1
|
Li C, Luo P, Guo F, Jia X, Shen M, Zhang T, Wang S, Du T. Identification of HSPG2 as a bladder pro-tumor protein through NID1/AKT signaling. Cancer Cell Int 2024; 24:345. [PMID: 39438949 PMCID: PMC11515648 DOI: 10.1186/s12935-024-03527-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
PURPOSE Heparan sulfate proteoglycans (HSPGs) are complex molecules found on the cell membrane and within the extracellular matrix, increasingly recognized for their role in tumor progression. This study aimed to investigate the involvement of Heparan sulfate proteoglycan 2 (HSPG2) in the progression of bladder cancer. METHODS We identified HSPG2 as a promoter of bladder tumor progression using single-cell RNA sequencing and transcriptome analysis of sequencing data from seven patient samples obtained from the Gene Expression Omnibus (GEO) database (GSE135337). Transcript profiles of 28 normal tissues and 407 bladder tumor tissues were analyzed for HSPG2 expression and survival outcomes using the Sanger tools and cBioPortal databases. HSPG2-overexpressing T24 and Biu-87 cell lines were generated, and cell proliferation and migration were assessed using CCK-8 and Transwell assays. Western blotting and immunostaining were performed to evaluate the activation of Nidogen-1 (NID1)/protein kinase B (AKT) signaling. Mouse models with patient-derived tumor organoids (HSPG2high and HSPG2low) were established to assess anticancer effects. RESULTS Our results demonstrated a marked upregulation of HSPG2 in malignant bladder tumors, which correlated significantly with poor patient prognosis. HSPG2 overexpression consistently enhanced bladder tumor cell proliferation and conferred chemotherapy resistance, as shown in both in vitro and in vivo experiments. Mechanistically, HSPG2 upregulated NID1 expression, leading to the activation of the AKT pro-survival signaling pathway and promoting sustained tumor growth in bladder cancer. CONCLUSION This study highlights the critical pro-tumor role of HSPG2/NID1/AKT signaling in bladder cancer and suggests its potential as a therapeutic target in clinical treatment.
Collapse
Affiliation(s)
- Cong Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Pengwei Luo
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Fengzhu Guo
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xu Jia
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Min Shen
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Ting Zhang
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Shusen Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Ting Du
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
2
|
Kulbay M, Marcotte E, Remtulla R, Lau THA, Paez-Escamilla M, Wu KY, Burnier MN. Uveal Melanoma: Comprehensive Review of Its Pathophysiology, Diagnosis, Treatment, and Future Perspectives. Biomedicines 2024; 12:1758. [PMID: 39200222 PMCID: PMC11352094 DOI: 10.3390/biomedicines12081758] [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: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/02/2024] Open
Abstract
Uveal melanoma (UM) is the most common intraocular malignancy in adults. Recent advances highlight the role of tumor-derived extracellular vesicles (TEV) and circulating hybrid cells (CHC) in UM tumorigenesis. Bridged with liquid biopsies, a novel technology that has shown incredible performance in detecting cancer cells or products derived from tumors in bodily fluids, it can significantly impact disease management and outcome. The aim of this comprehensive literature review is to provide a summary of current knowledge and ongoing advances in posterior UM pathophysiology, diagnosis, and treatment. The first section of the manuscript discusses the complex and intricate role of TEVs and CHCs. The second part of this review delves into the epidemiology, etiology and risk factors, clinical presentation, and prognosis of UM. Third, current diagnostic methods, ensued by novel diagnostic tools for the early detection of UM, such as liquid biopsies and artificial intelligence-based technologies, are of paramount importance in this review. The fundamental principles, limits, and challenges associated with these diagnostic tools, as well as their potential as a tracker for disease progression, are discussed. Finally, a summary of current treatment modalities is provided, followed by an overview of ongoing preclinical and clinical research studies to provide further insights on potential biomolecular pathway alterations and therapeutic targets for the management of UM. This review is thus an important resource for all healthcare professionals, clinicians, and researchers working in the field of ocular oncology.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Emily Marcotte
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Raheem Remtulla
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Tsz Hin Alexander Lau
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Manuel Paez-Escamilla
