapeutic target in sarcomas. Cancer Lett 2016; 382:245-254. [PMID: 27666777 DOI: 10.1016/j.canlet.2016.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
Abstract
Heparanase, the only known mammalian endoglycosidase degrading heparan sulfate (HS) chains of HS proteoglycans (HSPG), is a highly versatile protein affecting multiple events in tumor cells and their microenvironment. In several malignancies, deregulation of the heparanase/HSPG system has been implicated in tumor progression, hence representing a valuable therapeutic target. Currently, multiple agents interfering with the heparanase/HSPG axis are under clinical investigation. Sarcomas are characterized by a high biomolecular complexity and multiple levels of interconnection with microenvironment sustaining their growth and progression. The clinical management of advanced diseases remains a challenge. In several sarcoma subtypes, high levels of heparanase expression have been correlated with poor prognosis associated factors. On the other hand, expression of cell surface-associated HSPGs (i.e. glypicans and syndecans) has been found altered in specific sarcoma subtypes. Recent studies provided the preclinical proof-of-principle of the role of the heparanase/HSPG axis as therapeutic target in various sarcoma subtypes. Although currently there are no clinical trials evaluating agents targeting heparanase and/or HSPGs in sarcomas, we here provide arguments for this strategy as potentially able to implement the therapeutic options for sarcoma patients.
Collapse
|
11
|
Zeng YE, Yao XH, Yan ZP, Liu JX, Liu XH. Potential signaling pathway involved in sphingosine-1-phosphate-induced epithelial-mesenchymal transition in cancer. Oncol Lett 2016; 12:379-382. [PMID: 27347154 DOI: 10.3892/ol.2016.4661] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 05/24/2016] [Indexed: 12/19/2022] Open
Abstract
The developmental process of epithelial-mesenchymal transition (EMT) occurs when epithelial cells acquire invasive mesenchymal cell characteristics, and the activation of this process has been indicated to be involved in tumor progression. EMT could be induced by growth factors, cytokines and matrix metalloproteinases (MMPs). sphingosine-1-phosphate (S1P) is a biologically-active lipid that plays an important role in cancer metastasis. S1P also contributes to the activation of EMT. However, the mechanism underlying S1P-induced EMT is unclear. Increased evidence has demonstrated that the cell surface glycocalyx is closed associated with S1P and plays an important role in tumor progression, suggesting that S1P-induced EMT could be Snail-MMP signaling-dependent. Thus, we hypothesize that an S1P-glycocalyx-Snail-MMP signaling axis mediates S1P-induced EMT. This is an essential step towards improved understanding of the underlying mechanism involved in S1P-regulted EMT, and the development of novel diagnostic and anticancer therapeutic strategies.
Collapse
Affiliation(s)
- Y E Zeng
- Institute of Biomedical Engineering, School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xing-Hong Yao
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zhi-Ping Yan
- Institute of Biomedical Engineering, School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing-Xia Liu
- Institute of Biomedical Engineering, School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiao-Heng Liu
- Institute of Biomedical Engineering, School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
12
|
Choi S, Choi Y, Jun E, Kim IS, Kim SE, Jung SA, Oh ES. Shed syndecan-2 enhances tumorigenic activities of colon cancer cells. Oncotarget 2016; 6:3874-86. [PMID: 25686828 PMCID: PMC4414160 DOI: 10.18632/oncotarget.2885] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/09/2014] [Indexed: 12/14/2022] Open
Abstract
Because earlier studies showed the cell surface heparan sulfate proteoglycan, syndecan-2, sheds from colon cancer cells in culture, the functional roles of shed syndecan-2 were assessed. A non-cleavable mutant of syndecan-2 in which the Asn148-Leu149 residues were replaced with Asn148-Ile149, had decreased shedding, less cancer-associated activities of syndecan-2 in vitro, and less syndecan-2-mediated metastasis of mouse melanoma cells in vivo, suggesting the importance of shedding on syndecan-2-mediated pro-tumorigenic functions. Indeed, shed syndecan-2 from cancer-conditioned media and recombinant shed syndecan-2 enhanced cancer-associated activities, and depletion of shed syndecan-2 abolished these effects. Similarly, shed syndecan-2 was detected from sera of patients from advanced carcinoma (625.9 ng/ml) and promoted cancer-associated activities. Furthermore, a series of syndecan-2 deletion mutants showed that the tumorigenic activity of shed syndecan-2 resided in the C-terminus of the extracellular domain and a shed syndecan-2 synthetic peptide (16 residues) was sufficient to establish subcutaneous primary growth of HT29 colon cancer cells, pulmonary metastases (B16F10 cells), and primary intrasplenic tumor growth and liver metastases (4T1 cells). Taken together, these results demonstrate that shed syndecan-2 directly enhances colon cancer progression and may be a promising therapeutic target for controlling colon cancer development.
