1
|
Yang F, Zhao LY, Yang WQ, Chao S, Ling ZX, Sun BY, Wei LP, Zhang LJ, Yu LM, Cai GY. Quantitative proteomics and multi-omics analysis identifies potential biomarkers and the underlying pathological molecular networks in Chinese patients with multiple sclerosis. BMC Neurol 2024; 24:423. [PMID: 39478468 PMCID: PMC11526627 DOI: 10.1186/s12883-024-03926-3] [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: 06/28/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder caused by chronic inflammatory reactions in the central nervous system. Currently, little is known about the changes of plasma proteomic profiles in Chinese patients with MS (CpwMS) and its relationship with the altered profiles of multi-omics such as metabolomics and gut microbiome, as well as potential molecular networks that underlie the etiology of MS. To uncover the characteristics of proteomics landscape and potential multi-omics interaction networks in CpwMS, Plasma samples were collected from 22 CpwMS and 22 healthy controls (HCs) and analyzed using a Tandem Mass Tag (TMT)-based quantitative proteomics approach. Our results showed that the plasma proteomics pattern was significantly different in CpwMS compared to HCs. A total of 90 differentially expressed proteins (DEPs), such as LAMP1 and FCG2A, were identified in CpwMS plasma comparing to HCs. Furthermore, we also observed extensive and significant correlations between the altered proteomic profiles and the changes of metabolome, gut microbiome, as well as altered immunoinflammatory responses in MS-affected patients. For instance, the level of LAMP1 and ERN1 were significantly and positively correlated with the concentrations of metabolite L-glutamic acid and pro-inflammatory factor IL-17 (Padj < 0.05). However, they were negatively correlated with the amounts of other metabolites such as L-tyrosine and sphingosine 1-phosphate, as well as the concentrations of IL-8 and MIP-1α. This study outlined the underlying multi-omics integrated mechanisms that might regulate peripheral immunoinflammatory responses and MS progression. These findings are potentially helpful for developing new assisting diagnostic biomarker and therapeutic strategies for MS.
Collapse
Affiliation(s)
- Fan Yang
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China.
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Long-You Zhao
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China
| | - Wen-Qi Yang
- Department of Clinical Laboratory & Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shan Chao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Zong-Xin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo-Yao Sun
- Department of Clinical Laboratory & Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Li-Ping Wei
- Department of Clinical Laboratory & Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Li-Juan Zhang
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China
| | - Li-Mei Yu
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Guang-Yong Cai
- Lishui Key Laboratory of Brain Health and Severe Brain Disorders, Department of Rehabilitation & Clinical Laboratory, Lishui Second People's Hospital, Lishui, China.
| |
Collapse
|
2
|
Li JC, Huang ST, Feng F, Wang LJ, Chen TT, Li M, Liu LP. Relationship between low molecular weight heparin calcium therapy and prognosis in severe acute kidney injury in sepsis: Mendelian randomized analysis and retrospective study. Front Pharmacol 2024; 15:1389354. [PMID: 38915464 PMCID: PMC11194406 DOI: 10.3389/fphar.2024.1389354] [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: 02/23/2024] [Accepted: 05/22/2024] [Indexed: 06/26/2024] Open
Abstract
Background Sepsis-associated acute kidney injury (SA-AKI) poses an independent risk for mortality due to the absence of highly sensitive biomarkers and a specific treatment plan. Objective Investigate the association between low molecular weight heparin (LMWH) calcium therapy and prognosis in critically ill SA-AKI patients, and assess the causal relationship through Mendelian randomization (MR) analysis. Methods A single-center, retrospective, cross-sectional study included 90 SA-AKI patients and 30 septic patients without acute kidney injury (AKI) from the intensive care unit (ICU) of the First Hospital of Lanzhou University. SA-AKI patients were categorized into control or LMWH groups based on LMWH calcium usage. Primary outcome was renal function recovery, with secondary outcomes including 28-day mortality, ICU stay length, number of renal replacement therapy (RRT) recipients, and 90-day survival. MR and related sensitivity analyses explored causal effects. Results The combination of heparin-binding protein (HBP), heparanase (HPA), and neutrophil gelatinase-associated lipocalin (NGAL) demonstrated high diagnostic value for SA-AKI. MR analysis suggested a potential causal link between gene-predicted HBP and AKI (OR: 1.369, 95%CI: 1.040-1.801, p = 0.024). In the retrospective study, LMWH-treated patients exhibited improved renal function, reduced levels of HPA, HBP, Syndecan-1, and inflammation, along with enhanced immune function compared to controls. However, LMWH did not impact 28-day mortality, 90-day survival, or ICU stay length. Conclusion LMWH could enhance renal function in SA-AKI patients. MR analysis supports this causal link, underscoring the need for further validation in randomized controlled trials.
Collapse
Affiliation(s)
- Jian-Chun Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Shi-Tao Huang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Fei Feng
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Lin-Jun Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Ting-Ting Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Min Li
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Li-Ping Liu
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Jurjus R, Dosh L, Farhat R, Daccache T, El Masri J, Ghazi M, Hawi J, Leone A, Jurjus A. Lack of Syndecan-1 promotes the pathogenesis of experimental rheumatoid arthritis. Immunogenetics 2024; 76:145-154. [PMID: 38451352 DOI: 10.1007/s00251-024-01337-9] [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: 02/04/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
Syndecan-1 (Sdc-1), a transmembrane heparan sulfate protein, is implicated in several pathophysiological processes including rheumatoid arthritis (RA). The exact role of Syndican-1 in this autoimmune disease is still undetermined. This study explores the involvement level of Sdc-1 in the development of RA in a collagen II-induced arthritis mice model. RA was induced in two mice strains (wild-type BALB/c group and Sdc-1 knockout) by collagen II. Mice underwent regular clinical observations and scoring. After sacrifice, leg biopsies were taken from mice for histological examination, using a variety of stains. In addition, proteins were extracted, and molecular assessment of TNF-α was performed using the western blot technique. In the Sdc-1 knockout group, clinical scoring results showed a significantly more severe experimental RA; histology showed a significant increase in bone erosion, cartilage destruction, inflammation, and less granulated mast cells than the wild-type. In addition, molecular assessment of TNF-α showed more increase in expression in the Sdc-1 knockout models compared to the wild-type. Data suggest that lack of Sdc-1 enhances the inflammatory characteristics in RA. However, more molecular studies and investigations are needed to determine its exact role and possible mechanisms involved.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Arthritis, Experimental/immunology
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/immunology
- Collagen Type II/genetics
- Disease Models, Animal
- Mice, Inbred BALB C
- Mice, Knockout
- Syndecan-1/genetics
- Syndecan-1/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Rosalyn Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Laura Dosh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Rima Farhat
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Tatiana Daccache
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Jad El Masri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Maya Ghazi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Jihad Hawi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon
| | - Angelo Leone
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, Palermo, 90127, Italy
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Bliss Street, Beirut, 1107-2020, Lebanon.