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada;
| | - Miguel N. Burnier
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
3
|
Onyeisi JOS, Nader HB, Lopes CC. Effects of syndecan-4 silencing on the extracellular matrix remodeling in anoikis-resistant endothelial cells. Cell Biol Int 2024; 48:883-897. [PMID: 38591778 DOI: 10.1002/cbin.12158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/07/2024] [Accepted: 03/02/2024] [Indexed: 04/10/2024]
Abstract
Anoikis is a process of programmed cell death induced by the loss of cell/matrix interactions. In previous work, we have shown that the acquisition of anoikis resistance upregulates syndecan-4 (SDC4) expression in endothelial cells. In addition, SDC4 gene silencing by microRNA interference reverses the transformed phenotype of anoikis-resistant endothelial cells. Due to this role of SDC4 in regulating the behavior of anoikis-resistant endothelial cells, we have evaluated that the functional consequences of SDC4 silencing in the extracellular matrix (ECM) remodeling in anoikis-resistant rabbit aortic endothelial cells submitted to SDC4 gene silencing (miR-Syn4-Adh-1-EC). For this, we evaluated the expression of adhesive proteins, ECM receptors, nonreceptor protein-tyrosine kinases, and ECM-degrading enzymes and their inhibitors. Altered cell behavior was monitored by adhesion, migration, and tube formation assays. We found that SDC4 silencing led to a decrease in migration and angiogenic capacity of anoikis-resistant endothelial cells; this was accompanied by an increase in adhesion to fibronectin. Furthermore, after SDC4 silencing, we observed an increase in the expression of fibronectin, collagen IV, and vitronectin, and a decrease in the expression of integrin α5β1 and αvβ3, besides that, silenced cells show an increase in Src and FAK expression. Quantitative polymerase chain reaction and Western blot analysis demonstrated that SDC4 silencing leads to altered gene and protein expression of MMP2, MMP9, and HSPE. Compared with parental cells, SDC4 silenced cells showed a decrease in nitric oxide production and eNOS expression. In conclusion, these data demonstrate that SDC4 plays an important role in ECM remodeling. In addition, our findings represent an important step toward understanding the mechanism by which SDC4 can reverse the transformed phenotype of anoikis-resistant endothelial cells.
Collapse
Affiliation(s)
- Jessica Oyie Sousa Onyeisi
- Departamento de Bioquímica, Disciplina de Biologia Molecular, Universidade Federal de São Paulo, Sao Paulo, São Paulo, Brazil
| | - Helena Bonciani Nader
- Departamento de Bioquímica, Disciplina de Biologia Molecular, Universidade Federal de São Paulo, Sao Paulo, São Paulo, Brazil
| | - Carla Cristina Lopes
- Departamento de Bioquímica, Disciplina de Biologia Molecular, Universidade Federal de São Paulo, Sao Paulo, São Paulo, Brazil
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| |
Collapse
|
4
|
Wang Y, Huang Y, Zhu H, Guo Z, Cheng J, Zhang C, Zhong M. Exostoisns (EXT1/2) in Head and Neck Cancers: An In Silico Analysis and Clinical Correlates. Int Dent J 2024; 74:446-453. [PMID: 37989698 PMCID: PMC11123571 DOI: 10.1016/j.identj.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/26/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVES The exostosins (EXT), which are responsible for heparan sulfate backbone synthesis and play a vital role in tissue homeostasis, have been reported to be correlated with prognosis of various cancers. However, the expression, prognostic value, and immune infiltration of EXT1 and EXT2 in head and neck squamous cell carcinoma (HNSC) remain uncertain. METHODS GEPIA, UALCAN, and Xiantao bioinformatics tools were used to explore the EXT1 and EXT2 expression level in HNSC. GEPIA and Sangerbox were utilised to obtain the prognostic value of EXT1 and EXT2 in HNSC. Genetic alterations, immune cell infiltration, and single-cell analysis were conducted in cBioPortal, TIMER, and TISCH2. In addition, the expressions of EXT1 and EXT2 were validated by real-time polymerase chain reaction (PCR) in HNSC samples. RESULTS EXT1 and EXT2 were highly expressed in HNSC, especially in malignant cells. Only EXT2 was significantly negatively correlated to the prognosis of patients with HNSC. EXT1 and EXT2 were found to be associated with focal adhesin and cell adhesin molecule binding. EXT1 expression levels were considerably connected with CD8+ T cell infiltrating levels, whilst EXT2 expression levels were considerably negatively connected with infiltrating levels of CD4+ T cells, macrophages, neutrophils, and dendritic cells in HNSC. The gene mutation rates of EXT1 and EXT2 in HNSC were 7% and 2.8%, respectively. Moreover, EXT2 was validated to be highly expressed in HNSC samples by real-time PCR. CONCLUSION EXT2 was highly expressed and presented negative correlation with the prognosis and immune infiltration of HNSC, which might be a potential biomarker for HNSC.
