1
|
Seiler S, Rudolf F, Gomes FR, Pavlovic A, Nebel J, Seidenbecher CI, Foo LC. Astrocyte-derived factors regulate CNS myelination. Glia 2024; 72:2038-2060. [PMID: 39092473 DOI: 10.1002/glia.24596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
The role that astrocytes play in central nervous system (CNS) myelination is poorly understood. We investigated the contribution of astrocyte-derived factors to myelination and revealed a substantial overlap in the secretomes of human and rat astrocytes. Using in vitro myelinating co-cultures of primary retinal ganglion cells and cortical oligodendrocyte precursor cells, we discovered that factors secreted by resting astrocytes, but not reactive astrocytes, facilitated myelination. Soluble brevican emerged as a new enhancer of developmental myelination in vivo, CNS and its absence was linked to remyelination deficits following an immune-mediated damage in an EAE mouse model. The observed reduction of brevican expression in reactive astrocytes and human MS lesions suggested a potential link to the compromised remyelination characteristic of neurodegenerative diseases. Our findings suggested brevican's role in myelination may be mediated through interactions with binding partners such as contactin-1 and tenascin-R. Proteomic analysis of resting versus reactive astrocytes highlighted a shift in protein expression profiles, pinpointing candidates that either facilitate or impede CNS repair, suggesting that depending on their reactivity state, astrocytes play a dual role during myelination.
Collapse
Affiliation(s)
- Sybille Seiler
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Franziska Rudolf
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Filipa Ramilo Gomes
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Anto Pavlovic
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Jana Nebel
- Department Neurochemistry & Molecular Biology, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Constanze I Seidenbecher
- Department Neurochemistry & Molecular Biology, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Lynette C Foo
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| |
Collapse
|
2
|
Xu Y, Benedikt J, Ye L. Hyaluronic Acid Interacting Molecules Mediated Crosstalk between Cancer Cells and Microenvironment from Primary Tumour to Distant Metastasis. Cancers (Basel) 2024; 16:1907. [PMID: 38791985 PMCID: PMC11119954 DOI: 10.3390/cancers16101907] [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: 04/26/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Hyaluronic acid (HA) is a prominent component of the extracellular matrix, and its interactions with HA-interacting molecules (HAIMs) play a critical role in cancer development and disease progression. This review explores the multifaceted role of HAIMs in the context of cancer, focusing on their influence on disease progression by dissecting relevant cellular and molecular mechanisms in tumour cells and the tumour microenvironment. Cancer progression can be profoundly affected by the interactions between HA and HAIMs. They modulate critical processes such as cell adhesion, migration, invasion, and proliferation. The TME serves as a dynamic platform in which HAIMs contribute to the formation of a unique niche. The resulting changes in HA composition profoundly influence the biophysical properties of the TME. These modifications in the TME, in conjunction with HAIMs, impact angiogenesis, immune cell recruitment, and immune evasion. Therefore, understanding the intricate interplay between HAIMs and HA within the cancer context is essential for developing novel therapeutic strategies. Targeting these interactions offers promising avenues for cancer treatment, as they hold the potential to disrupt critical aspects of disease progression and the TME. Further research in this field is imperative for advancing our knowledge and the treatment of cancer.
Collapse
Affiliation(s)
- Yali Xu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK;
| | | | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| |
Collapse
|
3
|
Schlotterose L, Cossais F, Lucius R, Hattermann K. Breaking the circulus vitiosus of neuroinflammation: Resveratrol attenuates the human glial cell response to cytokines. Biomed Pharmacother 2023; 163:114814. [PMID: 37148859 DOI: 10.1016/j.biopha.2023.114814] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023] Open
Abstract
Neuroinflammation is both cause and effect of many neurodegenerative disorders. Activation of astrocytes and microglia leads to the release of cytokines and reactive oxygen species followed by blood-brain barrier leakage and neurotoxicity. Transient neuroinflammation is considered to be largely protective, however, chronic neuroinflammation contributes to the pathology of Alzheimer's disease, multiple sclerosis, traumatic brain injury, and many more. In this study, we focus on the aspect of cytokine-induced neuroinflammation in human microglia and astrocytes. Here we show by mRNA and protein analysis that cytokines, released not only by microglia but also by astrocytes, lead to a circuit of proinflammatory activation. Moreover, we present how the natural compound resveratrol can stop the circuit of proinflammatory activation and facilitate return to resting conditions. These results will contribute to distinguishing between the causes and the effects of neuroinflammation, a better understanding of underlying mechanisms, and potential treatment options.
Collapse
Affiliation(s)
| | | | - Ralph Lucius
- Institute of Anatomy, Kiel University, 24118 Kiel, Germany
| | | |
Collapse
|
4
|
Jin C, Zong Y. The role of hyaluronan in renal cell carcinoma. Front Immunol 2023; 14:1127828. [PMID: 36936902 PMCID: PMC10019822 DOI: 10.3389/fimmu.2023.1127828] [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: 12/20/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is associated with high mortality rates worldwide and survival among RCC patients has not improved significantly in the past few years. A better understanding of the pathogenesis of RCC can enable the development of more effective therapeutic strategies against RCC. Hyaluronan (HA) is a glycosaminoglycan located in the extracellular matrix (ECM) that has several roles in biology, medicine, and physiological processes, such as tissue homeostasis and angiogenesis. Dysregulated HA and its receptors play important roles in fundamental cellular and molecular biology processes such as cell signaling, immune modulation, tumor progression and angiogenesis. There is emerging evidence that alterations in the production of HA regulate RCC development, thereby acting as important biomarkers as well as specific therapeutic targets. Therefore, targeting HA or combining it with other therapies are promising therapeutic strategies. In this Review, we summarize the available data on the role of abnormal regulation of HA and speculate on its potential as a therapeutic target against RCC.
Collapse
Affiliation(s)
- Chenchen Jin
- Zhejiang Academy of Science & Technology for Inspection & Quarantine, Hangzhou, Zhejiang, China
| | - Yunfeng Zong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Salmikangas M, Laaksonen M, Edgren H, Salgado M, Suoranta A, Mattila P, Koljonen V, Böhling T, Sihto H. Neurocan expression associates with better survival and viral positivity in Merkel cell carcinoma. PLoS One 2023; 18:e0285524. [PMID: 37146093 PMCID: PMC10162530 DOI: 10.1371/journal.pone.0285524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
Merkel cell carcinoma (MCC) is a rare cutaneous neuroendocrine carcinoma that is frequently divided into Merkel cell polyomavirus negative and positive tumors due their distinct genomic and transcriptomic profiles, and disease outcomes. Although some prognostic factors in MCC are known, tumorigenic pathways, which that explain outcome differences in MCC are not fully understood. We investigated transcriptomes of 110 tissue samples of a formalin-fixed, paraffin-embedded MCC series by RNA sequencing to identify genes showing a bimodal expression pattern and predicting outcome in cancer and that potentially could play a role in tumorigenesis. We discovered 19 genes among which IGHM, IGKC, NCAN, OTOF, and USH2A were associated also with overall survival (all p-values < 0.05). From these genes, NCAN (neurocan) expression was detected in all 144 MCC samples by immunohistochemistry. Increased NCAN expression was associated with presence of Merkel cell polyomavirus DNA (p = 0.001) and viral large T antigen expression in tumor tissue (p = 0.004) and with improved MCC-specific survival (p = 0.027) and overall survival (p = 0.034). We conclude that NCAN expression is common in MCC, and further studies are warranted to investigate its role in MCC tumorigenesis.