| |
Collapse
|
4
|
Hinsinger G, Du Trieu De Terdonck L, Urbach S, Salvetat N, Rival M, Galoppin M, Ripoll C, Cezar R, Laurent-Chabalier S, Demattei C, Agherbi H, Castelnovo G, Lehmann S, Rigau V, Marin P, Thouvenot E. CD138 as a Specific CSF Biomarker of Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200230. [PMID: 38669615 PMCID: PMC11057439 DOI: 10.1212/nxi.0000000000200230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/30/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND AND OBJECTIVES The aim of this study was to identify novel biomarkers for multiple sclerosis (MS) diagnosis and prognosis, addressing the critical need for specific and prognostically valuable markers in the field. METHODS We conducted an extensive proteomic investigation, combining analysis of (1) CSF proteome from symptomatic controls, fast and slow converters after clinically isolated syndromes, and patients with relapsing-remitting MS (n = 10 per group) using label-free quantitative proteomics and (2) oligodendrocyte secretome changes under proinflammatory or proapoptotic conditions using stable isotope labeling by amino acids in cell culture. Proteins exhibiting differential abundance in both proteomic analyses were combined with other putative MS biomarkers, yielding a comprehensive list of 87 proteins that underwent quantification through parallel reaction monitoring (PRM) in a novel cohort, comprising symptomatic controls, inflammatory neurologic disease controls, and patients with MS at various disease stages (n = 10 per group). The 11 proteins that passed this qualification step were subjected to a new PRM assay within an expanded cohort comprising 158 patients with either MS at different disease stages or other inflammatory or noninflammatory neurologic disease controls. RESULTS This study unveiled a promising biomarker signature for MS, including previously established candidates, such as chitinase 3-like protein 1, chitinase 3-like protein 2, chitotriosidase, immunoglobulin kappa chain region C, neutrophil gelatinase-associated lipocalin, and CD27. In addition, we identified novel markers, namely cat eye syndrome critical region protein 1 (adenosine deaminase 2, a therapeutic target in multiple sclerosis) and syndecan-1, a proteoglycan, also known as plasma cell surface marker CD138 and acting as chitinase 3-like protein 1 receptor implicated in inflammation and cancer signaling. CD138 exhibited good diagnostic accuracy in distinguishing MS from inflammatory neurologic disorders (area under the curve [AUC] = 0.85, CI 0.75-0.95). CD138 immunostaining was also observed in the brains of patients with MS and cultured oligodendrocyte precursor cells but was absent in astrocytes. DISCUSSION These findings identify CD138 as a specific CSF biomarker for MS and suggest the selective activation of the chitinase 3-like protein 1/CD138 pathway within the oligodendrocyte lineage in MS. They offer promising prospects for improving MS diagnosis and prognosis by providing much-needed specificity and clinical utility. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that CD138 distinguishes multiple sclerosis from other inflammatory neurologic disorders with an AUC of 0.85 (95% CI 0.75-0.95).
Collapse
Affiliation(s)
- Geoffrey Hinsinger
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Lucile Du Trieu De Terdonck
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Serge Urbach
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Nicolas Salvetat
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Manon Rival
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Manon Galoppin
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Chantal Ripoll
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Renaud Cezar
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Sabine Laurent-Chabalier
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Christophe Demattei
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Hanane Agherbi
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Giovanni Castelnovo
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Sylvain Lehmann
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Valérie Rigau
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Philippe Marin
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| | - Eric Thouvenot
- From the IGF (G.H., L.D.T.D.T., S.U., M.R., M.G., C.R., P.M., E.T.), Université de Montpellier, CNRS, INSERM, Montpellier; Sys2Diag (N.S.), UMR 9005 CNRS / ALCEDIAG, Montpellier; Department of Neurology (M.R., H.A., G.C., E.T.), Nîmes University Hospital; IRMB (R.C.), Université de Montpellier, INSERM; Department of Immunology (R.C.), Nîmes University Hospital; Department of Biostatistics (S.L.-C., C.D.), Clinical Epidemiology, Public Health, and Innovation in Methodology, Nîmes University Hospital, Université de Montpellier; Biochemistry Department (S.L.), Hôpital Saint-Eloi; and Department of Pathology (V.R.), Montpellier University Hospital, France
| |
Collapse
|
5
|
Baucom MR, Weissman N, Price AD, England L, Schuster RM, Pritts TA, Goodman MD. Syndecan-1 as the Effect or Effector of the Endothelial Inflammatory Response? J Surg Res 2024; 295:611-618. [PMID: 38096775 DOI: 10.1016/j.jss.2023.10.010] [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: 03/01/2023] [Revised: 09/11/2023] [Accepted: 10/27/2023] [Indexed: 02/25/2024]
Abstract
INTRODUCTION Syndecan-1 is a heparan sulfate proteoglycan found in the glycocalyx of vascular endothelial cells. Serum levels of syndecan-1 have repeatedly been demonstrated to increase following traumatic injury and shock, but it is unclear whether syndecan-1 plays an active role in the inflammatory response or is simply a biomarker of a state of hypoperfusion. The aim of this study was to identify the role of syndecan-1 role in the inflammatory process in the absence of trauma. METHODS Male mice were randomized into five groups (n = 3). Four groups received increasing concentrations of syndecan-1 (1, 10, 100, and 1000pg/mL per blood volume) and a fifth group was given normal saline as a control via intravenous injection. These concentrations were selected based on previous syndecan-1 enzyme-linked immunosorbent assay data acquired following induced hemorrhagic shock in mice resulting in serum levels of 10-6000 pg/mL. Mice from each group were sacrificed at 1-, 4-, and 24-h time points for serum biomarker evaluation. A multiplex enzyme-linked immunosorbent assay was performed to analyze proinflammatory cytokines and chemokines including interleukin (IL)-1a, IL-1b, IL-2, IL-3, IL-4, IL-6, IL-10, IL-12, IL-17, monocyte chemoattractant protein-1, TNF-α, macrophage inflammatory protein-1α, granulocyte-macrophage colony-stimulating factor, and normal T cell expressed and presumably secreted levels. Whole blood was analyzed via rotational thromboelastometry in a separate group of mice dosed with syndecan-1 at 1000 pg/mL and compared to sham mice at 1 h. RESULTS Tumor necrosis factor-α was significantly elevated in the 1000 pg/mL group compared to sham animals. There were no significant changes in IL-1a, IL-1b, IL-2, IL-3, IL-4, IL-6, IL-10, IL-12, monocyte chemoattractant protein--1, macrophage inflammatory protein-1α, granulocyte-macrophage colony-stimulating factor, or normal T cell expressed and presumably secretedat 1, 4, and 24 h for any group when compared to mice receiving saline alone. No significant differences were noted in coagulability between the 1000 pg/mL syndecan-1 group and shams at 1 h CONCLUSIONS: Inflammatory cytokine concentrations did not change with increasing dosage of syndecan-1 within mice at any timepoint, except for an acute change in tumor necrosis factor-α which was transient. Based on our results, syndecan-1 appears to be a biomarker for inflammation rather than an active participant in eliciting an inflammatory response. Further research will focus on the role of syndecan-1 following hemorrhagic shock.