Collapse
Affiliation(s)
- Yiping Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College; Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Yan Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College; Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Houwei Zhu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College; Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Zhenzhen Guo
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College; Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Jun Cheng
- Department of Oral Histopathology, Stomatological Hospital of Xiamen Medical College; Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Churen Zhang
- Department of Stomatology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Ming Zhong
- Department of Oral Histopathology, School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Disease, China Medical University, Shenyang, China
| |
Collapse
|
5
|
Shen H, Zhang C, Li S, Liang Y, Lee LT, Aggarwal N, Wun KS, Liu J, Nadarajan SP, Weng C, Ling H, Tay JK, Wang DY, Yao SQ, Hwang IY, Lee YS, Chang MW. Prodrug-conjugated tumor-seeking commensals for targeted cancer therapy. Nat Commun 2024; 15:4343. [PMID: 38773197 PMCID: PMC11109227 DOI: 10.1038/s41467-024-48661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/08/2024] [Indexed: 05/23/2024] Open
Abstract
Prodrugs have been explored as an alternative to conventional chemotherapy; however, their target specificity remains limited. The tumor microenvironment harbors a range of microorganisms that potentially serve as tumor-targeting vectors for delivering prodrugs. In this study, we harness bacteria-cancer interactions native to the tumor microbiome to achieve high target specificity for prodrug delivery. We identify an oral commensal strain of Lactobacillus plantarum with an intrinsic cancer-binding mechanism and engineer the strain to enable the surface loading of anticancer prodrugs, with nasopharyngeal carcinoma (NPC) as a model cancer. The engineered commensals show specific binding to NPC via OppA-mediated recognition of surface heparan sulfate, and the loaded prodrugs are activated by tumor-associated biosignals to release SN-38, a chemotherapy compound, near NPC. In vitro experiments demonstrate that the prodrug-loaded microbes significantly increase the potency of SN-38 against NPC cell lines, up to 10-fold. In a mouse xenograft model, intravenous injection of the engineered L. plantarum leads to bacterial colonization in NPC tumors and a 67% inhibition in tumor growth, enhancing the efficacy of SN-38 by 54%.
Collapse
Affiliation(s)
- Haosheng Shen
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National Centre for Engineering Biology (NCEB), Singapore, Singapore
| | - Changyu Zhang
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Shengjie Li
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuanmei Liang
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National Centre for Engineering Biology (NCEB), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Ting Lee
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National Centre for Engineering Biology (NCEB), Singapore, Singapore
| | - Nikhil Aggarwal
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National Centre for Engineering Biology (NCEB), Singapore, Singapore
| | - Kwok Soon Wun
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National Centre for Engineering Biology (NCEB), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Liu
- Department of Otolaryngology, Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Saravanan Prabhu Nadarajan
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng Weng
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Hua Ling
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Wilmar International Limited, Singapore, Singapore
| | - Joshua K Tay
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - De Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - In Young Hwang
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore.
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Food, Chemical and Biotechnology, Singapore Institute of Technology, Singapore, Singapore.
| | - Yung Seng Lee
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Matthew Wook Chang
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore.
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- National Centre for Engineering Biology (NCEB), Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
6
|
Han M, Zhu H, Chen X, Luo X. 6-O-endosulfatases in tumor metastasis: heparan sulfate proteoglycans modification and potential therapeutic targets. Am J Cancer Res 2024; 14:897-916. [PMID: 38455409 PMCID: PMC10915330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Metastasis is the leading cause of cancer-associated mortality. Although advances in the targeted treatment and immunotherapy have improved the management of some cancers, the prognosis of metastatic cancers remains unsatisfied. Therefore, the specific mechanisms in tumor metastasis need further investigation. 6-O-endosulfatases (SULFs), comprising sulfatase1 (SULF1) and sulfatase 2 (SULF2), play pivotal roles in the post-synthetic modifications of heparan sulfate proteoglycans (HSPGs). Consequently, these extracellular enzymes can regulate a variety of downstream pathways by modulating HSPGs function. During the past decades, researchers have detected the expression of SULF1 and SULF2 in most cancers and revealed their roles in tumor progression and metastasis. Herein we reviewed the metastasis steps which SULFs participated in, elucidated the specific roles and mechanisms of SULFs in metastasis process, and discussed the effects of SULFs in different types of cancers. Moreover, we summarized the role of targeting SULFs in combination therapy to treat metastatic cancers, which provided some novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Mengzhen Han