Collapse
Affiliation(s)
- Marko Salmikangas
- Department of Pathology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | - Marco Salgado
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anu Suoranta
- Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Pirkko Mattila
- Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Virve Koljonen
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tom Böhling
- Department of Pathology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Sihto
- Department of Pathology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
6
|
Fu Z, Zhu G, Luo C, Chen Z, Dou Z, Chen Y, Zhong C, Su S, Liu F. Matricellular protein tenascin C: Implications in glioma progression, gliomagenesis, and treatment. Front Oncol 2022; 12:971462. [PMID: 36033448 PMCID: PMC9413079 DOI: 10.3389/fonc.2022.971462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Matricellular proteins are nonstructural extracellular matrix components that are expressed at low levels in normal adult tissues and are upregulated during development or under pathological conditions. Tenascin C (TNC), a matricellular protein, is a hexameric and multimodular glycoprotein with different molecular forms that is produced by alternative splicing and post-translational modifications. Malignant gliomas are the most common and aggressive primary brain cancer of the central nervous system. Despite continued advances in multimodal therapy, the prognosis of gliomas remains poor. The main reasons for such poor outcomes are the heterogeneity and adaptability caused by the tumor microenvironment and glioma stem cells. It has been shown that TNC is present in the glioma microenvironment and glioma stem cell niches, and that it promotes malignant properties, such as neovascularization, proliferation, invasiveness, and immunomodulation. TNC is abundantly expressed in neural stem cell niches and plays a role in neurogenesis. Notably, there is increasing evidence showing that neural stem cells in the subventricular zone may be the cells of origin of gliomas. Here, we review the evidence regarding the role of TNC in glioma progression, propose a potential association between TNC and gliomagenesis, and summarize its clinical applications. Collectively, TNC is an appealing focus for advancing our understanding of gliomas.
Collapse
Affiliation(s)
- Zaixiang Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ganggui Zhu
- Department of Neurosurgery, Hangzhou First People’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chao Luo
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Zihang Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhangqi Dou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yike Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chen Zhong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Su
- Department of Neurosurgery, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Fuyi Liu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Fuyi Liu,
| |
Collapse
|
7
|
Tondepu C, Karumbaiah L. Glycomaterials to Investigate the Functional Role of Aberrant Glycosylation in Glioblastoma. Adv Healthc Mater 2022; 11:e2101956. [PMID: 34878733 PMCID: PMC9048137 DOI: 10.1002/adhm.202101956] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/30/2021] [Indexed: 02/03/2023]
Abstract
Glioblastoma (GBM) is a stage IV astrocytoma that carries a dismal survival rate of ≈10 months postdiagnosis and treatment. The highly invasive capacity of GBM and its ability to escape therapeutic challenges are key factors contributing to the poor overall survival rate. While current treatments aim to target the cancer cell itself, they fail to consider the significant role that the GBM tumor microenvironment (TME) plays in promoting tumor progression and therapeutic resistance. The GBM tumor glycocalyx and glycan-rich extracellular matrix (ECM), which are important constituents of the TME have received little attention as therapeutic targets. A wide array of aberrantly modified glycans in the GBM TME mediate tumor growth, invasion, therapeutic resistance, and immunosuppression. Here, an overview of the landscape of aberrant glycan modifications in GBM is provided, and the design and utility of 3D glycomaterials are discussed as a tool to evaluate glycan-mediated GBM progression and therapeutic efficacy. The development of alternative strategies to target glycans in the TME can potentially unveil broader mechanisms of restricting tumor growth and enhancing the efficacy of tumor-targeting therapeutics.
Collapse
Affiliation(s)
- Chaitanya Tondepu
- Regenerative Bioscience Science Center, University of Georgia, Athens, GA, 30602, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Science Center, University of Georgia, Athens, GA, 30602, USA
- Division of Neuroscience, Biomedical & Translational Sciences Institute, University of Georgia, Athens, GA, 30602, USA
- Edgar L. Rhodes Center for ADS, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
8
|
Structural and Functional Modulation of Perineuronal Nets: In Search of Important Players with Highlight on Tenascins. Cells 2021; 10:cells10061345. [PMID: 34072323 PMCID: PMC8230358 DOI: 10.3390/cells10061345] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the brain plays a crucial role in providing optimal conditions for neuronal function. Interactions between neurons and a specialized form of ECM, perineuronal nets (PNN), are considered a key mechanism for the regulation of brain plasticity. Such an assembly of interconnected structural and regulatory molecules has a prominent role in the control of synaptic plasticity. In this review, we discuss novel ways of studying the interplay between PNN and its regulatory components, particularly tenascins, in the processes of synaptic plasticity, mechanotransduction, and neurogenesis. Since enhanced neuronal activity promotes PNN degradation, it is possible to study PNN remodeling as a dynamical change in the expression and organization of its constituents that is reflected in its ultrastructure. The discovery of these subtle modifications is enabled by the development of super-resolution microscopy and advanced methods of image analysis.
Collapse
|
9
|
Barkovskaya A, Buffone A, Žídek M, Weaver VM. Proteoglycans as Mediators of Cancer Tissue Mechanics. Front Cell Dev Biol 2020; 8:569377. [PMID: 33330449 PMCID: PMC7734320 DOI: 10.3389/fcell.2020.569377] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
Proteoglycans are a diverse group of molecules which are characterized by a central protein backbone that is decorated with a variety of linear sulfated glycosaminoglycan side chains. Proteoglycans contribute significantly to the biochemical and mechanical properties of the interstitial extracellular matrix where they modulate cellular behavior by engaging transmembrane receptors. Proteoglycans also comprise a major component of the cellular glycocalyx to influence transmembrane receptor structure/function and mechanosignaling. Through their ability to initiate biochemical and mechanosignaling in cells, proteoglycans elicit profound effects on proliferation, adhesion and migration. Pathologies including cancer and cardiovascular disease are characterized by perturbed expression of proteoglycans where they compromise cell and tissue behavior by stiffening the extracellular matrix and increasing the bulkiness of the glycocalyx. Increasing evidence indicates that a bulky glycocalyx and proteoglycan-enriched extracellular matrix promote malignant transformation, increase cancer aggression and alter anti-tumor therapy response. In this review, we focus on the contribution of proteoglycans to mechanobiology in the context of normal and transformed tissues. We discuss the significance of proteoglycans for therapy response, and the current experimental strategies that target proteoglycans to sensitize cancer cells to treatment.