Collapse
Affiliation(s)
- Matthew R Baucom
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | - Adam D Price
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Lisa England
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | - Timothy A Pritts
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | |
Collapse
|
6
|
Baucom MR, Wallen TE, Ammann AM, England LG, Schuster RM, Pritts TA, Goodman MD. Blood component resuscitative strategies to mitigate endotheliopathy in a murine hemorrhagic shock model. J Trauma Acute Care Surg 2023; 95:21-29. [PMID: 37012625 PMCID: PMC10330005 DOI: 10.1097/ta.0000000000003942] [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] [Indexed: 04/05/2023]
Abstract
BACKGROUND Resuscitation with plasma components has been shown to improve endotheliopathy induced by hemorrhagic shock, but the optimal resuscitation strategy to preserve the endothelial glycocalyx has yet to be defined. The aim of this study was to determine if resuscitation with lactated Ringer's (LR), whole blood (WB), packed red blood cells (RBCs), platelet-rich plasma (PRP), platelet poor plasma, balanced RBC:PRP (1:1), or day 14 (d14) RBC would best minimize endothelial damage following shock. METHODS Male C57BL/6 mice were hemorrhaged to a goal mean arterial pressure of 25 mm Hg for 1 hour. Unshocked sham mice served as controls. Mice were then resuscitated with equal volumes of LR, WB, RBC, PRP, platelet poor plasma, 1:1, or d14 RBC and then sacrificed at 1, 4, or 24 hours (n = 5). Serum was analyzed for syndecan-1, ubiquitin C-terminal hydrolase L1, and cytokine concentrations. Lungs underwent syndecan-1 immunostaining, and lung injury scores were calculated after hematoxylin and eosin. Proteolytic cleavage of the endothelial glycocalyx was assessed by serum matrix metalloprotease 9 levels. RESULTS Serum syndecan-1 and ubiquitin C-terminal hydrolase L1 levels were significantly increased following resuscitation with d14 RBC compared with other groups. Early elevation in lung syndecan-1 staining was noted in LR-treated mice, while d14 mice showed decreased staining compared with sham mice following shock. Lung injury scores were significantly elevated 4 hours after resuscitation with LR and d14 RBC compared with WB. Serum matrix metalloprotease 9 levels were significantly increased at 1 and 4 hours in d14 mice compared with sham mice. Systemic inflammation was increased in animals receiving LR, 1:1, or d14 RBC. CONCLUSION Resuscitation with WB following hemorrhagic shock reduces endothelial syndecan-1 shedding and mitigates lung injury. Aged RBC and LR fail to attenuate endothelial injury following hemorrhagic shock. Further research will be necessary to determine the effect of each of these resuscitative fluids in a hemorrhagic shock model with the addition of tissue injury.
Collapse
Affiliation(s)
- Matthew R Baucom
- From the Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | | | | | | | | | | |
Collapse
|
7
|
Revisiting the Syndecans: Master Signaling Regulators with Prognostic and Targetable Therapeutic Values in Breast Carcinoma. Cancers (Basel) 2023; 15:cancers15061794. [PMID: 36980680 PMCID: PMC10046401 DOI: 10.3390/cancers15061794] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Syndecans (SDC1 to 4), a family of cell surface heparan sulfate proteoglycans, are frequently expressed in mammalian tissues. SDCs are aberrantly expressed either on tumor or stromal cells, influencing cancer initiation and progression through their pleiotropic role in different signaling pathways relevant to proliferation, cell-matrix adhesion, migration, invasion, metastasis, cancer stemness, and angiogenesis. In this review, we discuss the key roles of SDCs in the pathogenesis of breast cancer, the most common malignancy in females worldwide, focusing on the prognostic significance and molecular regulators of SDC expression and localization in either breast tumor tissue or its microenvironmental cells and the SDC-dependent epithelial–mesenchymal transition program. This review also highlights the molecular mechanisms underlying the roles of SDCs in regulating breast cancer cell behavior via modulation of nuclear hormone receptor signaling, microRNA expression, and exosome biogenesis and functions, as well as summarizing the potential of SDCs as promising candidate targets for therapeutic strategies against breast cancer.
Collapse
|
8
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
9
|
Purushothaman A, Mohajeri M, Lele TP. The role of glycans in the mechanobiology of cancer. J Biol Chem 2023; 299:102935. [PMID: 36693448 PMCID: PMC9930169 DOI: 10.1016/j.jbc.2023.102935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Although cancer is a genetic disease, physical changes such as stiffening of the extracellular matrix also commonly occur in cancer. Cancer cells sense and respond to extracellular matrix stiffening through the process of mechanotransduction. Cancer cell mechanotransduction can enhance cancer-promoting cell behaviors such as survival signaling, proliferation, and migration. Glycans, carbohydrate-based polymers, have recently emerged as important mediators and/or modulators of cancer cell mechanotransduction. Stiffer tumors are characterized by increased glycan content on cancer cells and their associated extracellular matrix. Here we review the role of cancer-associated glycans in coupled mechanical and biochemical alterations during cancer progression. We discuss the recent evidence on how increased expression of different glycans, in the form of glycoproteins and proteoglycans, contributes to both mechanical changes in tumors and corresponding cancer cell responses. We conclude with a summary of emerging tools that can be used to modify glycans for future studies in cancer mechanobiology.