Collapse
Affiliation(s)
- Sojoong Choi
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea
| | - Youngsil Choi
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Eunsung Jun
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea.,Department of Biochemistry and Cell Biology, School of Medicine and Cell & Matrix Research Institute, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Seong-Eun Kim
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Republic of Korea
| | - Sung-Ae Jung
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Republic of Korea
| | - Eok-Soo Oh
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Republic of Korea
| |
Collapse
|
13
|
Akl MR, Nagpal P, Ayoub NM, Prabhu SA, Gliksman M, Tai B, Hatipoglu A, Goy A, Suh KS. Molecular and clinical profiles of syndecan-1 in solid and hematological cancer for prognosis and precision medicine. Oncotarget 2015; 6:28693-715. [PMID: 26293675 PMCID: PMC4745686 DOI: 10.18632/oncotarget.4981] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/11/2015] [Indexed: 12/18/2022] Open
Abstract
Syndecan-1 (SDC1, CD138) is a key cell surface adhesion molecule essential for maintaining cell morphology and interaction with the surrounding microenvironment. Deregulation of SDC1 contributes to cancer progression by promoting cell proliferation, metastasis, invasion and angiogenesis, and is associated with relapse through chemoresistance. SDC1 expression level is also associated with responses to chemotherapy and with prognosis in multiple solid and hematological cancers, including multiple myeloma and Hodgkin lymphoma. At the tissue level, the expression levels of SDC1 and the released extracellular domain of SDC1 correlate with tumor malignancy, phenotype, and metastatic potential for both solid and hematological tumors in a tissue-specific manner. The SDC1 expression profile varies among cancer types, but the differential expression signatures between normal and cancer cells in epithelial and stromal compartments are directly associated with aggressiveness of tumors and patient's clinical outcome and survival. Therefore, relevant biomarkers of SDC signaling may be useful for selecting patients that would most likely respond to a particular therapy at the time of diagnosis or perhaps for predicting relapse. In addition, the reciprocal expression signature of SDC between tumor epithelial and stromal compartments may have synergistic value for patient selection and the prediction of clinical outcome.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Ahmet Hatipoglu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
14
|
Surviladze Z, Sterkand RT, Ozbun MA. Interaction of human papillomavirus type 16 particles with heparan sulfate and syndecan-1 molecules in the keratinocyte extracellular matrix plays an active role in infection. J Gen Virol 2015; 96:2232-2241. [PMID: 26289843 PMCID: PMC4681067 DOI: 10.1099/vir.0.000147] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/13/2015] [Indexed: 12/18/2022] Open
Abstract
Oncogenic human papillomaviruses (HPVs) attach predominantly to extracellular matrix (ECM) components during infection of cultured keratinocytes and in the rodent vaginal challenge model in vivo. However, the mechanism of virion transfer from the ECM to receptors that mediate entry into host cells has not been determined. In this work we strove to assess the role of heparan sulfate (HS) chains in HPV16 binding to the ECM and determine how HPV16 release from the ECM is regulated. We also assessed the extent to which capsids released from the ECM are infectious. We show that a large fraction of HPV16 particles binds to the ECM via HS chains, and that syndecan-1 (snd-1) molecules present in the ECM are involved in virus binding. Inhibiting the normal processing of snd-1 and HS molecules via matrix metalloproteinases and heparanase dramatically reduces virus release from the ECM, cellular uptake and infection. Conversely, exogenous heparinase activates each of these processes. We confirm that HPV16 released from the ECM is infectious in keratinocytes. Use of a specific inhibitor shows furin is not involved in HPV16 release from ECM attachment factors and corroborates other studies showing only the intracellular activity of furin is responsible for modulating HPV infectivity. These data suggest that our recently proposed model, describing the action of HS proteoglycan processing enzymes in releasing HPV16 from the cell surface in complex with the attachment factor snd-1, is also relevant to the release of HPV16 particles from the ECM to promote efficient infection of keratinocytes.