Collapse
Affiliation(s)
- Anna Barkovskaya
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander Buffone
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Martin Žídek
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Valerie M. Weaver
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
10
|
Islam S, Watanabe H. Versican: A Dynamic Regulator of the Extracellular Matrix. J Histochem Cytochem 2020; 68:763-775. [PMID: 33131383 DOI: 10.1369/0022155420953922] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Versican is a large chondroitin sulfate/dermatan sulfate proteoglycan belonging to the aggrecan/lectican family. In adults, this proteoglycan serves as a structural macromolecule of the extracellular matrix in the brain and large blood vessels. In contrast, versican is transiently expressed at high levels during development and under pathological conditions when the extracellular matrix dramatically changes, including in the inflammation and repair process. There are many reports showing the upregulation of versican in cancer, which correlates with cancer aggressiveness. Versican has four classical splice variants, and all the variants contain G1 and G3 domains at N- and C-termini, respectively. There are two glycosaminoglycan attachment domains CSα and CSβ. The largest V0 variant contains both CSα and CSβ, V1 contains CSβ, V2 contains CSα, and the shortest G3 variant has neither of them. Versican degradation is initiated by cleavage at a site in the CSβ domain by ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) proteinases. The N-terminal fragment containing the G1 domain has been reported to exert various biological functions, although its mechanisms of action have not yet been elucidated. In this review, we describe the role of versican in inflammation and cancer and also address the biological function of versikine.
Collapse
Affiliation(s)
- Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
11
|
Reinhard J, Wagner N, Krämer MM, Jarocki M, Joachim SC, Dick HB, Faissner A, Kakkassery V. Expression Changes and Impact of the Extracellular Matrix on Etoposide Resistant Human Retinoblastoma Cell Lines. Int J Mol Sci 2020; 21:ijms21124322. [PMID: 32560557 PMCID: PMC7352646 DOI: 10.3390/ijms21124322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
Retinoblastoma (RB) represents the most common malignant childhood eye tumor worldwide. Several studies indicate that the extracellular matrix (ECM) plays a crucial role in tumor growth and metastasis. Moreover, recent studies indicate that the ECM composition might influence the development of resistance to chemotherapy drugs. The objective of this study was to evaluate possible expression differences in the ECM compartment of the parental human cell lines WERI-RB1 (retinoblastoma 1) and Y79 and their Etoposide resistant subclones via polymerase chain reaction (PCR). Western blot analyses were performed to analyze protein levels. To explore the influence of ECM molecules on RB cell proliferation, death, and cluster formation, WERI-RB1 and resistant WERI-ETOR cells were cultivated on Fibronectin, Laminin, Tenascin-C, and Collagen IV and analyzed via time-lapse video microscopy as well as immunocytochemistry. We revealed a significantly reduced mRNA expression of the proteoglycans Brevican, Neurocan, and Versican in resistant WERI-ETOR compared to sensitive WERI-RB1 cells. Also, for the glycoproteins α1-Laminin, Fibronectin, Tenascin-C, and Tenascin-R as well as Collagen IV, reduced expression levels were observed in WERI-ETOR. Furthermore, a downregulation was detected for the matrix metalloproteinases MMP2, MMP7, MMP9, the tissue-inhibitor of metalloproteinase TIMP2, the Integrin receptor subunits ITGA4, ITGA5 and ITGB1, and all receptor protein tyrosine phosphatase β/ζ isoforms. Downregulation of Brevican, Collagen IV, Tenascin-R, MMP2, TIMP2, and ITGA5 was also verified in Etoposide resistant Y79 cells compared to sensitive ones. Protein levels of Tenascin-C and MMP-2 were comparable in both WERI cell lines. Interestingly, Fibronectin displayed an apoptosis-inducing effect on WERI-RB1 cells, whereas an anti-apoptotic influence was observed for Tenascin-C. Conversely, proliferation of WERI-ETOR cells was enhanced on Tenascin-C, while an anti-proliferative effect was observed on Fibronectin. In WERI-ETOR, cluster formation was decreased on the substrates Collagen IV, Fibronectin, and Tenascin-C. Collectively, we noted a different ECM mRNA expression and behavior of Etoposide resistant compared to sensitive RB cells. These findings may indicate a key role of ECM components in chemotherapy resistance formation of RB.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany; (N.W.); (M.M.K.); (M.J.); (A.F.)
- Correspondence: (J.R.); (V.K.); Tel.: +49-234-32-24-314 (J.R.); +49-451-500-43911 (V.K.); Fax: +49-234-32-143-13 (J.R.); +49-451-500-43914 (V.K.)
| | - Natalie Wagner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany; (N.W.); (M.M.K.); (M.J.); (A.F.)
| | - Miriam M. Krämer
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany; (N.W.); (M.M.K.); (M.J.); (A.F.)
| | - Marvin Jarocki
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany; (N.W.); (M.M.K.); (M.J.); (A.F.)
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany; (S.C.J.); (H.B.D.)
| | - H. Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany; (S.C.J.); (H.B.D.)
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany; (N.W.); (M.M.K.); (M.J.); (A.F.)
| | - Vinodh Kakkassery
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany; (S.C.J.); (H.B.D.)
- Department of Ophthalmology, University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
- Correspondence: (J.R.); (V.K.); Tel.: +49-234-32-24-314 (J.R.); +49-451-500-43911 (V.K.); Fax: +49-234-32-143-13 (J.R.); +49-451-500-43914 (V.K.)
| |
Collapse
|
12
|
Galardi A, Colletti M, Lavarello C, Di Paolo V, Mascio P, Russo I, Cozza R, Romanzo A, Valente P, De Vito R, Pascucci L, Peinado H, Carcaboso AM, Petretto A, Locatelli F, Di Giannatale A. Proteomic Profiling of Retinoblastoma-Derived Exosomes Reveals Potential Biomarkers of Vitreous Seeding. Cancers (Basel) 2020; 12:cancers12061555. [PMID: 32545553 PMCID: PMC7352325 DOI: 10.3390/cancers12061555] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma (RB) is the most common tumor of the eye in early childhood. Although recent advances in conservative treatment have greatly improved the visual outcome, local tumor control remains difficult in the presence of massive vitreous seeding. Traditional biopsy has long been considered unsafe in RB, due to the risk of extraocular spread. Thus, the identification of new biomarkers is crucial to design safer diagnostic and more effective therapeutic approaches. Exosomes, membrane-derived nanovesicles that are secreted abundantly by aggressive tumor cells and that can be isolated from several biological fluids, represent an interesting alternative for the detection of tumor-associated biomarkers. In this study, we defined the protein signature of exosomes released by RB tumors (RBT) and vitreous seeding (RBVS) primary cell lines by high resolution mass spectrometry. A total of 5666 proteins were identified. Among these, 5223 and 3637 were expressed in exosomes RBT and one RBVS group, respectively. Gene enrichment analysis of exclusively and differentially expressed proteins and network analysis identified in RBVS exosomes upregulated proteins specifically related to invasion and metastasis, such as proteins involved in extracellular matrix (ECM) remodeling and interaction, resistance to anoikis and the metabolism/catabolism of glucose and amino acids.