Collapse
Affiliation(s)
- Anurag Purushothaman
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA.
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Tanmay P Lele
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA; Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA; Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA; Department of Translational Medical Sciences, Texas A&M University, Houston, Texas, USA.
| |
Collapse
|
10
|
Su X, Wang B, Zhou Z, Li Z, Tong S, Chen S, Zhang N, Liu S, Zhang M. A positive feedback loop of heparanase/syndecan1/nerve growth factor regulates cancer pain progression. Korean J Pain 2023; 36:60-71. [PMID: 36536517 PMCID: PMC9812689 DOI: 10.3344/kjp.22277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 12/24/2022] Open
Abstract
Background The purpose of this research was to assess the role of heparanase (HPSE)/syndecan1 (SDC1)/nerve growth factor (NGF) on cancer pain from melanoma. Methods The influence of HPSE on the biological function of melanoma cells and cancer pain in a mouse model was evaluated. Immunohistochemical staining was used to analyze HPSE and SDC1. HPSE, NGF, and SDC1 were detected using western blot. Inflammatory factors were detected using ELISA assay. Results HPSE promoted melanoma cell viability, proliferation, migration, invasion, and tumor growth, as well as cancer pain, while SST0001 treatment reversed the promoting effect of HPSE. HPSE up-regulated NGF, and NGF feedback promoted HPSE. High expression of NGF reversed the inhibitory effect of HPSE down-regulation on melanoma cell phenotype deterioration, including cell viability, proliferation, migration, and invasion. SST0001 down-regulated SDC1 expression. SDC1 reversed the inhibitory effect of SST0001 on cancer pain. Conclusions The results showed that HPSE promoted melanoma development and cancer pain by interacting with NGF/SDC1. It provides new insights to better understand the role of HPSE in melanoma and also provides a new direction for cancer pain treatment.
Collapse
Affiliation(s)
- Xiaohu Su
- Department of Anesthesiology, Suqian First People’s Hospital, Suqian City, Jiangsu Province, China
| | - Bingwu Wang
- Cancer Institute, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Zhaoyun Zhou
- Department of Anesthesiology, Tai’an Central Hospital, Tai’an City, Shandong Province, China
| | - Zixian Li
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Song Tong
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Simin Chen
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Nan Zhang
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Su Liu
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Maoyin Zhang
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China,Correspondence: Maoyin Zhang Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, No. 99, Huaihai West Road, Quanshan District, Xuzhou City, Jiangsu Province 221002, China, Tel: +86-18168777315, Fax: +86-0516-85805911, E-mail:
| |
Collapse
|
11
|
Yarimizu K, Nakane M, Onodera Y, Matsuuchi T, Suzuki H, Yoshioka M, Kudo M, Kawamae K. Prognostic Value of Antithrombin Activity Levels in the Early Phase of Intensive Care: A 2-Center Retrospective Cohort Study. Clin Appl Thromb Hemost 2023; 29:10760296231218711. [PMID: 38099709 PMCID: PMC10725115 DOI: 10.1177/10760296231218711] [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: 06/29/2023] [Revised: 10/19/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
To investigate the relationship between antithrombin (AT) activity level and prognosis in patients requiring intensive care. Patients whose AT activity was measured within 24 h of intensive care unit (ICU) admission were enrolled for analysis. The primary endpoint was mortality at discharge. Prognostic accuracy was examined using receiver operating characteristic (ROC) curves and cox hazard regression analysis. Patients were divided into 6 groups based on predicted mortality, and a χ2 independence test was performed on the prognostic value of AT activity for each predicted mortality; P < .05 was considered significant. A total of 281 cases were analyzed. AT activity was associated with mortality at discharge (AT% [interquartile range, IQR]): survivor group, 69 (56-86) versus nonsurvivor group, 56 (44-73), P = .0003). We found an increasing risk for mortality in both the lowest level of AT activity (<50%; hazard ratio [HR] 2.43, 95% confidence interval [CI] 1.20-4.89, P = .01) and the middle-low level of AT activity (≥ 50% and < 70%; HR 2.06, 95% CI 1.06-4.02, P = .03), compared with the normal AT activity level (≥ 70%). ROC curve analysis showed that the prediction accuracy of AT was an area under the curve (AUC) of 0.66 (cutoff 58%, sensitivity 61.4%, specificity 68.2%, P = .0003). AT activity was significantly prognostic in the group with 20% to 50% predicted mortality (AUC 0.74, sensitivity: 24.0%-55.5%, specificity: 83.3%-93.0%). An early decrease in AT activity level in ICU patients may be a predictor of mortality at discharge.
Collapse
Affiliation(s)
- Kenya Yarimizu
- Department of Anesthesiology, Yamagata University Hospital, Yamagata, Japan
| | - Masaki Nakane
- Department of Emergency and Critical Care Medicine, Yamagata University Hospital, Yamagata, Japan
| | - Yu Onodera
- Department of Anesthesiology, Yamagata University Hospital, Yamagata, Japan
| | - Taro Matsuuchi
- Department of Anesthesia, Nihonkai General Hospital, Yamagata, Japan
| | - Hiroto Suzuki
- Department of Anesthesiology, Yamagata University Hospital, Yamagata, Japan
| | - Masatomo Yoshioka
- Department of Emergency Medicine, Nihonkai General Hospital, Yamagata, Japan
| | - Masaya Kudo
- Department of Anesthesia, Nihonkai General Hospital, Yamagata, Japan
| | - Kaneyuki Kawamae
- Department of Anesthesiology, Yamagata University Hospital, Yamagata, Japan
| |
Collapse
|
12
|
Masola V, Franchi M, Zaza G, Atsina FM, Gambaro G, Onisto M. Heparanase regulates EMT and cancer stem cell properties in prostate tumors. Front Oncol 2022; 12:918419. [PMID: 35965510 PMCID: PMC9363836 DOI: 10.3389/fonc.2022.918419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer displays a certain phenotypic plasticity that allows for the transition of cells from the epithelial to the mesenchymal state. This process, known as epithelial–mesenchymal transition (EMT), is one of the factors that give the tumor cells greater invasive and migratory capacity with subsequent formation of metastases. In addition, many cancers, including prostate cancer, are derived from a cell population that shows the properties of stem cells. These cells, called cancer stem cells (CSCs) or tumor-initiating cells, not only initiate the tumor process and growth but are also able to mediate metastasis and drug resistance. However, the impact of EMT and CSCs in prostate cancer progression and patient survival is still far from fully understood. Heparanase (HPSE), the sole mammalian endoglycosidase capable of degrading heparan sulfate (HS), is also involved in prostate cancer progression. We had previously proved that HPSE regulates EMT in non-cancerous pathologies. Two prostate cancer cell lines (DU145 and PC3) were silenced and overexpressed for HPSE. Expression of EMT and stemness markers was evaluated. Results showed that the expression of several EMT markers are modified by HPSE expression in both the prostate cancer cell lines analyzed. In the same way, the stemness markers and features are also modulated by HPSE expression. Taken together, the present findings seem to prove a new mechanism of action of HPSE in sustaining prostate cancer growth and diffusion. As for other tumors, these results highlight the importance of HPSE as a potential pharmacological target in prostate cancer treatment.