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary DiseasesWuhan 430030, Hubei, China
| | - He Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary DiseasesWuhan 430030, Hubei, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary DiseasesWuhan 430030, Hubei, China
| | - Xin Luo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary DiseasesWuhan 430030, Hubei, China
| |
Collapse
|
7
|
Solaimuthu B, Khatib A, Tanna M, Karmi A, Hayashi A, Abu Rmaileh A, Lichtenstein M, Takoe S, Jolly MK, Shaul YD. The exostosin glycosyltransferase 1/STAT3 axis is a driver of breast cancer aggressiveness. Proc Natl Acad Sci U S A 2024; 121:e2316733121. [PMID: 38215181 PMCID: PMC10801894 DOI: 10.1073/pnas.2316733121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
The epithelial-mesenchymal transition (EMT) program is crucial for transforming carcinoma cells into a partially mesenchymal state, enhancing their chemoresistance, migration, and metastasis. This shift in cell state is tightly regulated by cellular mechanisms that are not yet fully characterized. One intriguing EMT aspect is the rewiring of the proteoglycan landscape, particularly the induction of heparan sulfate proteoglycan (HSPG) biosynthesis. This proteoglycan functions as a co-receptor that accelerates cancer-associated signaling pathways through its negatively-charged residues. However, the precise mechanisms through which EMT governs HSPG biosynthesis and its role in cancer cell plasticity remain elusive. Here, we identified exostosin glycosyltransferase 1 (EXT1), a central enzyme in HSPG biosynthesis, to be selectively upregulated in aggressive tumor subtypes and cancer cell lines, and to function as a key player in breast cancer aggressiveness. Notably, ectopic expression of EXT1 in epithelial cells is sufficient to induce HSPG levels and the expression of known mesenchymal markers, subsequently enhancing EMT features, including cell migration, invasion, and tumor formation. Additionally, EXT1 loss in MDA-MB-231 cells inhibits their aggressiveness-associated traits such as migration, chemoresistance, tumor formation, and metastasis. Our findings reveal that EXT1, through its role in HSPG biosynthesis, governs signal transducer and activator of transcription 3 (STAT3) signaling, a known regulator of cancer cell aggressiveness. Collectively, we present the EXT1/HSPG/STAT3 axis as a central regulator of cancer cell plasticity that directly links proteoglycan synthesis to oncogenic signaling pathways.
Collapse
Affiliation(s)
- Balakrishnan Solaimuthu
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Anees Khatib
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Mayur Tanna
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Abdelrahman Karmi
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Arata Hayashi
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Areej Abu Rmaileh
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Michal Lichtenstein
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Suranjana Takoe
- Department of Biological Sciences, Indian Institute of Science Education and Research, Berhampur760010, India
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore560012, India
| | - Yoav David Shaul
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| |
Collapse
|
8
|
Giusti V, Kaur G, Giusto E, Civiero L. Brain clearance of protein aggregates: a close-up on astrocytes. Mol Neurodegener 2024; 19:5. [PMID: 38229094 DOI: 10.1186/s13024-024-00703-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 01/18/2024] Open
Abstract
Protein misfolding and accumulation defines a prevailing feature of many neurodegenerative disorders, finally resulting in the formation of toxic intra- and extracellular aggregates. Intracellular aggregates can enter the extracellular space and be subsequently transferred among different cell types, thus spreading between connected brain districts.Although microglia perform a predominant role in the removal of extracellular aggregated proteins, mounting evidence suggests that astrocytes actively contribute to the clearing process. However, the molecular mechanisms used by astrocytes to remove misfolded proteins are still largely unknown.Here we first provide a brief overview of the progressive transition from soluble monomers to insoluble fibrils that characterizes amyloid proteins, referring to α-Synuclein and Tau as archetypical examples. We then highlight the mechanisms at the basis of astrocyte-mediated clearance with a focus on their potential ability to recognize, collect, internalize and digest extracellular protein aggregates. Finally, we explore the potential of targeting astrocyte-mediated clearance as a future therapeutic approach for the treatment of neurodegenerative disorders characterized by protein misfolding and accumulation.
Collapse
Affiliation(s)
| | - Gurkirat Kaur
- Department of Biology, University of Padova, Padua, Italy
| | | | - Laura Civiero
- IRCCS San Camillo Hospital, Venice, Italy.