Collapse
Affiliation(s)
- Angela Galardi
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
| | - Marta Colletti
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
- Correspondence: ; Tel.: +39-066859-3516
| | - Chiara Lavarello
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS, Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy; (C.L.); (A.P.)
| | - Virginia Di Paolo
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
| | - Paolo Mascio
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
| | - Ida Russo
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
| | - Raffaele Cozza
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
| | - Antonino Romanzo
- Ophtalmology Unit, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’ Onofrio 4, 00165 Rome, Italy; (A.R.); (P.V.)
| | - Paola Valente
- Ophtalmology Unit, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’ Onofrio 4, 00165 Rome, Italy; (A.R.); (P.V.)
| | - Rita De Vito
- Department of Pathology, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza di Sant’ Onofrio 4, 00165 Rome, Italy;
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy;
| | - Hector Peinado
- Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, 28029 Madrid, Spain;
| | - Angel M. Carcaboso
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, 08950 Esplugues de Llobregat, Spain;
| | - Andrea Petretto
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS, Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy; (C.L.); (A.P.)
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
- Department of Ginecology/Obstetrics & Pediatrics, Sapienza University of Rome, 00185 Roma, Italy
| | - Angela Di Giannatale
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.); (V.D.P.); (P.M.); (I.R.); (R.C.); (F.L.); (A.D.G.)
| |
Collapse
|
13
|
Giamanco KA, Matthews RT. The Role of BEHAB/Brevican in the Tumor Microenvironment: Mediating Glioma Cell Invasion and Motility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:117-132. [PMID: 32845505 DOI: 10.1007/978-3-030-48457-6_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malignant gliomas are the most common tumors in the central nervous system (CNS) and, unfortunately, are also the most deadly. The lethal nature of malignant gliomas is due in large part to their unique and distinctive ability to invade the surrounding neural tissue. The invasive and dispersive nature of these tumors makes them particularly challenging to treat, and currently there are no effective therapies for malignant gliomas. The brain tumor microenvironment plays a particularly important role in mediating the invasiveness of gliomas, and, therefore, understanding its function is key to developing novel therapies to treat these deadly tumors. A defining aspect of the tumor microenvironment of gliomas is the unique composition of the extracellular matrix that enables tumors to overcome the typically inhibitory environment found in the CNS. One conspicuous component of the glioma tumor microenvironment is the neural-specific ECM molecule, brain-enriched hyaluronan binding (BEHAB)/brevican (B/b). B/b is highly overexpressed in gliomas, and its expression in these tumors contributes importantly to the tumor invasiveness and aggressiveness. However, B/b is a complicated protein with multiple splice variants, cleavage products, and glycoforms that contribute to its complex functions in these tumors and provide unique targets for tumor therapy. Here we review the role of B/b in glioma tumor microenvironment and explore targeting of this protein for glioma therapy.
Collapse
Affiliation(s)
- Kristin A Giamanco
- Department of Biological and Environmental Sciences, Western Connecticut State University, Danbury, CT, USA
| | - Russell T Matthews
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
14
|
Belousov A, Titov S, Shved N, Garbuz M, Malykin G, Gulaia V, Kagansky A, Kumeiko V. The Extracellular Matrix and Biocompatible Materials in Glioblastoma Treatment. Front Bioeng Biotechnol 2019; 7:341. [PMID: 31803736 PMCID: PMC6877546 DOI: 10.3389/fbioe.2019.00341] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
During cancer genesis, the extracellular matrix (ECM) in the human brain undergoes important transformations, starting to resemble embryonic brain cell milieu with a much denser structure. However, the stiffness of the tumor ECM does not preclude cancer cells from migration. The importance of the ECM role in normal brain tissue as well as in tumor homeostasis has engaged much effort in trials to implement ECM as a target and an instrument in the treatment of brain cancers. This review provides a detailed analysis of both experimental and applied approaches in combined therapy for gliomas in adults. In general, matrix materials for glioma treatment should have properties facilitating the simplest delivery into the body. Hence, to deliver an artificial implant directly into the operation cavity it should be packed into a gel form, while for bloodstream injections matrix needs to be in the form of polymer micelles, nanoparticles, etc. Furthermore, the delivered material should mimic biomechanical properties of the native tissue, support vital functions, and slow down or stop the proliferation of surrounding cells for a prolonged period. The authors propose a two-step approach aimed, on the one hand, at elimination of remaining cancer cells and on the other hand, at restoring normal brain tissue. Thereby, the first bioartificial matrix to be applied should have relatively low elastic modulus should be loaded with anticancer drugs, while the second material with a higher elastic modulus for neurite outgrowth support should contain specific factors stimulating neuroregeneration.
Collapse
Affiliation(s)
- Andrei Belousov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Sergei Titov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
| | - Nikita Shved
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Mikhail Garbuz
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Grigorii Malykin
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Valeriia Gulaia
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Alexander Kagansky
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
15
|
Morad G, Moses MA. Brainwashed by extracellular vesicles: the role of extracellular vesicles in primary and metastatic brain tumour microenvironment. J Extracell Vesicles 2019; 8:1627164. [PMID: 31275532 PMCID: PMC6598504 DOI: 10.1080/20013078.2019.1627164] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/08/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Brain malignancies, including primary and metastatic brain tumours, are often associated with high mortality, reflecting a need for more effective diagnostics and therapeutics. Despite the different cells of origin, primary and metastatic brain tumours share the same microenvironment, which affects the survival mechanisms adopted by these tumours. Elucidating the mechanisms by which primary and metastatic brain tumours interact with the brain microenvironment can uncover potential targets for clinical applications. Extracellular vesicles have been recognized as intercellular communicators that can contribute to cancer progression and have shown promise as potential cancer biomarkers and therapeutics. Here, we outline the contribution of extracellular vesicles in the tumour–microenvironment interactions in primary and metastatic brain tumours with the goal of providing a guide for future translational research in this area.