Collapse
Affiliation(s)
- Valentina Masola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Maurizio Onisto, ; Valentina Masola,
| | - Marco Franchi
- Department of Life Quality Sciences, University of Bologna, Rimini, Italy
| | - Gianluigi Zaza
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | | - Giovanni Gambaro
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Maurizio Onisto, ; Valentina Masola,
| |
Collapse
|
13
|
Yin T, Hao J, Jiang Q, Xu X, Xu B, Lv H, Liu W, Xiao Y, Jiao L, Wang J, Zhang L. Dynamics of intracranial and peripheral plasma Syndecan‐1 after ischemic stroke with large vessel occlusion. CNS Neurosci Ther 2022; 28:1648-1650. [PMID: 35790077 PMCID: PMC9437231 DOI: 10.1111/cns.13898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tengkun Yin
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| | - Jiheng Hao
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| | - Qunlong Jiang
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| | - Xin Xu
- Department of Neurosurgery Xuanwu Hospital, Capital Medical University Beijing China
| | - Bin Xu
- Department of Neurosurgery Xuanwu Hospital, Capital Medical University Beijing China
- School of Clinical Medicine Weifang Medical College Weifang China
| | - Hang Lv
- Department of Neurosurgery Xuanwu Hospital, Capital Medical University Beijing China
- School of Clinical Medicine Weifang Medical College Weifang China
| | - Weidong Liu
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| | - Yilei Xiao
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| | - Liqun Jiao
- Department of Neurosurgery Xuanwu Hospital, Capital Medical University Beijing China
| | - Jiyue Wang
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| | - Liyong Zhang
- From the Department of Neurosurgery Liaocheng People's Hospital Liaocheng China
| |
Collapse
|
14
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Oravecz O, Balogh A, Romero R, Xu Y, Juhasz K, Gelencser Z, Xu Z, Bhatti G, Pique-Regi R, Peterfia B, Hupuczi P, Kovalszky I, Murthi P, Tarca AL, Papp Z, Matko J, Than NG. Proteoglycans: Systems-Level Insight into Their Expression in Healthy and Diseased Placentas. Int J Mol Sci 2022; 23:5798. [PMID: 35628608 PMCID: PMC9147780 DOI: 10.3390/ijms23105798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/04/2023] Open
Abstract
Proteoglycan macromolecules play key roles in several physiological processes (e.g., adhesion, proliferation, migration, invasion, angiogenesis, and apoptosis), all of which are important for placentation and healthy pregnancy. However, their precise roles in human reproduction have not been clarified. To fill this gap, herein, we provide an overview of the proteoglycans' expression and role in the placenta, in trophoblast development, and in pregnancy complications (pre-eclampsia, fetal growth restriction), highlighting one of the most important members of this family, syndecan-1 (SDC1). Microarray data analysis showed that of 34 placentally expressed proteoglycans, SDC1 production is markedly the highest in the placenta and that SDC1 is the most upregulated gene during trophoblast differentiation into the syncytiotrophoblast. Furthermore, placental transcriptomic data identified dysregulated proteoglycan genes in pre-eclampsia and in fetal growth restriction, including SDC1, which is supported by the lower concentration of syndecan-1 in maternal blood in these syndromes. Overall, our clinical and in vitro studies, data analyses, and literature search pointed out that proteoglycans, as important components of the placenta, may regulate various stages of placental development and participate in the maintenance of a healthy pregnancy. Moreover, syndecan-1 may serve as a useful marker of syncytialization and a prognostic marker of adverse pregnancy outcomes. Further studies are warranted to explore the role of proteoglycans in healthy and complicated pregnancies, which may help in diagnostic or therapeutic developments.
Collapse
Affiliation(s)
- Orsolya Oravecz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Kata Juhasz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Zsolt Gelencser
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Zhonghui Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Balint Peterfia
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | | | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| | - Padma Murthi
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia;
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women’s Hospital, Parkville, VIC 3502, Australia
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA
| | - Zoltan Papp
- Maternity Private Clinic, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
| | - Janos Matko
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
- Maternity Private Clinic, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| |
Collapse
|
16
|
D'Arcy C, Zimmermann CC, Espinoza-Sanchez NA, Greve B, Schmidt A, Kiesel L, von Wahlde MK, Götte M. The heparan sulphate proteoglycan Syndecan-1 (CD138) regulates tumour progression in a 3D model of ductal carcinoma in situ of the breast. IUBMB Life 2022; 74:955-968. [PMID: 35587107 DOI: 10.1002/iub.2623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/25/2022] [Indexed: 11/07/2022]
Abstract
Ductal carcinoma in situ (DCIS) is a form of breast cancer that is restricted to the lactiferous ducts and has not yet invaded the surrounding breast tissue. Dysregulation of the transmembrane heparan sulphate proteoglycan Syndecan-1 (Sdc-1) plays a role in tumour progression of invasive breast cancer (IBC). In DCIS, Sdc-1, c-Met and E-cadherin are part of a proangiogenic expression signature. In this study, we employed a siRNA knockdown approach in the DCIS model cell line MCF10A DCIS.com to investigate a potential connection between Sdc-1 and epithelial mesenchymal transition (EMT), proteolysis and the Rho kinase pathway. Analysis of gene expression data of the TNMplot.com database revealed that Sdc-1 expression was higher in primary breast tumours compared to metastases. The impact of Sdc-1-depletion on the cellular phenotype was investigated in a Matrigel-based three-dimensional cell culture model. Sdc-1 depletion resulted in the formation of larger spheroids and the formation of invasive protrusions. Application of matrix metalloproteinase (MMP) and Rho kinase inhibitors could block the Sdc-1-induced phenotype. qPCR analysis of Sdc-1-depleted cells in two-dimensional culture revealed upregulated expression of the EMT-markers CDH1, FN-1, CLDN1, the proteolysis markers MMP3, and MMP9, and HPSE, while MMP2, VIM and ROCK-2 were downregulated. Immunocytochemistry confirmed upregulation of MMP9 and fibronectin, the latter being particular prominent after ROCK inhibition. STRING analysis confirmed an interaction of the investigated gene products at the protein level. Our results suggest that diminished Sdc-1 expression plays a role in DCIS progression to IBC through deregulation of proteolytic factors and a partial EMT.