- Department of Biology, University of Padova, Padua, Italy.
| |
Collapse
|
9
|
Li Y, Yang KD, Duan HY, Du YN, Ye JF. Phage-based peptides for pancreatic cancer diagnosis and treatment: alternative approach. Front Microbiol 2023; 14:1231503. [PMID: 37601380 PMCID: PMC10433397 DOI: 10.3389/fmicb.2023.1231503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Pancreatic cancer is a devastating disease with a high mortality rate and a lack of effective therapies. The challenges associated with early detection and the highly aggressive nature of pancreatic cancer have limited treatment options, underscoring the urgent need for better disease-modifying therapies. Peptide-based biotherapeutics have become an attractive area of research due to their favorable properties such as high selectivity and affinity, chemical modifiability, good tissue permeability, and easy metabolism and excretion. Phage display, a powerful technique for identifying peptides with high affinity and specificity for their target molecules, has emerged as a key tool in the discovery of peptide-based drugs. Phage display technology involves the use of bacteriophages to express peptide libraries, which are then screened against a target of interest to identify peptides with desired properties. This approach has shown great promise in cancer diagnosis and treatment, with potential applications in targeting cancer cells and developing new therapies. In this comprehensive review, we provide an overview of the basic biology of phage vectors, the principles of phage library construction, and various methods for binding affinity assessment. We then describe the applications of phage display in pancreatic cancer therapy, targeted drug delivery, and early detection. Despite its promising potential, there are still challenges to be addressed, such as optimizing the selection process and improving the pharmacokinetic properties of phage-based drugs. Nevertheless, phage display represents a promising approach for the development of novel targeted therapies in pancreatic cancer and other tumors.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Hao-yu Duan
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Ya-nan Du
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
10
|
Yang H, Wang L. Heparan sulfate proteoglycans in cancer: Pathogenesis and therapeutic potential. Adv Cancer Res 2023; 157:251-291. [PMID: 36725112 DOI: 10.1016/bs.acr.2022.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The heparan sulfate proteoglycans (HSPGs) are glycoproteins that consist of a proteoglycan "core" protein and covalently attached heparan sulfate (HS) chain. HSPGs are ubiquitously expressed in mammalian cells on the cell surface and in the extracellular matrix (ECM) and secretory vesicles. Within HSPGs, the protein cores determine when and where HSPG expression takes place, and the HS chains mediate most of HSPG's biological roles through binding various protein ligands, including cytokines, chemokines, growth factors and receptors, morphogens, proteases, protease inhibitors, and ECM proteins. Through these interactions, HSPGs modulate cell proliferation, adhesion, migration, invasion, and angiogenesis to display essential functions in physiology and pathology. Under physiological conditions, the expression and localization of HSPGs are finely regulated to orchestrate their physiological functions, and this is disrupted in cancer. The HSPG dysregulation elicits multiple oncogenic signaling, including growth factor signaling, ECM and Integrin signaling, chemokine and immune signaling, cancer stem cell, cell differentiation, apoptosis, and senescence, to prompt cell transformation, proliferation, tumor invasion and metastasis, tumor angiogenesis and inflammation, and immunotolerance. These oncogenic roles make HSPGs an attractive pharmacological target for anti-cancer therapy. Several therapeutic strategies have been under development, including anti-HSPG antibodies, peptides and HS mimetics, synthetic xylosides, and heparinase inhibitors, and shown promising anti-cancer efficacy. Therefore, much progress has been made in this line of study. However, it needs to bear in mind that the roles of HSPGs in cancer can be either oncogenic or tumor-suppressive, depending on the HSPG and the cancer cell type with the underlying mechanisms that remain obscure. Further studies need to address these to fill the knowledge gap and rationalize more efficient therapeutic targeting.
Collapse
Affiliation(s)
- Hua Yang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lianchun Wang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Bryd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| |
Collapse
|
11
|
Setting the stage for universal pharmacological targeting of the glycocalyx. CURRENT TOPICS IN MEMBRANES 2023; 91:61-88. [PMID: 37080681 DOI: 10.1016/bs.ctm.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
All cells in the human body are covered by a complex meshwork of sugars as well as proteins and lipids to which these sugars are attached, collectively termed the glycocalyx. Over the past few decades, the glycocalyx has been implicated in a range of vital cellular processes in health and disease. Therefore, it has attracted considerable interest as a therapeutic target. Considering its omnipresence and its relevance for various areas of cell biology, the glycocalyx should be a versatile platform for therapeutic intervention, however, the full potential of the glycocalyx as therapeutic target is yet to unfold. This might be attributable to the fact that glycocalyx alterations are currently discussed mainly in the context of specific diseases. In this perspective review, we shift the attention away from a disease-centered view of the glycocalyx, focusing on changes in glycocalyx state. Furthermore, we survey important glycocalyx-targeted drugs currently available and finally discuss future steps. We hope that this approach will inspire a unified, holistic view of the glycocalyx in disease, helping to stimulate novel glycocalyx-targeted therapy strategies.