Collapse
Affiliation(s)
- Golnaz Morad
- The Vascular Biology Program and The Department of Surgery, Boston Children's Hospital, Boston, MA, USA.,Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Marsha A Moses
- The Vascular Biology Program and The Department of Surgery, Boston Children's Hospital, Boston, MA, USA.,Department of Surgery, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Saxena T, Lyon JG, Pai SB, Pare D, Amero J, Karumbaia L, Carroll SL, Gaupp E, Bellamkonda RV. Engineering Controlled Peritumoral Inflammation to Constrain Brain Tumor Growth. Adv Healthc Mater 2019; 8:e1801076. [PMID: 30537355 PMCID: PMC6657526 DOI: 10.1002/adhm.201801076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/23/2018] [Indexed: 02/06/2023]
Abstract
Brain tumors remain a great clinical challenge, in part due to their capacity to invade into eloquent, inoperable regions of the brain. In contrast, inflammation in the central nervous system (CNS) due to injuries activates microglia and astrocytes culminating in an astroglial scar that typically "walls-off" the injury site. Here, the hypothesis is tested that targeting peritumoral cells surrounding tumors to activate them via an inflammatory stimulus that recapitulates the sequelae of a traumatic CNS injury, could generate an environment that would wall-off and contain invasive tumors in the brain. Gold nanoparticles coated with inflammatory polypeptides to target stromal cells in close vicinity to glioblastoma (GBM) tumors, in order to activate these cells and stimulate stromal CNS inflammation, are engineered. It is reported that this approach significantly contains tumors in rodent models of GBM relative to control treatments (reduction in tumor volume by over 300% in comparison to controls), by the activation of the innate and adaptive immune response, and by triggering pathways related to cell clustering. Overall, this report outlines an approach to contain invasive tumors that can complement adjuvant interventions for invasive GBM such as radiation and chemotherapy.
Collapse
Affiliation(s)
- Tarun Saxena
- Department of Biomedical Engineering Pratt School of Engineering
Duke University, 101 Science Drive, Durham, NC 27705, USA
| | - Johnathan G. Lyon
- Department of Biomedical Engineering Pratt School of Engineering
Duke University, 101 Science Drive, Durham, NC 27705, USA
| | - S. Balakrishna Pai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
Institute of Technology & Emory School of Medicine, UA Whitaker
Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Daniel Pare
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
Institute of Technology & Emory School of Medicine, UA Whitaker
Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Jessica Amero
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
Institute of Technology & Emory School of Medicine, UA Whitaker
Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Lohitash Karumbaia
- Regenerative Bioscience Center, The University of Georgia, 425
River Road, ADS Complex, Athens, GA 30602, USA
| | - Sheridan L. Carroll
- Department of Biomedical Engineering Pratt School of Engineering
Duke University, 101 Science Drive, Durham, NC 27705, USA
| | - Eric Gaupp
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
Institute of Technology & Emory School of Medicine, UA Whitaker
Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Ravi V. Bellamkonda
- Department of Biomedical Engineering Pratt School of Engineering
Duke University, 101 Science Drive, Durham, NC 27705, USA,
| |
Collapse
|
17
|
Virga J, Bognár L, Hortobágyi T, Csősz É, Kalló G, Zahuczki G, Steiner L, Hutóczki G, Reményi-Puskár J, Klekner A. The Expressional Pattern of Invasion-Related Extracellular Matrix Molecules in CNS Tumors. Cancer Invest 2018; 36:492-503. [PMID: 30501525 DOI: 10.1080/07357907.2018.1545855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Aim of the study: Astrocytomas are primary CNS malignancies which infiltrate the peritumoral tissue, even when they are low-grade. Schwannomas are also primary CNS tumors, however, they do not show peritumoral infiltration similarly to brain metastases which almost never invade the neighboring parts of brain. Extracellular matrix is altered in composition in various cancer types and is proposed to play an important role in the development of invasiveness of astrocytic tumors. This study aims to identify differences in the ECM composition of CNS tumors with different invasiveness.Materials and methods: The mRNA and protein levels of ECM components were measured by QRT-PCR and mass-spectrometry, respectively, in grade II astrocytoma, NSCLC brain metastasis, schwannomas, and non-tumor brain control samples. Expressional data was analyzed statistically with ANOVA and nearest neighbor search.Results: There is a significant difference in the expressional pattern of invasion-related ECM components among various CNS tumors, especially among those of different embryonic origin. Non-invasive tumors show only slight differences in the expressional pattern of ECM molecules. Tumor samples can be separated based on their expressional pattern using statistical classifiers, therefore the ECM composition seems to be typical of various cancer types.Conclusions: Differences in the expressional pattern of the ECM could be responsible for the different invasiveness of various CNS tumors.
Collapse
Affiliation(s)
- József Virga
- Department of Neurosurgery, University of Debrecen, Debrecen, Hungary
| | - László Bognár
- Department of Neurosurgery, University of Debrecen, Debrecen, Hungary
| | - Tibor Hortobágyi
- Department of Neuropathology, University of Debrecen, Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Gergő Kalló
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Gábor Zahuczki
- UD-GenoMed Medical Genomic Technologies Research & Development Services Ltd., Debrecen, Hungary
| | - László Steiner
- UD-GenoMed Medical Genomic Technologies Research & Development Services Ltd., Debrecen, Hungary
| | - Gábor Hutóczki
- Department of Neurosurgery, University of Debrecen, Debrecen, Hungary
| | | | - Almos Klekner
- Department of Neurosurgery, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
18
|
Klein JA, Meng L, Zaia J. Deep Sequencing of Complex Proteoglycans: A Novel Strategy for High Coverage and Site-specific Identification of Glycosaminoglycan-linked Peptides. Mol Cell Proteomics 2018; 17:1578-1590. [PMID: 29773674 DOI: 10.1074/mcp.ra118.000766] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/09/2018] [Indexed: 12/17/2022] Open
Abstract
Proteoglycans are distributed in all animal tissues and play critical, multifaceted, physiological roles. Expressed in a spatially and temporally regulated manner, these molecules regulate interactions among growth factors and cell surface receptors and play key roles in basement membranes and other extracellular matrices. Because of the high degree of glycosylation by glycosaminoglycan (GAG), N-glycan and mucin-type O-glycan classes, the peptide sequence coverage of complex proteoglycans is revealed poorly by standard mass spectrometry-based proteomics methods. As a result, there is little information concerning how proteoglycan site specific glycosylation changes during normal and pathological processes. Here, we developed a workflow to improve sequence coverage and identification of glycosylated peptides in proteoglycans. We applied this workflow to the small leucine-rich proteoglycan decorin and three hyalectan proteoglycans: neurocan, brevican, and aggrecan.We characterized glycosylation of these proteoglycans using LC-MS methods easily implemented on instruments widely used in proteomics laboratories. For decorin, we assigned the linker-glycosite and three N-glycosylation sites. For neurocan and brevican, we identified densely glycosylated mucin-like regions in the extended domains. For aggrecan, we identified 50 linker-glycosites and mucin-type O-glycosites in the extended region and N-glycosites in the globular domains, many of which are novel and have not been observed previously. Most importantly, we demonstrate an LC-MS and bioinformatics approach that will enable routine analysis of proteoglycan glycosylation from biological samples to assess their role in pathophysiology.