Collapse
Affiliation(s)
- Zurab Surviladze
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| | - Rosa T. Sterkand
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| | - Michelle A. Ozbun
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
15
|
Cigliana G, Torti E, Gulli F, De Santis E, Dell'Abate MT, Colacicco L, Pisani F, Conti L, Basile U. Relationship between circulating syndecan-1 levels (CD138s) and serum free light chains in monoclonal gammopathies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:37. [PMID: 26025441 PMCID: PMC4495691 DOI: 10.1186/s13046-015-0155-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/07/2015] [Indexed: 12/21/2022]
Abstract
Background Monoclonal gammopathies encompass a wide range of diseases characterized by the monoclonal expansion of a B-cell clone. Despite emerging therapeutic strategies, chances of survival of patients who are affected are still scarce, which implies that new tools are necessary not only for the diagnosis but also for the follow-up of patients affected by such diseases. In this context, the use of free light chains (FLCs) has been incorporated into many guidelines. Likewise, tumor microenvironment is consistently gaining importance as role player in tumor pathogenesis. Specifically, Syndecan-1 (CD138), a heparan-sulfate proteoglycan is attracting interests as it is highly expressed and shed by myeloma plasma-cells. The aim of our study was to analyze CD138 levels in the serum of patients affected by multiple myeloma or light chain only disease, and to compare the values obtained with free light chain (FLC) kappa, lambda and FLC ratio in both groups of patients. Methods 84 patients affected by Multiple Myeloma and Light Chain Myeloma were recruited for this study. Serum CD138 was assessed by ELISA (Diaclone Research, France) and FLC values were quantified by nephelometry (Freelite TM Human Kappa and Lambda Free Kits, The Binding Site, UK). Data was analyzed by GraphPad Prism software and Statgraph. Results We observed higher CD138 mean values in myeloma patients compared to the light chain only myeloma group. A positive linear regression of CD138 and FLC was observed in the light chain only cohort as opposed to myeloma patients which show an inverse trend. Conclusions The study highlighted an existing relationship between FLCs and CD138 and wishes to seek also a correlation in order to rapidly and efficiently perform diagnosis and different diagnostic schemes.
Collapse
Affiliation(s)
- Giovanni Cigliana
- Department of Prevention and Diagnostic Oncology, Laboratory of Clinical Pathology -National Cancer Institute "Regina Elena", Rome, Italy.
| | - Eleonora Torti
- Department of Laboratory Medicine, School of Medicine, Catholic University of the Sacred Heart, Largo A. Gemelli 8, Rome, ZIP CODE: 00168, Italy.
| | - Francesca Gulli
- School of Medicine - Institute of Internal Medicine, Catholic University of the Sacred Heart, Rome, Italy.
| | - Elena De Santis
- Department of Laboratory Medicine, School of Medicine, Catholic University of the Sacred Heart, Largo A. Gemelli 8, Rome, ZIP CODE: 00168, Italy.
| | - Maria Teresa Dell'Abate
- Department of Laboratory Medicine, School of Medicine, Catholic University of the Sacred Heart, Largo A. Gemelli 8, Rome, ZIP CODE: 00168, Italy.
| | - Luigi Colacicco
- Department of Laboratory Medicine, School of Medicine, Catholic University of the Sacred Heart, Largo A. Gemelli 8, Rome, ZIP CODE: 00168, Italy.
| | - Francesco Pisani
- Hematology and Transplantation, Italian National Cancer Institute "Regina Elena", Rome, Italy.
| | - Laura Conti
- Department of Prevention and Diagnostic Oncology, Laboratory of Clinical Pathology -National Cancer Institute "Regina Elena", Rome, Italy.
| | - Umberto Basile
- Department of Laboratory Medicine, School of Medicine, Catholic University of the Sacred Heart, Largo A. Gemelli 8, Rome, ZIP CODE: 00168, Italy.