Collapse
|
12
|
Enzymatic Digestion of Cell-surface Heparan Sulfate Alters the Radiation Response in Triple-negative Breast Cancer Cells. Arch Med Res 2022; 53:826-839. [PMID: 36411172 DOI: 10.1016/j.arcmed.2022.11.004] [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: 08/31/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIM Radiation resistance represents a major challenge in the treatment of breast cancer. As heparan sulfate (HS) chains are known to contribute to tumorigenesis, we aimed to investigate the interplay between HS degradation and radiation response in triple-negative breast cancer (TNBC) cells. METHODS HS chains were degraded in vitro as TNBC cells MDA-MB-231 and HCC1806 were treated with heparinase I and III. Subsequently, radioresistance was determined via colony formation assay after doses of 2, 4 and 6 Gy. Cell cycle profile, stem cell characteristics, expression of HS, activation of beta integrins, and apoptosis were determined by flow cytometry. Additionally, cell motility was analyzed via wound-healing assays, and expression and activation of FAK, CDK-6, Src, and Erk1/2 were quantified by western blot pre- and post-irradiation. Finally, the expression of cytokines was analyzed using a cytokine array. RESULTS Radiation promoted cell cycle changes, while heparinase treatment induced apoptosis in both cell lines. Colony formation assays showed significantly increased radio-resistance for both cell lines after degradation of HS. Cell migration was similarly upregulated after degradation of HS compared to controls. This effect was even more prominent after irradiation. Interestingly, FAK, a marker of radioresistance, was significantly activated in the heparinase-treated group. Additionally, we found Src to be dysregulated in MDA-MB-231 cells. Finally, we observed differential secretion of GRO, CXCL1, IGFBP1, IL8, Angiogenin, and Osteoprotegerin after HS degradation and radiotherapy. CONCLUSION Our results suggest an influence of HS chains on the development of radioresistance in TNBC.
Collapse
|
13
|
Noborn F, Nilsson J, Larson G. Site-specific glycosylation of proteoglycans: a revisited frontier in proteoglycan research. Matrix Biol 2022; 111:289-306. [PMID: 35840015 DOI: 10.1016/j.matbio.2022.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/11/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022]
Abstract
Proteoglycans (PGs), a class of carbohydrate-modified proteins, are present in essentially all metazoan organisms investigated to date. PGs are composed of glycosaminoglycan (GAG) chains attached to various core proteins and are important for embryogenesis and normal homeostasis. PGs exert many of their functions via their GAG chains and understanding the details of GAG-ligand interactions has been an essential part of PG research. Although PGs are also involved in many diseases, the number of GAG-related drugs used in the clinic is yet very limited, indicating a lack of detailed structure-function understanding. Structural analysis of PGs has traditionally been obtained by first separating the GAG chains from the core proteins, after which the two components are analyzed separately. While this strategy greatly facilitates the analysis, it precludes site-specific information and introduces either a "GAG" or a "core protein" perspective on the data interpretation. Mass-spectrometric (MS) glycoproteomic approaches have recently been introduced, providing site-specific information on PGs. Such methods have revealed a previously unknown structural complexity of the GAG linkage regions and resulted in identification of several novel CSPGs and HSPGs in humans and in model organisms, thereby expanding our view on PG complexity. In light of these findings, we discuss here if the use of such MS-based techniques, in combination with various functional assays, can also be used to expand our functional understanding of PGs. We have also summarized the site-specific information of all human PGs known to date, providing a theoretical framework for future studies on site-specific functional analysis of PGs in human pathophysiology.
Collapse
Affiliation(s)
- Fredrik Noborn
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Laboratory Medicine, Sundsvall County Hospital, Sweden.
| | - Jonas Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Proteomics Core Facility, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Kaczor-Kamińska M, Kamiński K, Wróbel M. Heparan Sulfate, Mucopolysaccharidosis IIIB and Sulfur Metabolism Disorders. Antioxidants (Basel) 2022; 11:antiox11040678. [PMID: 35453363 PMCID: PMC9026333 DOI: 10.3390/antiox11040678] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
Mucopolysaccharidosis, type IIIB (MPS IIIB) is a rare disease caused by mutations in the N-alpha-acetylglucosaminidase (NAGLU) gene resulting in decreased or absent enzyme activity. On the cellular level, the disorder is characterized by the massive lysosomal storage of heparan sulfate (HS)—one species of glycosaminoglycans. HS is a sulfur-rich macromolecule, and its accumulation should affect the turnover of total sulfur in cells; according to the studies presented here, it, indeed, does. The lysosomal degradation of HS in cells produces monosaccharides and inorganic sulfate (SO42−). Sulfate is a product of L-cysteine metabolism, and any disruption of its levels affects the entire L-cysteine catabolism pathway, which was first reported in 2019. It is known that L-cysteine level is elevated in cells with the Naglu−/− gene mutation and in selected tissues of individuals with MPS IIIB. The level of glutathione and the Naglu−/− cells’ antioxidant potential are significantly reduced, as well as the activity of 3-mercaptopyruvate sulfurtransferase (MPST, EC 2.8.1.2) and the level of sulfane sulfur-containing compounds. The direct reason is not yet known. This paper attempts to identify some of cause-and-effect correlations that may lead to this condition and identifies research directions that should be explored.