Collapse
Affiliation(s)
- Joshua A Klein
- From the ‡Department of Biochemistry, Center for Biomedical Mass Spectrometry.,§Bioinformatics Program Boston University, Boston, Massachusetts 02118
| | - Le Meng
- From the ‡Department of Biochemistry, Center for Biomedical Mass Spectrometry
| | - Joseph Zaia
- From the ‡Department of Biochemistry, Center for Biomedical Mass Spectrometry; .,§Bioinformatics Program Boston University, Boston, Massachusetts 02118
| |
Collapse
|
19
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
20
|
Herrera J, Henke CA, Bitterman PB. Extracellular matrix as a driver of progressive fibrosis. J Clin Invest 2018; 128:45-53. [PMID: 29293088 DOI: 10.1172/jci93557] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The extracellular matrix (ECM) is dynamically tuned to optimize physiological function. Its major properties, including composition and mechanics, profoundly influence cell biology. Cell-ECM interactions operate through an integrated set of sensor and effector circuits that use several classes of receptors and signal transduction pathways. At the single-cell level, the ECM governs differentiation, metabolism, motility, orientation, proliferation, and survival. At the cell population level, the ECM provides higher-order guidance that is essential for physiological function. When pathological changes in the ECM lead to impairment of organ function, we use the term "fibrosis." In this Review, we differentiate fibrosis initiation from progression and focus primarily on progressive lung fibrosis impairing organ function. We present a working model to explain how the altered ECM is not only a consequence but also a driver of fibrosis. Additionally, we advance the concept that fibrosis progression occurs in a fibrogenic niche that is composed of a fibrogenic ECM that nurtures fibrogenic mesenchymal progenitor cells and their fibrogenic progeny.
Collapse
|
21
|
Su Z, Kishida S, Tsubota S, Sakamoto K, Cao D, Kiyonari S, Ohira M, Kamijo T, Narita A, Xu Y, Takahashi Y, Kadomatsu K. Neurocan, an extracellular chondroitin sulfate proteoglycan, stimulates neuroblastoma cells to promote malignant phenotypes. Oncotarget 2017; 8:106296-106310. [PMID: 29290949 PMCID: PMC5739734 DOI: 10.18632/oncotarget.22435] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022] Open
Abstract
Neurocan (NCAN), a secreted chondroitin sulfate proteoglycan, is one of the major inhibitory molecules for axon regeneration in nervous injury. However, its role in cancer is not clear. Here we observed that high NCAN expression was closely associated with the unfavorable outcome of neuroblastoma (NB). NCAN was also highly and ubiquitously expressed in the early lesions and terminal tumor of TH-MYCN mice, a NB model. Interestingly, exogenous NCAN (i.e., overexpression, recombinant protein and conditioned medium) transformed adherent NB cells into spheres whose malignancies in vitro (anchorage-independent growth and chemoresistance) and in vivo (xenograft tumor growth) were potentiated. Both chondroitin sulfate sugar chains and NCAN's core protein were essential for the sphere formation. The CSG3 domain was essential in the moiety of NCAN. Our comprehensive microarray analysis and RT-qPCR of mRNA expression suggested that NCAN treatment promoted cell division, and urged cells to undifferentiated state. The knockdown of NCAN in tumor sphere cells cultured from TH-MYCN mice resulted in growth suppression in vitro and in vivo. Our findings suggest that NCAN, which stimulates NB cells to promote malignant phenotypes, is an extracellular molecule providing a growth advantage to cancer cells.
Collapse
Affiliation(s)
- Zhendong Su
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Satoshi Kishida
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Dongliang Cao
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shinichi Kiyonari
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Miki Ohira
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Saitama, Japan
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Saitama, Japan
| | - Atsushi Narita
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yinyan Xu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
22
|
Theocharis AD, Karamanos NK. Proteoglycans remodeling in cancer: Underlying molecular mechanisms. Matrix Biol 2017; 75-76:220-259. [PMID: 29128506 DOI: 10.1016/j.matbio.2017.10.008] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Extracellular matrix is a highly dynamic macromolecular network. Proteoglycans are major components of extracellular matrix playing key roles in its structural organization and cell signaling contributing to the control of numerous normal and pathological processes. As multifunctional molecules, proteoglycans participate in various cell functions during morphogenesis, wound healing, inflammation and tumorigenesis. Their interactions with matrix effectors, cell surface receptors and enzymes enable them with unique properties. In malignancy, extensive remodeling of tumor stroma is associated with marked alterations in proteoglycans' expression and structural variability. Proteoglycans exert diverse functions in tumor stroma in a cell-specific and context-specific manner and they mainly contribute to the formation of a permissive provisional matrix for tumor growth affecting tissue organization, cell-cell and cell-matrix interactions and tumor cell signaling. Proteoglycans also modulate cancer cell phenotype and properties, the development of drug resistance and tumor stroma angiogenesis. This review summarizes the proteoglycans remodeling and their novel biological roles in malignancies with particular emphasis to the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
23
|
Labrousse-Arias D, Martínez-Ruiz A, Calzada MJ. Hypoxia and Redox Signaling on Extracellular Matrix Remodeling: From Mechanisms to Pathological Implications. Antioxid Redox Signal 2017; 27:802-822. [PMID: 28715969 DOI: 10.1089/ars.2017.7275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is an essential modulator of cell behavior that influences tissue organization. It has a strong relevance in homeostasis and translational implications for human disease. In addition to ECM structural proteins, matricellular proteins are important regulators of the ECM that are involved in a myriad of different pathologies. Recent Advances: Biochemical studies, animal models, and study of human diseases have contributed to the knowledge of molecular mechanisms involved in remodeling of the ECM, both in homeostasis and disease. Some of them might help in the development of new therapeutic strategies. This review aims to review what is known about some of the most studied matricellular proteins and their regulation by hypoxia and redox signaling, as well as the pathological implications of such regulation. CRITICAL ISSUES Matricellular proteins have complex regulatory functions and are modulated by hypoxia and redox signaling through diverse mechanisms, in some cases with controversial effects that can be cell or tissue specific and context dependent. Therefore, a better understanding of these regulatory processes would be of great benefit and will open new avenues of considerable therapeutic potential. FUTURE DIRECTIONS Characterizing the specific molecular mechanisms that modulate matricellular proteins in pathological processes that involve hypoxia and redox signaling warrants additional consideration to harness the potential therapeutic value of these regulatory proteins. Antioxid. Redox Signal. 27, 802-822.