| |
Collapse
|
16
|
Kim SY, Choi EJ, Yun JA, Jung ES, Oh ST, Kim JG, Kang WK, Lee SH. Syndecan-1 expression is associated with tumor size and EGFR expression in colorectal carcinoma: a clinicopathological study of 230 cases. Int J Med Sci 2015; 12:92-9. [PMID: 25589885 PMCID: PMC4293174 DOI: 10.7150/ijms.10497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 12/07/2014] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Syndecan-1 (SDC1) is reported to modulate several key processes of tumorigenesis and has variable expression in many cancers. To date, the cause of altered expression has not been elucidated. In this study, we compared SDC1 expression with various clinicopathological parameters and molecular markers to evaluate its clinical significance in colorectal carcinoma. METHODS We screened for SDC1 expression using immunohistochemistry in 230 surgical specimens of primary colorectal carcinoma from patients consecutively treated between 2008 and 2011 at Seoul St. Mary's Hospital, The Catholic University of Korea. The relationship between SDC1 expression and various clinicopathological parameters and molecular markers was analyzed. RESULTS The tumors were principally located in the left colon (71.3%) and rectum (33.5%). There were 216 (93.9%) adenocarcinomas, 10 (4.3%) mucinous adenocarcinomas, and 4 other tumors. Most of the carcinomas were pT3 (68.3%) and pT4 (22.2%). There was regional lymph node metastasis in 140 patients. SDC1 expression was identified in the cancer cells of 212 (96.8%) colon cancer cases. Of the SDC1-positive cases, 131 showed predominantly membranous immunopositivity, and 81 showed a predominantly cytoplasmic staining pattern. Mixed membranous and cytoplasmic staining was observed in 154 cases. In 93 cases, stromal SDC1 reactivity was noted. Epithelial SDC1 immunopositivity was significantly associated with tumor size (p=0.016) and epidermal growth factor receptor expression (p=0.006). However, it was not significantly correlated with lymph node metastasis, distant metastasis, lymphatic or vascular invasion, or KRAS mutation. In addition, stromal SDC1 immunopositivity was significantly associated with the male sex (p=0.018). CONCLUSIONS The expression profile of SDC1 may be of clinical value in colorectal cancer and may help in identifying aggressive forms of colorectal carcinoma. Further studies are needed in order to better understand the role of SDC1 in the progression and invasiveness of colorectal carcinoma.
Collapse
Affiliation(s)
- Su Young Kim
- 1. Department of Pathology, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Eun Ji Choi
- 1. Department of Pathology, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Jeong A Yun
- 1. Department of Pathology, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Eun Sun Jung
- 2. Department of Hospital Pathology, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Seung Taek Oh
- 3. Department of Surgery, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Jun Gi Kim
- 3. Department of Surgery, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Won Kyung Kang
- 3. Department of Surgery, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| | - Sung Hak Lee
- 2. Department of Hospital Pathology, The Catholic University of Korea, School of Medicine, Seocho-gu Banpodaero 222, Seoul 137-701, South Korea
| |
Collapse
|
17
|
Zaragosi LE, Dadone B, Michiels JF, Marty M, Pedeutour F, Dani C, Bianchini L. Syndecan-1 regulates adipogenesis: new insights in dedifferentiated liposarcoma tumorigenesis. Carcinogenesis 2014; 36:32-40. [PMID: 25344834 DOI: 10.1093/carcin/bgu222] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Syndecan-1 (SDC1/CD138) is one of the main cell surface proteoglycans and is involved in crucial biological processes. Only a few studies have analyzed the role of SDC1 in mesenchymal tumor pathogenesis. In particular, its involvement in adipose tissue tumors has never been investigated. Dedifferentiated liposarcoma, one of the most frequent types of malignant adipose tumors, has a high potential of recurrence and metastastic evolution. Classical chemotherapy is inefficient in metastatic dedifferentiated liposarcoma and novel biological markers are needed for improving its treatment. In this study, we have analyzed the expression of SDC1 in well-differentiated/dedifferentiated liposarcomas and showed that SDC1 is highly overexpressed in dedifferentiated liposarcoma compared with normal adipose tissue and lipomas. Silencing of SDC1 in liposarcoma cells impaired cell viability and proliferation. Using the human multipotent adipose-derived stem cell model of human adipogenesis, we showed that SDC1 promotes proliferation of undifferentiated adipocyte progenitors and inhibits their adipogenic differentiation. Altogether, our results support the hypothesis that SDC1 might be involved in liposarcomagenesis. It might play a prominent role in the dedifferentiation process occurring when well-differentiated liposarcoma progress to dedifferentiated liposarcoma. Targeting SDC1 in these tumors might provide a novel therapeutic strategy.