Collapse
Affiliation(s)
- Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
- Correspondence: ; Tel.: +48-12-422-7400
| | - Kamil Kamiński
- Department of Physical Chemistry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa St., 30-387 Krakow, Poland;
| | - Maria Wróbel
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
| |
Collapse
|
15
|
Piperigkou Z, Tzaferi K, Makrokanis G, Cheli K, Karamanos NK. The microRNA-cell surface proteoglycan axis in cancer progression. Am J Physiol Cell Physiol 2022; 322:C825-C832. [PMID: 35294845 DOI: 10.1152/ajpcell.00041.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proteoglycans consist one of the major extracellular matrix class of biomolecules that demonstrate nodal roles in cancer progression. Μodern diagnostic and therapeutic approaches include proteoglycan detection and pharmacological targeting in various cancers. Proteoglycans orchestrate critical signaling pathways for cancer development and progression through dynamic interactions with matrix components. It is well established that the epigenetic signatures of cancer cells play critical role in guiding their functional properties and metastatic potential. Secreted microRNAs (miRNAs) reside in a complex network with matrix proteoglycans, thus affecting cell-cell and cell-matrix communication. This mini-review aims to highlight current knowledge on the proteoglycan-mediated signaling cascades that regulate miRNA biogenesis in cancer. Moreover, the miRNA-mediated proteoglycan regulation during cancer progression and mechanistic aspects on the way that proteoglycans affect miRNA expression are presented. Recent advances on the role of cell surface proteoglycans in exosome biogenesis and miRNA packaging and expression are also discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Kyriaki Tzaferi
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - George Makrokanis
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konsatntina Cheli
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
16
|
Zha Z, Liu Y, Miao Y, Liao S, Wang SY, Tang H, Yin H. Preparation and characterization of 2-deacetyl-3-O-sulfo-heparosan and its antitumor effects via the fibroblast growth factor receptor pathway. Int J Biol Macromol 2022; 201:47-58. [PMID: 34998873 DOI: 10.1016/j.ijbiomac.2021.12.098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/24/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022]
Abstract
Heparosan, with a linear chain of disaccharide repeating units of → 4) β-D-glucuronic acid (GlcA) (1 → 4)-α-D-N-acetylglucosamine (GlcNAc) (1→, is a potential starting chemical for heparin synthesis. However, the chemoenzymatic synthesis of single-site sulfated heparosan and its antitumor activity have not been studied. In this study, 2-deacetyl-3-O-sulfo-heparosan (DSH) was prepared successively by the N-deacetylation chemical reaction and enzymatic modification of human 3-O-sulfotransferase-1 (3-OST-1). Structural characterization of DSH was shown the success of the sulfation with the sulfation degree of 0.87. High performance gel permeation chromatography (HPGPC) analysis revealed that DSH had only one symmetrical sharp peak with a molecular weight of 9.6334 × 104 Da. Biological function studies showed that DSH could inhibit tumor cell (A549, HepG2 and HCT116) viability and induce the apoptosis of A549 cells. Further in vitro mechanistic studies showed that DSH may induce apoptosis via the JNK signaling pathway, and the upstream signal of this process may be fibroblast growth factor receptors. These results indicated that DSH could be developed as one of a potential chemical for tumor treatment.
Collapse
Affiliation(s)
- Zhengqi Zha
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yaoyao Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yinghua Miao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Shiying Liao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Su-Yan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Huiling Tang
- Department of Pharmacy, Jiangsu Food and Pharmaceutical Science College, Huaian 223003, People's Republic of China.