Collapse
Affiliation(s)
- David Labrousse-Arias
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain
| | - Antonio Martínez-Ruiz
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,2 Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) , Madrid, Spain
| | - María J Calzada
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,3 Departmento de Medicina, Universidad Autónoma de Madrid , Madrid, Spain
| |
Collapse
|
24
|
Asgharzadeh MR, Barar J, Pourseif MM, Eskandani M, Jafari Niya M, Mashayekhi MR, Omidi Y. Molecular machineries of pH dysregulation in tumor microenvironment: potential targets for cancer therapy. BIOIMPACTS : BI 2017; 7:115-133. [PMID: 28752076 PMCID: PMC5524986 DOI: 10.15171/bi.2017.15] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/28/2017] [Accepted: 06/06/2017] [Indexed: 12/30/2022]
Abstract
Introduction: Cancer is an intricate disorder/dysfunction of cells that can be defined as a genetic heterogeneity in human disease. Therefore, it is characterized by several adaptive complex hallmarks. Among them, the pH dysregulation appears as a symbol of aberrant functions within the tumor microenvironment (TME). In comparison with normal tissues, in the solid tumors, we face with an irregular acidification and alkalinization of the extracellular and intracellular fluids. Methods: In this study, we comprehensively discussed the most recent reports on the hallmarks of solid tumors to provide deep insights upon the molecular machineries involved in the pH dysregulation of solid tumors and their impacts on the initiation and progression of cancer. Results: The dysregulation of pH in solid tumors is fundamentally related to the Warburg effect and hypoxia, leading to expression of a number of molecular machineries, including: NHE1, H+ pump V-ATPase, CA-9, CA-12, MCT-1, GLUT-1. Activation of proton exchangers and transporters (PETs) gives rise to formation of TME. This condition favors the cancer cells to evade from the anoikis and apoptosis, granting them aggressive and metastasis phenotype, as well as resistance to chemotherapy and radiation therapy. This review aimed to discuss the key molecular changes of tumor cells in terms of bio-energetics and cancer metabolism in relation with pH dysregulation. During this phenomenon, the intra- and extracellular metabolites are altered and/or disrupted. Such molecular alterations provide molecular hallmarks for direct targeting of the PETs by potent relevant inhibitors in combination with conventional cancer therapies as ultimate therapy against solid tumors. Conclusion: Taken all, along with other treatment strategies, targeting the key molecular machineries related to intra- and extracellular metabolisms within the TME is proposed as a novel strategy to inhibit or block PETs that are involved in the pH dysregulation of solid tumors.
Collapse
Affiliation(s)
- Mohammad Reza Asgharzadeh
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Jafari Niya
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | | | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Qi XT, Zhan JS, Xiao LM, Li L, Xu HX, Fu ZB, Zhang YH, Zhang J, Jia XH, Ge G, Chai RC, Gao K, Yu ACH. The Unwanted Cell Migration in the Brain: Glioma Metastasis. Neurochem Res 2017; 42:1847-1863. [PMID: 28478595 DOI: 10.1007/s11064-017-2272-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 12/19/2022]
Abstract
Cell migration is identified as a highly orchestrated process. It is a fundamental and essential phenomenon underlying tissue morphogenesis, wound healing, and immune response. Under dysregulation, it contributes to cancer metastasis. Brain is considered to be the most complex organ in human body containing many types of neural cells with astrocytes playing crucial roles in monitoring both physiological and pathological functions. Astrocytoma originates from astrocytes and its most malignant type is glioblastoma multiforme (WHO Grade IV astrocytoma), which is capable to infiltrate widely into the neighboring brain tissues making a complete resection of tumors impossible. Very recently, we have reviewed the mechanisms for astrocytes in migration. Given the fact that astrocytoma shares many histological features with astrocytes, we therefore attempt to review the mechanisms for glioma cells in migration and compare them to normal astrocytes, hoping to obtain a better insight into the dysregulation of migratory mechanisms contributing to their metastasis in the brain.
Collapse
Affiliation(s)
- Xue Tao Qi
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Jiang Shan Zhan
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Li Ming Xiao
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Lina Li
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China.
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China.
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China.
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China.
| | - Han Xiao Xu
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Human Anatomy, Guizhou Medical University, Guian New Area, Guiyang, Guizhou, 550025, China
| | - Zi Bing Fu
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Yan Hao Zhang
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Jing Zhang
- Department of Pathology, Peking University Health Science Center and Peking University Third Hospital, Beijing, 100191, China
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Xi Hua Jia
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Guo Ge
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Human Anatomy, Guizhou Medical University, Guian New Area, Guiyang, Guizhou, 550025, China
| | - Rui Chao Chai
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Kai Gao
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Albert Cheung Hoi Yu
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China.
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China.
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China.
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China.
- Laboratory of Translational Medicine, Institute of Systems Biomedicine, Peking University, Beijing, 100191, China.
| |
Collapse
|
26
|
Oliveira-Ferrer L, Legler K, Milde-Langosch K. Role of protein glycosylation in cancer metastasis. Semin Cancer Biol 2017; 44:141-152. [PMID: 28315783 DOI: 10.1016/j.semcancer.2017.03.002] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 02/06/2023]
Abstract
Although altered glycosylation has been detected in human cancer cells decades ago, only investigations in the last years have enormously increased our knowledge about the details of protein glycosylation and its role in tumour progression. Many proteins, which are heavily glycosylated, i.e. adhesion proteins or proteases, play an important role in cancer metastasis that represents the crucial and frequently life-threatening step in progression of most tumour types. Compared to normal tissue, tumour cells often show altered glycosylation patters with appearance of new tumour-specific antigens. In this review, we give an overview about the role of glycosylation in tumour metastasis, describing recent results about O-glycans, N-glycans and glycosaminoglycans. We show that glycan structures, glycosylated proteins and glycosylation enzymes have influence on different steps of the metastatic process, including epithelial-mesenchymal transition (EMT), migration, invasion/intravasation and extravasation of tumour cells. Regarding the important role of cancer metastasis for patients survival, further knowledge about the consequences of altered glycosylation patterns in tumour cells is needed which might eventually lead to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Karen Legler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
27
|
Tumor Grade versus Expression of Invasion-Related Molecules in Astrocytoma. Pathol Oncol Res 2017; 24:35-43. [DOI: 10.1007/s12253-017-0194-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
|
28
|
Tenascin-C and fibronectin expression divide early stage tongue cancer into low- and high-risk groups. Br J Cancer 2017; 116:640-648. [PMID: 28095396 PMCID: PMC5344290 DOI: 10.1038/bjc.2016.455] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/18/2016] [Accepted: 12/20/2016] [Indexed: 12/23/2022] Open
Abstract
Background: Oral tongue squamous cell carcinoma (OTSCC) metastasises early, especially to regional lymph nodes. There is an ongoing debate on which early stage (T1-T2N0) patients should be treated with elective neck dissection. We need prognosticators for early stage tongue cancer. Methods: Mice immunisation with human mesenchymal stromal cells resulted in production of antibodies against tenascin-C (TNC) and fibronectin (FN), which were used to stain 178 (98 early stage), oral tongue squamous cell carcinoma samples. Tenascin-C and FN expression in the stroma (negative, moderate or abundant) and tumour cells (negative or positive) were assessed. Similar staining was obtained using corresponding commercial antibodies. Results: Expression of TNC and FN in the stroma, but not in the tumour cells, proved to be excellent prognosticators both in all stages and in early stage cases. Among early stages, when stromal TNC was negative, the 5-year survival rate was 88%. Correspondingly, when FN was negative, no cancer deaths were observed. Five-year survival rates for abundant expression of TNC and FN were 43% and 25%, respectively. Conclusions: Stromal TNC and, especially, FN expressions differentiate patients into low- and high-risk groups. Surgery alone of early stage primary tumours might be adequate when stromal FN is negative. Aggressive treatments should be considered when both TNC and FN are abundant.