Collapse
Affiliation(s)
- Laure-Emmanuelle Zaragosi
- Institute of Biology Valrose, UMR7277 CNRS/UMR1091 INSERM/University of Nice-Sophia Antipolis, 06108 Nice, France, Present address: CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, University of Nice- Sophia Antipolis, 06560 Sophia Antipolis, France
| | - Bérengère Dadone
- Department of Pathology, Nice University Hospital, 06202 Nice, France, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284/INSERM U1081, University of Nice-Sophia Antipolis, 06107 Nice, France, Laboratory of Solid Tumor Genetics, Nice University Hospital, 06107 Nice, France and
| | - Jean-François Michiels
- Department of Pathology, Nice University Hospital, 06202 Nice, France, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284/INSERM U1081, University of Nice-Sophia Antipolis, 06107 Nice, France
| | - Marion Marty
- Department of Pathology, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Florence Pedeutour
- Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284/INSERM U1081, University of Nice-Sophia Antipolis, 06107 Nice, France, Laboratory of Solid Tumor Genetics, Nice University Hospital, 06107 Nice, France and
| | - Christian Dani
- Institute of Biology Valrose, UMR7277 CNRS/UMR1091 INSERM/University of Nice-Sophia Antipolis, 06108 Nice, France
| | - Laurence Bianchini
- Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284/INSERM U1081, University of Nice-Sophia Antipolis, 06107 Nice, France,
| |
Collapse
|
18
|
Barbouri D, Afratis N, Gialeli C, Vynios DH, Theocharis AD, Karamanos NK. Syndecans as modulators and potential pharmacological targets in cancer progression. Front Oncol 2014; 4:4. [PMID: 24551591 PMCID: PMC3910246 DOI: 10.3389/fonc.2014.00004] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/09/2014] [Indexed: 12/17/2022] Open
Abstract
Extracellular matrix (ECM) components form a dynamic network of key importance for cell function and properties. Key macromolecules in this interplay are syndecans (SDCs), a family of transmembrane heparan sulfate proteoglycans (HSPGs). Specifically, heparan sulfate (HS) chains with their different sulfation pattern have the ability to interact with growth factors and their receptors in tumor microenvironment, promoting the activation of different signaling cascades that regulate tumor cell behavior. The affinity of HS chains with ligands is altered during malignant conditions because of the modification of chain sequence/sulfation pattern. Furthermore, matrix degradation enzymes derived from the tumor itself or the tumor microenvironment, like heparanase and matrix metalloproteinases, ADAM as well as ADAMTS are involved in the cleavage of SDCs ectodomain at the HS and protein core level, respectively. Such released soluble SDCs "shed SDCs" in the ECM interact in an autocrine or paracrine manner with the tumor or/and stromal cells. Shed SDCs, upon binding to several matrix effectors, such as growth factors, chemokines, and cytokines, have the ability to act as competitive inhibitors for membrane proteoglycans, and modulate the inflammatory microenvironment of cancer cells. It is notable that SDCs and their soluble counterparts may affect either the behavior of cancer cells and/or their microenvironment during cancer progression. The importance of these molecules has been highlighted since HSPGs have been proposed as prognostic markers of solid tumors and hematopoietic malignancies. Going a step further down the line, the multi-actions of SDCs in many levels make them appealing as potential pharmacological targets, either by targeting directly the tumor or indirectly the adjacent stroma.
Collapse
Affiliation(s)
- Despoina Barbouri
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikolaos Afratis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Chrisostomi Gialeli
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Demitrios H. Vynios
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
19
|
Poblete CE, Fulla J, Gallardo M, Muñoz V, Castellón EA, Gallegos I, Contreras HR. Increased SNAIL expression and low syndecan levels are associated with high Gleason grade in prostate cancer. Int J Oncol 2014; 44:647-54. [PMID: 24424718 PMCID: PMC3928469 DOI: 10.3892/ijo.2014.2254] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/27/2013] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer (PC) is a leading male oncologic malignancy wideworld. During malignant transformation, normal epithelial cells undergo genetic and morphological changes known as epithelial-mesenchymal transition (EMT). Several regulatory genes and specific marker proteins are involved in PC EMT. Recently, syndecans have been associated with malignancy grade and Gleason score in PC. Considering that SNAIL is mainly a gene repressor increased in PC and that syndecan promoters have putative binding sites for this repressor, we propose that SNAIL might regulate syndecan expression during PC EMT. The aim of this study was to analyze immunochemically the expression of SNAIL, syndecans 1 and 2 and other EMT markers in a tissue microarray (TMA) of PC samples and PC cell lines. The TMAs included PC samples of different Gleason grade and benign prostatic hyperplasia (BPH) samples, as non-malignant controls. PC3 and LNCaP cell lines were used as models of PC representing different tumorigenic capacities. Semi-quantitative immunohistochemistry was performed on TMAs and fluorescence immunocytochemistry and western blot analysis were conducted on cell cultures. Results show that SNAIL exhibits increased expression in high Gleason specimens compared to low histological grade and BPH samples. Accordingly, PC3 cells show higher SNAIL expression levels compared to LNCaP cells. Conversely, syndecan 1, similarly to E-cadherin (a known marker of EMT), shows a decreased expression in high Gleason grades samples and PC3 cells. Interestingly, syndecan 2 shows no changes associated to histological grade. It is concluded that increased SNAIL levels in advanced PC are associated with low expression of syndecan 1. The mechanism by which SNAIL regulates the expression of syndecan 1 remains to be investigated.