Collapse
Affiliation(s)
- Christopher D'Arcy
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | | | - Nancy Adriana Espinoza-Sanchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Department of Radiotherapy-Radiooncology, Department of Radiotherapy and Radiooncology, University Hospital Münster, Münster, Germany
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Department of Radiotherapy and Radiooncology, University Hospital Münster, Münster, Germany
| | - Annika Schmidt
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | | | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
17
|
Advances in the Immunomodulatory Properties of Glycoantigens in Cancer. Cancers (Basel) 2022; 14:cancers14081854. [PMID: 35454762 PMCID: PMC9032556 DOI: 10.3390/cancers14081854] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/28/2022] Open
Abstract
Simple Summary This work reviews the role of aberrant glycosylation in cancer cells during tumour growth and spreading, as well as in immune evasion. The interaction of tumour-associated glycans with the immune system through C-type lectin receptors can favour immune escape but can also provide opportunities to develop novel tumour immunotherapy strategies. This work highlights the main findings in this area and spotlights the challenges that remain to be investigated. Abstract Aberrant glycosylation in tumour progression is currently a topic of main interest. Tumour-associated carbohydrate antigens (TACAs) are expressed in a wide variety of epithelial cancers, being both a diagnostic tool and a potential treatment target, as they have impact on patient outcome and disease progression. Glycans affect both tumour-cell biology properties as well as the antitumor immune response. It has been ascertained that TACAs affect cell migration, invasion and metastatic properties both when expressed by cancer cells or by their extracellular vesicles. On the other hand, tumour-associated glycans recognized by C-type lectin receptors in immune cells possess immunomodulatory properties which enable tumour growth and immune response evasion. Yet, much remains unknown, concerning mechanisms involved in deregulation of glycan synthesis and how this affects cell biology on a major level. This review summarises the main findings to date concerning how aberrant glycans influence tumour growth and immunity, their application in cancer treatment and spotlights of unanswered challenges remaining to be solved.
Collapse
|
18
|
Masola V, Greco N, Gambaro G, Franchi M, Onisto M. Heparanase as active player in endothelial glycocalyx remodeling. Matrix Biol Plus 2022; 13:100097. [PMID: 35036899 PMCID: PMC8749438 DOI: 10.1016/j.mbplus.2021.100097] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
The surface of all animal cells is coated with a layer of carbohydrates linked in various ways to the outer side of the plasma membrane. These carbohydrates are mainly bound to proteins in the form of glycoproteins and proteoglycans and together with the glycolipids constitute the so-called glycocalyx. In particular, the endothelial glycocalyx that covers the luminal layer of the endothelium is composed of glycosaminoglycans (heparan sulphate -HS and hyaluronic acid -HA), proteoglycans (syndecans and glypicans) and adsorbed plasma proteins. Thanks to its ability to absorb water, this structure contributes to making the surface of the vessels slippery but at the same time acts by modulating the mechano-transduction of the vessels, the vascular permeability and the adhesion of leukocytes in thus regulating several physiological and pathological events. Among the various enzymes involved in the degradation of the glycocalyx, heparanase (HPSE) has been shown to be particularly involved. This enzyme is responsible for the cutting of heparan sulfate (HS) chains at the level of the proteoglycans of the endothelial glycocalyx whose dysfunction appears to have a role in organ fibrosis, sepsis and viral infection. In this mini-review, we describe the mechanisms by which HPSE contributes to glycocalyx remodeling and then examine the role of glycocalyx degradation in the development of pathological conditions and pharmacological strategies to preserve glycocalyx during disease pathogenesis.
Collapse
Affiliation(s)
- Valentina Masola
- Renal Unit, Dept. of Medicine, University Hospital of Verona, Verona, Italy.,Dept. of Biomedical Sciences, University of Padova, Padua, Italy
| | - Nicola Greco
- Dept. of Biomedical Sciences, University of Padova, Padua, Italy
| | - Giovanni Gambaro
- Renal Unit, Dept. of Medicine, University Hospital of Verona, Verona, Italy
| | - Marco Franchi
- Dept. of Life Quality Sciences, University of Bologna, Rimini, Italy
| | - Maurizio Onisto
- Dept. of Biomedical Sciences, University of Padova, Padua, Italy
| |
Collapse
|
19
|
Rapraeger AC. Syndecans and Their Synstatins: Targeting an Organizer of Receptor Tyrosine Kinase Signaling at the Cell-Matrix Interface. Front Oncol 2021; 11:775349. [PMID: 34778093 PMCID: PMC8578902 DOI: 10.3389/fonc.2021.775349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/27/2021] [Indexed: 01/11/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) and integrin matrix receptors have well-established roles in tumor cell proliferation, invasion and survival, often functioning in a coordinated fashion at sites of cell-matrix adhesion. Central to this coordination are syndecans, another class of matrix receptor, that organize RTKs and integrins into functional units, relying on docking motifs in the syndecan extracellular domains to capture and localize RTKs (e.g., EGFR, IGF-1R, VEGFR2, HER2) and integrins (e.g., αvβ3, αvβ5, α4β1, α3β1, α6β4) to sites of adhesion. Peptide mimetics of the docking motifs in the syndecans, called “synstatins”, prevent assembly of these receptor complexes, block their signaling activities and are highly effective against tumor cell invasion and survival and angiogenesis. This review describes our current understanding of these four syndecan-coupled mechanisms and their inhibitory synstatins (SSTNIGF1R, SSTNVEGFR2, SSTNVLA-4, SSTNEGFR and SSTNHER2).