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
17
|
Saif MW. From Screening to Treatment of Pancreatic Cancer: A Comprehensive Review. JOP : JOURNAL OF THE PANCREAS 2021; 22:70-79. [PMID: 34483790 PMCID: PMC8411391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is a devastating malignancy, associated with a grim prognosis, due to its silent presentation and lack of diagnostic tests. In addition, treatment options are limited to few agents, such as 5-FU, irinotecan, oxaliplatin, gemcitabine and nab-paclitaxel. METHODS We performed a literature search for relevant published clinical trials, abstracts of trials in progress and ongoing or planned trials for the treatment of APC using Pubmed.com, ClinicalTrials.gov and American Society of Clinical Oncology (ASCO) abstract search as sources. We present an in-depth analysis of the phase I-III clinical trials determining the role and efficacy of different modalities. We also describe rationale for future investigation. DISCUSSION Despite advances in first-line and second-line therapies for APC, median OS remains short of a year. We need collaborative efforts between the cooperative groups, institutions, community practices and industry to work together in enrolling these patients in clinical trials. In addition to use new technologies, such as organoids, we must pay attention to the palliative aspect of care for these patients from the beginning including nutritionist, social worker and supportive care health providers to assist with goals of care, symptom management and end of life discussions.
Collapse
Affiliation(s)
- Muhammad Wasif Saif
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine, Feinstein Institute for Medical Research, Lake Success, NY, USA
| |
Collapse
|
18
|
Nassar E, Hassan N, El-Ghonaimy EA, Hassan H, Abdullah MS, Rottke TV, Kiesel L, Greve B, Ibrahim SA, Götte M. Syndecan-1 Promotes Angiogenesis in Triple-Negative Breast Cancer through the Prognostically Relevant Tissue Factor Pathway and Additional Angiogenic Routes. Cancers (Basel) 2021; 13:cancers13102318. [PMID: 34066023 PMCID: PMC8150756 DOI: 10.3390/cancers13102318] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Triple-negative breast cancer is an aggressive subtype of breast cancer characterized by tumor angiogenesis and poor patient survival. Here, we analyzed the function of the cell surface molecule Syndecan-1 in tumor angiogenesis in a 3D cell culture system. As a novel finding, we demonstrate that downregulation of Syndecan-1 reduces angiogenesis by decreasing the amount of angiogenesis factors of the tissue factor pathway. Furthermore, we show that the components of this pathway are associated with the prognosis of breast cancer patients. Our study identifies Syndecan-1 and the tissue factor pathway as novel potential therapeutic targets in the aggressive triple-negative subtype of breast cancer, for which no targeted therapies are currently available. Abstract Triple-negative breast cancer (TNBC) is characterized by increased angiogenesis, metastasis, and poor survival. Dysregulation of the cell surface heparan sulfate proteoglycan and signaling co-receptor Syndecan-1 is linked to poor prognosis. To study its role in angiogenesis, we silenced Syndecan-1 in TNBC cell lines using a 3D human umbilical vein endothelial cell (HUVEC) co-culture system. Syndecan-1 siRNA depletion in SUM-149, MDA-MB-468, and MDA-MB-231 cells decreased HUVEC tubule network formation. Angiogenesis array revealed reduced VEGF-A and tissue factor (TF) in the Syndecan-1-silenced secretome. qPCR independently confirmed altered expression of F3, F7, F2R/PAR1, F2RL1/PAR2, VEGF-A, EDN1, IGFBP1, and IGFBP2 in SUM-149, MDA-MB-231, and MDA-MB-468 cells. ELISA revealed reduced secreted endothelin-1 (SUM-149, MDA-MB-468) and TF (all cell lines) upon Syndecan-1 depletion, while TF pathway inhibitor treatment impaired angiogenesis. Survival analysis of 3951 patients demonstrated that high expression of F3 and F7 are associated with better relapse-free survival, whereas poor survival was observed in TNBC and p53 mutant basal breast cancer (F3) and in ER-negative and HER2-positive breast cancer (F2R, F2RL1). STRING protein network analysis revealed associations of Syndecan-1 with VEGF-A and IGFBP1, further associated with the TF and ET-1 pathways. Our study suggests that TNBC Syndecan-1 regulates angiogenesis via the TF and additional angiogenic pathways and marks its constituents as novel prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Eyyad Nassar
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
| | - Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Eslam A. El-Ghonaimy
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
- Department of Zoology, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Hebatallah Hassan
- Department of Zoology, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Mahmoud Salah Abdullah
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Theresa V. Rottke
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
| | - Burkhard Greve
- Department of Radiotherapy and Radiooncology, University Hospital Münster, 48149 Münster, Germany;
| | - Sherif Abdelaziz Ibrahim
- Department of Zoology, Faculty of Science, Cairo University, 12613 Giza, Egypt;
- Correspondence: (S.A.I.); (M.G.)
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
- Correspondence: (S.A.I.); (M.G.)
| |
Collapse
|