Collapse
Affiliation(s)
- Cristian E Poblete
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Fulla
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Marcela Gallardo
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Valentina Muñoz
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Enrique A Castellón
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ivan Gallegos
- Pathological Anatomy Service, Clinic Hospital, University of Chile, Santiago, Chile
| | - Hector R Contreras
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
20
|
Ramani VC, Sanderson RD. Chemotherapy stimulates syndecan-1 shedding: a potentially negative effect of treatment that may promote tumor relapse. Matrix Biol 2013; 35:215-22. [PMID: 24145151 DOI: 10.1016/j.matbio.2013.10.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 01/01/2023]
Abstract
In patients with multiple myeloma, the heparan sulfate proteoglycan syndecan-1 (CD138) is shed from the surface of tumor cells and accumulates in the serum and within the extracellular matrix of the bone marrow where it promotes tumor growth and metastasis. In the present study we discovered that commonly used anti-myeloma drugs stimulate syndecan-1 shedding both in vitro and in animals bearing myeloma tumors. Enhanced shedding is accompanied by increased syndecan-1 synthesis prior to drug induced tumor cell death. Addition of a caspase inhibitor blocks the drug-induced shedding of syndecan-1 in vitro indicating that shedding is linked to the onset of apoptosis. ADAM inhibitors or siRNA targeting ADAMs blocked drug-induced shedding suggesting that upregulation or activation of ADAMs is responsible for cleaving syndecan-1 from the tumor cell surface. These results reveal that myeloma chemotherapy stimulates synthesis and shedding of syndecan-1, a potentially negative side effect that may lead to the accumulation of high levels of syndecan-1 to establish a microenvironment that nurtures relapse and promotes tumor progression. Interestingly, we also found that chemotherapeutic drugs stimulated syndecan-1 shedding from pancreatic cancer cells as well, indicating that drug-induced shedding of syndecan-1 may occur in many cancer types. Overall, our results indicate that the use of metalloproteinase inhibitors (to inhibit syndecan-1 shedding) in combination with chemotherapy may represent a novel therapeutic strategy to prevent re-establishment of a microenvironment conducive for tumor relapse.
Collapse
Affiliation(s)
- Vishnu C Ramani
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ralph D Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
21
|
Ramani VC, Purushothaman A, Stewart MD, Thompson CA, Vlodavsky I, Au JLS, Sanderson RD. The heparanase/syndecan-1 axis in cancer: mechanisms and therapies. FEBS J 2013; 280:2294-306. [PMID: 23374281 DOI: 10.1111/febs.12168] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 12/21/2022]
Abstract
Heparanase is an endoglucuronidase that cleaves heparan sulfate chains of proteoglycans. In many malignancies, high heparanase expression and activity correlate with an aggressive tumour phenotype. A major consequence of heparanase action in cancer is a robust up-regulation of growth factor expression and increased shedding of syndecan-1 (a transmembrane heparan sulfate proteoglycan). Substantial evidence indicates that heparanase and syndecan-1 work together to drive growth factor signalling and regulate cell behaviours that enhance tumour growth, dissemination, angiogenesis and osteolysis. Preclinical and clinical studies have demonstrated that therapies targeting the heparanase/syndecan-1 axis hold promise for blocking the aggressive behaviour of cancer.