Collapse
Affiliation(s)
- Alan C Rapraeger
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
20
|
Syndecan-1 Depletion Has a Differential Impact on Hyaluronic Acid Metabolism and Tumor Cell Behavior in Luminal and Triple-Negative Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms22115874. [PMID: 34070901 PMCID: PMC8198019 DOI: 10.3390/ijms22115874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/18/2022] Open
Abstract
Glycosaminoglycans (GAGs) and proteoglycans (PGs) are major components of the glycocalyx. The secreted GAG and CD44 ligand hyaluronic acid (HA), and the cell surface PG syndecan-1 (Sdc-1) modulate the expression and activity of cytokines, chemokines, growth factors, and adhesion molecules, acting as critical regulators of tumor cell behavior. Here, we studied the effect of Sdc-1 siRNA depletion and HA treatment on hallmark processes of cancer in breast cancer cell lines of different levels of aggressiveness. We analyzed HA synthesis, and parameters relevant to tumor progression, including the stem cell phenotype, Wnt signaling constituents, cell cycle progression and apoptosis, and angiogenic markers in luminal MCF-7 and triple-negative MDA-MB-231 cells. Sdc-1 knockdown enhanced HAS-2 synthesis and HA binding in MCF-7, but not in MDA-MB-231 cells. Sdc-1-depleted MDA-MB-231 cells showed a reduced CD24-/CD44+ population. Furthermore, Sdc-1 depletion was associated with survival signals in both cell lines, affecting cell cycle progression and apoptosis evasion. These changes were linked to the altered expression of KLF4, MSI2, and miR-10b and differential changes in Erk, Akt, and PTEN signaling. We conclude that Sdc-1 knockdown differentially affects HA metabolism in luminal and triple-negative breast cancer model cell lines and impacts the stem phenotype, cell survival, and angiogenic factors.
Collapse
|
21
|
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
|
22
|
Syndecan-1 (CD138), Carcinomas and EMT. Int J Mol Sci 2021; 22:ijms22084227. [PMID: 33921767 PMCID: PMC8072910 DOI: 10.3390/ijms22084227] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cell surface proteoglycans are known to be important regulators of many aspects of cell behavior. The principal family of transmembrane proteoglycans is the syndecans, of which there are four in mammals. Syndecan-1 is mostly restricted to epithelia, and bears heparan sulfate chains that are capable of interacting with a large array of polypeptides, including extracellular matrix components and potent mediators of proliferation, adhesion and migration. For this reason, it has been studied extensively with respect to carcinomas and tumor progression. Frequently, but not always, syndecan-1 levels decrease as tumor grade, stage and invasiveness and dedifferentiation increase. This parallels experiments that show depletion of syndecan-1 can be accompanied by loss of cadherin-mediated adhesion. However, in some tumors, levels of syndecan-1 increase, but the characterization of its distribution is relevant. There can be loss of membrane staining, but acquisition of cytoplasmic and/or nuclear staining that is abnormal. Moreover, the appearance of syndecan-1 in the tumor stroma, either associated with its cellular component or the collagenous matrix, is nearly always a sign of poor prognosis. Given its relevance to myeloma progression, syndecan-1-directed antibody—toxin conjugates are being tested in clinical and preclinical trials, and may have future relevance to some carcinomas.
Collapse
|
23
|
Antiviral strategies should focus on stimulating the biosynthesis of heparan sulfates, not their inhibition. Life Sci 2021; 277:119508. [PMID: 33865880 PMCID: PMC8046744 DOI: 10.1016/j.lfs.2021.119508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022]
Abstract
Antiviral strategies for viruses that utilize proteoglycan core proteins (syndecans and glypicans) as receptors should focus on heparan sulfate (HS) biosynthesis rather than on inhibition of these sugar chains. Here, we show that heparin and certain xylosides, which exhibit in vitro viral entry inhibitory properties against HSV-1, HSV-2, HPV-16, HPV-31, HVB, HVC, HIV-1, HTLV-1, SARS-CoV-2, HCMV, DENV-1, and DENV-2, stimulated HS biosynthesis at the cell surface 2- to 3-fold for heparin and up to 10-fold for such xylosides. This is consistent with the hypothesis from a previous study that for core protein attachment, viruses are glycosylated at HS attachment sites (i.e., serine residues intended to receive the D-xylose molecule for initiating HS chains). Heparanase overexpression, endocytic entry, and syndecan shedding enhancement, all of which are observed during viral infection, lead to glycocalyx deregulation and appear to be direct consequences of this hypothesis. In addition to the appearance of type 2 diabetes and the degradation of HS observed during viral infection, we linked this hypothesis to that proposed in a previous publication.
Collapse
|
24
|
Vlodavsky I, Barash U, Nguyen HM, Yang SM, Ilan N. Biology of the Heparanase-Heparan Sulfate Axis and Its Role in Disease Pathogenesis. Semin Thromb Hemost 2021; 47:240-253. [PMID: 33794549 DOI: 10.1055/s-0041-1725066] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell surface proteoglycans are important constituents of the glycocalyx and participate in cell-cell and cell-extracellular matrix (ECM) interactions, enzyme activation and inhibition, and multiple signaling routes, thereby regulating cell proliferation, survival, adhesion, migration, and differentiation. Heparanase, the sole mammalian heparan sulfate degrading endoglycosidase, acts as an "activator" of HS proteoglycans, thus regulating tissue hemostasis. Heparanase is a multifaceted enzyme that together with heparan sulfate, primarily syndecan-1, drives signal transduction, immune cell activation, exosome formation, autophagy, and gene transcription via enzymatic and nonenzymatic activities. An important feature is the ability of heparanase to stimulate syndecan-1 shedding, thereby impacting cell behavior both locally and distally from its cell of origin. Heparanase releases a myriad of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate-heparanase axis plays pivotal roles in creating a permissive environment for cell proliferation, differentiation, and function, often resulting in the pathogenesis of diseases such as cancer, inflammation, endotheliitis, kidney dysfunction, tissue fibrosis, and viral infection.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
25
|
Berdiaki A, Neagu M, Giatagana EM, Kuskov A, Tsatsakis AM, Tzanakakis GN, Nikitovic D. Glycosaminoglycans: Carriers and Targets for Tailored Anti-Cancer Therapy. Biomolecules 2021; 11:395. [PMID: 33800172 PMCID: PMC8001210 DOI: 10.3390/biom11030395] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/25/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment (TME) is composed of cancerous, non-cancerous, stromal, and immune cells that are surrounded by the components of the extracellular matrix (ECM). Glycosaminoglycans (GAGs), natural biomacromolecules, essential ECM, and cell membrane components are extensively altered in cancer tissues. During disease progression, the GAG fine structure changes in a manner associated with disease evolution. Thus, changes in the GAG sulfation pattern are immediately correlated to malignant transformation. Their molecular weight, distribution, composition, and fine modifications, including sulfation, exhibit distinct alterations during cancer development. GAGs and GAG-based molecules, due to their unique properties, are suggested as promising effectors for anticancer therapy. Considering their participation in tumorigenesis, their utilization in drug development has been the focus of both industry and academic research efforts. These efforts have been developing in two main directions; (i) utilizing GAGs as targets of therapeutic strategies and (ii) employing GAGs specificity and excellent physicochemical properties for targeted delivery of cancer therapeutics. This review will comprehensively discuss recent developments and the broad potential of GAG utilization for cancer therapy.
Collapse
Affiliation(s)
- Aikaterini Berdiaki
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.B.); (E.-M.G.); (G.N.T.)
| | - Monica Neagu
- Department of Immunology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
| | - Eirini-Maria Giatagana
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.B.); (E.-M.G.); (G.N.T.)
| | - Andrey Kuskov
- Department of Technology of Chemical Pharmaceutical and Cosmetic Substances, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Aristidis M. Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - George N. Tzanakakis
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.B.); (E.-M.G.); (G.N.T.)
- Laboratory of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.B.); (E.-M.G.); (G.N.T.)
| |
Collapse
|
26
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 362] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
27
|
Gopal S, Arokiasamy S, Pataki C, Whiteford JR, Couchman JR. Syndecan receptors: pericellular regulators in development and inflammatory disease. Open Biol 2021; 11:200377. [PMID: 33561383 PMCID: PMC8061687 DOI: 10.1098/rsob.200377] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
The syndecans are the major family of transmembrane proteoglycans, usually bearing multiple heparan sulfate chains. They are present on virtually all nucleated cells of vertebrates and are also present in invertebrates, indicative of a long evolutionary history. Genetic models in both vertebrates and invertebrates have shown that syndecans link to the actin cytoskeleton and can fine-tune cell adhesion, migration, junction formation, polarity and differentiation. Although often associated as co-receptors with other classes of receptors (e.g. integrins, growth factor and morphogen receptors), syndecans can nonetheless signal to the cytoplasm in discrete ways. Syndecan expression levels are upregulated in development, tissue repair and an array of human diseases, which has led to the increased appreciation that they may be important in pathogenesis not only as diagnostic or prognostic agents, but also as potential targets. Here, their functions in development and inflammatory diseases are summarized, including their potential roles as conduits for viral pathogen entry into cells.
Collapse
Affiliation(s)
- Sandeep Gopal
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Samantha Arokiasamy
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Csilla Pataki
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - James R. Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - John R. Couchman
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
28
|
Rangarajan S, Richter JR, Richter RP, Bandari SK, Tripathi K, Vlodavsky I, Sanderson RD. Heparanase-enhanced Shedding of Syndecan-1 and Its Role in Driving Disease Pathogenesis and Progression. J Histochem Cytochem 2020; 68:823-840. [PMID: 32623935 PMCID: PMC7711244 DOI: 10.1369/0022155420937087] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/29/2020] [Indexed: 02/08/2023] Open
Abstract
Both heparanase and syndecan-1 are known to be present and active in disease pathobiology. An important feature of syndecan-1 related to its role in pathologies is that it can be shed from the surface of cells as an intact ectodomain composed of the extracellular core protein and attached heparan sulfate and chondroitin sulfate chains. Shed syndecan-1 remains functional and impacts cell behavior both locally and distally from its cell of origin. Shedding of syndecan-1 is initiated by a variety of stimuli and accomplished predominantly by the action of matrix metalloproteinases. The accessibility of these proteases to the core protein of syndecan-1 is enhanced, and shedding facilitated, when the heparan sulfate chains of syndecan-1 have been shortened by the enzymatic activity of heparanase. Interestingly, heparanase also enhances shedding by upregulating the expression of matrix metalloproteinases. Recent studies have revealed that heparanase-induced syndecan-1 shedding contributes to the pathogenesis and progression of cancer and viral infection, as well as other septic and non-septic inflammatory states. This review discusses the heparanase/shed syndecan-1 axis in disease pathogenesis and progression, the potential of targeting this axis therapeutically, and the possibility that this axis is widespread and of influence in many diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Israel Vlodavsky
- The University of Alabama at Birmingham, Birmingham, Alabama, and Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
29
|
Hassan N, Greve B, Espinoza-Sánchez NA, Götte M. Cell-surface heparan sulfate proteoglycans as multifunctional integrators of signaling in cancer. Cell Signal 2020; 77:109822. [PMID: 33152440 DOI: 10.1016/j.cellsig.2020.109822] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
Proteoglycans (PGs) represent a large proportion of the components that constitute the extracellular matrix (ECM). They are a diverse group of glycoproteins characterized by a covalent link to a specific glycosaminoglycan type. As part of the ECM, heparan sulfate (HS)PGs participate in both physiological and pathological processes including cell recruitment during inflammation and the promotion of cell proliferation, adhesion and motility during development, angiogenesis, wound repair and tumor progression. A key function of HSPGs is their ability to modulate the expression and function of cytokines, chemokines, growth factors, morphogens, and adhesion molecules. This is due to their capacity to act as ligands or co-receptors for various signal-transducing receptors, affecting pathways such as FGF, VEGF, chemokines, integrins, Wnt, notch, IL-6/JAK-STAT3, and NF-κB. The activation of those pathways has been implicated in the induction, progression, and malignancy of a tumor. For many years, the study of signaling has allowed for designing specific drugs targeting these pathways for cancer treatment, with very positive results. Likewise, HSPGs have become the subject of cancer research and are increasingly recognized as important therapeutic targets. Although they have been studied in a variety of preclinical and experimental models, their mechanism of action in malignancy still needs to be more clearly defined. In this review, we discuss the role of cell-surface HSPGs as pleiotropic modulators of signaling in cancer and identify them as promising markers and targets for cancer treatment.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Biotechnology Program, Department of Chemistry, Faculty of Science, Cairo University, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany.
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.
| |
Collapse
|