Collapse
Affiliation(s)
- Vishnu C Ramani
- Department of Pathology, University of Alabama at Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Strand ME, Herum KM, Rana ZA, Skrbic B, Askevold ET, Dahl CP, Vistnes M, Hasic A, Kvaløy H, Sjaastad I, Carlson CR, Tønnessen T, Gullestad L, Christensen G, Lunde IG. Innate immune signaling induces expression and shedding of the heparan sulfate proteoglycan syndecan-4 in cardiac fibroblasts and myocytes, affecting inflammation in the pressure-overloaded heart. FEBS J 2013; 280:2228-47. [DOI: 10.1111/febs.12161] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 01/21/2013] [Accepted: 01/28/2013] [Indexed: 12/22/2022]
|
23
|
Choi S, Kang DH, Oh ES. Targeting syndecans: a promising strategy for the treatment of cancer. Expert Opin Ther Targets 2013; 17:695-705. [DOI: 10.1517/14728222.2013.773313] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
24
|
Surviladze Z, Dziduszko A, Ozbun MA. Essential roles for soluble virion-associated heparan sulfonated proteoglycans and growth factors in human papillomavirus infections. PLoS Pathog 2012; 8:e1002519. [PMID: 22346752 PMCID: PMC3276557 DOI: 10.1371/journal.ppat.1002519] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 12/19/2011] [Indexed: 12/12/2022] Open
Abstract
A subset of human papillomavirus (HPV) infections is causally related to the development of human epithelial tumors and cancers. Like a number of pathogens, HPV entry into target cells is initiated by first binding to heparan sulfonated proteoglycan (HSPG) cell surface attachment factors. The virus must then move to distinct secondary receptors, which are responsible for particle internalization. Despite intensive investigation, the mechanism of HPV movement to and the nature of the secondary receptors have been unclear. We report that HPV16 particles are not liberated from bound HSPG attachment factors by dissociation, but rather are released by a process previously unreported for pathogen-host cell interactions. Virus particles reside in infectious soluble high molecular weight complexes with HSPG, including syndecan-1 and bioactive compounds, like growth factors. Matrix mellatoproteinase inhibitors that block HSPG and virus release from cells interfere with virus infection. Employing a co-culture assay, we demonstrate HPV associated with soluble HSPG-growth factor complexes can infect cells lacking HSPG. Interaction of HPV-HSPG-growth factor complexes with growth factor receptors leads to rapid activation of signaling pathways important for infection, whereas a variety of growth factor receptor inhibitors impede virus-induced signaling and infection. Depletion of syndecan-1 or epidermal growth factor and removal of serum factors reduce infection, while replenishment of growth factors restores infection. Our findings support an infection model whereby HPV usurps normal host mechanisms for presenting growth factors to cells via soluble HSPG complexes as a novel method for interacting with entry receptors independent of direct virus-cell receptor interactions. A subset of the >120 different types of human papillomaviruses (HPVs) are the most common cause of sexually transmitted infections. Certain HPVs are also associated with approximately 5% of all cancers worldwide. Like many pathogens, HPVs bind first to heparan sulfate proteoglycans (HSPGs) on cells before moving to more specific uptake receptors. However, relatively little is known about the mechanism(s) that triggers the translocation of HPV from HSPGs to the receptors that facilitate entry. As obligate parasites, viruses have evolved numerous means to hijack host cell functions to cause infection. We report two novel mechanisms of pathogen-host interactions. First, bound HPV particles are liberated from cells in an active complex with HSPGs and growth factors rather than dissociating from the sugars to engage secondary receptors. Second, HPV uses the specificity of the associated growth factors to bridge to their cognate receptors as opposed to direct binding to a cell internalization receptor. Signals transduced during these interactions are important for HPV infection. Our study provides new insights into the transmission of a significant viral pathogen and reveals novel means whereby microbes may repurpose normal cell functions during infection of their hosts. Likewise, this work uncovers new targets for HPV prophylaxis.
Collapse
Affiliation(s)
- Zurab Surviladze
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail: (ZS); (MAO)
| | - Agnieszka Dziduszko
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Michelle A. Ozbun
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail: (ZS); (MAO)
| |
Collapse
|
25
|
Metwaly HA, Al-Gayyar MMH, Eletreby S, Ebrahim MA, El-Shishtawy MM. Relevance of serum levels of interleukin-6 and syndecan-1 in patients with hepatocellular carcinoma. Sci Pharm 2012; 80:179-188. [PMID: 22396913 PMCID: PMC3293347 DOI: 10.3797/scipharm.1110-07] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 12/23/2011] [Indexed: 02/08/2023] Open
Abstract
Syndecan-1 is a trans-membrane heparan sulfate proteoglycan that localizes in epithelial cells and has been shown to be present in normal hepatocytes. It is thought to be involved in processes such as cell growth, differentiation and adhesion. However, the clinical data regarding syndecan-1 in patients with hepatocellular carcinoma (HCC) are scarce and controversial. Therefore, we need to evaluate the effects of HCC on the serum levels of syndecan-1. Thus, 40 patients with HCC and 31 patients with liver cirrhosis were physically examined. Blood samples were taken for measurements of routine markers (sGPT, sGOT, bilirubin, albumin, and α-fetoprotein), as well as serum levels of interleukin (IL)-6 and syndecan-1. Patients with liver cirrhosis showed significant increase in serum IL-6 as compared with HCC patients and the control subjects. Serum level of syndecan-1 was significantly increased in HCC patients as compared with the cirrhotic and control groups. In addition, significant positive correlations between syndecan-1 and serum levels of ALT, AST in HCC patients were found. Moreover, syndecan-1 increased significantly with increasing stage of Barcelona-Clinic Liver Cancer Group diagnostic and treatment strategy. In conclusion, the development of HCC is accompanied by a significant elevation in serum syndecan-1 levels. The increase in serum syndecan-1 may be linked with progression of HCC.
Collapse
Affiliation(s)
- Heba A. Metwaly
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | | | - Shahira Eletreby
- Department of Internal Medicine, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | | | - Mamdouh M. El-Shishtawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
26
|
Lendorf ME, Manon-Jensen T, Kronqvist P, Multhaupt HAB, Couchman JR. Syndecan-1 and syndecan-4 are independent indicators in breast carcinoma. J Histochem Cytochem 2011; 59:615-29. [PMID: 21430259 DOI: 10.1369/0022155411405057] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Syndecan proteoglycans may be key regulators of tumor invasion and metastasis because this four-member family of transmembrane receptors regulates cell adhesion, proliferation, and differentiation. Their expression can also serve as prognostic markers. In breast carcinomas, syndecan-1 overexpression correlates with poor prognosis and aggressive phenotype. Syndecan-4 is expressed in most breast carcinoma cell lines, but its role in malignancy is unclear. A possible relationship between syndecan-1 and syndecan-4 expression and established prognostic factors in breast carcinomas was examined. Duplicate samples of 114 benign and malignant breast disease cases were stained for the two syndecans. Clinicopathological information was available for all cases. Syndecan-1 was detected in 72.8% of cases, with significant association between its expression and histological tumor type (p<0.05) and high grade tumors (p<0.05). Syndecan-4 was expressed in 66.7% of cases; expression correlated significantly with positive estrogen (p<0.01) and progesterone (p<0.01) receptor status. Independent expression of the two syndecans was noted from an analysis of single and double positive cases. There was a statistical relationship between syndecan-1 presence in high-grade tumors and absence of syndecan-4, whereas syndecan-4 presence in cases positive for estrogen and progesterone receptor associated with syndecan-1 absence. These syndecans may, therefore, have distinct roles in regulating breast carcinoma cell behavior.
Collapse
Affiliation(s)
- Maria E Lendorf
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
27
|
Mendez MP, Monroy YK, Du M, Preston AM, Tolle L, Lin Y, VanDussen KL, Samuelson LC, Standiford TJ, Curtis JL, Beck JM, Christensen PJ, Paine R. Overexpression of sICAM-1 in the alveolar epithelial space results in an exaggerated inflammatory response and early death in Gram negative pneumonia. Respir Res 2011; 12:12. [PMID: 21247482 PMCID: PMC3034680 DOI: 10.1186/1465-9921-12-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 01/19/2011] [Indexed: 11/19/2022] Open
Abstract
Background A sizeable body of data demonstrates that membrane ICAM-1 (mICAM-1) plays a significant role in host defense in a site-specific fashion. On the pulmonary vascular endothelium, mICAM-1 is necessary for normal leukocyte recruitment during acute inflammation. On alveolar epithelial cells (AECs), we have shown previously that the presence of normal mICAM-1 is essential for optimal alveolar macrophage (AM) function. We have also shown that ICAM-1 is present in the alveolar space as a soluble protein that is likely produced through cleavage of mICAM-1. Soluble intercellular adhesion molecule-1 (sICAM-1) is abundantly present in the alveolar lining fluid of the normal lung and could be generated by proteolytic cleavage of mICAM-1, which is highly expressed on type I AECs. Although a growing body of data suggesting that intravascular sICAM-1 has functional effects, little is known about sICAM-1 in the alveolus. We hypothesized that sICAM-1 in the alveolar space modulates the innate immune response and alters the response to pulmonary infection. Methods Using the surfactant protein C (SPC) promoter, we developed a transgenic mouse (SPC-sICAM-1) that constitutively overexpresses sICAM-1 in the distal lung, and compared the responses of wild-type and SPC-sICAM-1 mice following intranasal inoculation with K. pneumoniae. Results SPC-sICAM-1 mice demonstrated increased mortality and increased systemic dissemination of organisms compared with wild-type mice. We also found that inflammatory responses were significantly increased in SPC-sICAM-1 mice compared with wild-type mice but there were no difference in lung CFU between groups. Conclusions We conclude that alveolar sICAM-1 modulates pulmonary inflammation. Manipulating ICAM-1 interactions therapeutically may modulate the host response to Gram negative pulmonary infections.
Collapse
Affiliation(s)
- Michael P Mendez
- Division of Pulmonary and Critical Care Medicine, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|