Collapse
|
29
|
Binder MJ, McCoombe S, Williams ED, McCulloch DR, Ward AC. The extracellular matrix in cancer progression: Role of hyalectan proteoglycans and ADAMTS enzymes. Cancer Lett 2016; 385:55-64. [PMID: 27838414 DOI: 10.1016/j.canlet.2016.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 01/08/2023]
Abstract
Remodelling of the extracellular matrix (ECM) has emerged as a key factor in cancer progression. Proteoglycans, including versican and other hyalectans, represent major structural elements of the ECM where they interact with other important molecules, including the glycosaminoglycan hyaluronan and the CD44 cell surface receptor. The hyalectan proteoglycans are regulated through cleavage by the proteolytic actions of A Disintegrin-like And Metalloproteinase domain with Thrombospondin-1 motif (ADAMTS) family members. Alteration in the balance between hyalectan proteoglycans and ADAMTS enzymes has been proposed to be a crucial factor in cancer progression either in a positive or negative manner depending on the context. Further complexity arises due to the formation of bioactive cleavage products, such as versikine, which may also play a role, and non-enzymatic functions for ADAMTS proteins. This research is providing fresh insights into cancer biology and opportunities for the development of new diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Marley J Binder
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Scott McCoombe
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland 4000, Australia
| | - Daniel R McCulloch
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| |
Collapse
|
30
|
Polisetty RV, Gautam P, Gupta MK, Sharma R, Gowda H, Renu D, Shivakumar BM, Lakshmikantha A, Mariswamappa K, Ankathi P, Purohit AK, Uppin MS, Sundaram C, Sirdeshmukh R. Microsomal membrane proteome of low grade diffuse astrocytomas: Differentially expressed proteins and candidate surveillance biomarkers. Sci Rep 2016; 6:26882. [PMID: 27246909 PMCID: PMC4887981 DOI: 10.1038/srep26882] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/03/2016] [Indexed: 01/27/2023] Open
Abstract
Diffuse astrocytoma (DA; WHO grade II) is a low-grade, primary brain neoplasm with high potential of recurrence as higher grade malignant form. We have analyzed differentially expressed membrane proteins from these tumors, using high-resolution mass spectrometry. A total of 2803 proteins were identified, 340 of them differentially expressed with minimum of 2 fold change and based on ≥2 unique peptides. Bioinformatics analysis of this dataset also revealed important molecular networks and pathways relevant to tumorigenesis, mTOR signaling pathway being a major pathway identified. Comparison of 340 differentially expressed proteins with the transcript data from Grade II diffuse astrocytomas reported earlier, revealed about 190 of the proteins correlate in their trends in expression. Considering progressive and recurrent nature of these tumors, we have mapped the differentially expressed proteins for their secretory potential, integrated the resulting list with similar list of proteins from anaplastic astrocytoma (WHO Grade III) tumors and provide a panel of proteins along with their proteotypic peptides, as a resource that would be useful for investigation as circulatory plasma markers for post-treatment surveillance of DA patients.
Collapse
Affiliation(s)
| | - Poonam Gautam
- Centre for Cellular and Molecular Biology (CSIR), Hyderabad, India
| | - Manoj Kumar Gupta
- Centre for Cellular and Molecular Biology (CSIR), Hyderabad, India.,Institute of Bioinformatics, Bangalore, India.,Manipal University, Madhav Nagar, Manipal, India
| | | | | | | | | | | | | | - Praveen Ankathi
- Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | | | - Megha S Uppin
- Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | - Challa Sundaram
- Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | - Ravi Sirdeshmukh
- Centre for Cellular and Molecular Biology (CSIR), Hyderabad, India.,Institute of Bioinformatics, Bangalore, India.,Neuro-Oncology, Mazumdar Shaw Center for Translational Research, Narayana Health, Bangalore, India
| |
Collapse
|
31
|
Reinhard J, Brösicke N, Theocharidis U, Faissner A. The extracellular matrix niche microenvironment of neural and cancer stem cells in the brain. Int J Biochem Cell Biol 2016; 81:174-183. [PMID: 27157088 DOI: 10.1016/j.biocel.2016.05.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/25/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
Numerous studies demonstrated that neural stem cells and cancer stem cells (NSCs/CSCs) share several overlapping characteristics such as self-renewal, multipotency and a comparable molecular repertoire. In addition to the intrinsic cellular properties, NSCs/CSCs favor a similar environment to acquire and maintain their characteristics. In the present review, we highlight the shared properties of NSCs and CSCs in regard to their extracellular microenvironment called the NSC/CSC niche. Moreover, we point out that extracellular matrix (ECM) molecules and their complementary receptors influence the behavior of NSCs/CSCs as well as brain tumor progression. Here, we focus on the expression profile and functional importance of the ECM glycoprotein tenascin-C, the chondroitin sulfate proteoglycan DSD-1-PG/phosphacan but also on other important glycoprotein/proteoglycan constituents. Within this review, we specifically concentrate on glioblastoma multiforme (GBM). GBM is the most common malignant brain tumor in adults and is associated with poor prognosis despite intense and aggressive surgical and therapeutic treatment. Recent studies indicate that GBM onset is driven by a subpopulation of CSCs that display self-renewal and recapitulate tumor heterogeneity. Based on the CSC hypothesis the cancer arises just from a small subpopulation of self-sustaining cancer cells with the exclusive ability to self-renew and maintain the tumor. Besides the fundamental stem cell properties of self-renewal and multipotency, GBM stem cells share further molecular characteristics with NSCs, which we would like to review in this article.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Nicole Brösicke
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Ursula Theocharidis
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany.
| |
Collapse
|
32
|
Hutóczki G, Bognár L, Tóth J, Scholtz B, Zahuczky G, Hanzély Z, Csősz É, Reményi-Puskár J, Kalló G, Hortobágyi T, Klekner A. Effect of Concomitant Radiochemotherapy on Invasion Potential of Glioblastoma. Pathol Oncol Res 2015; 22:155-60. [PMID: 26450124 DOI: 10.1007/s12253-015-9989-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/29/2015] [Indexed: 01/22/2023]
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults with inevitable recurrence after oncotherapy. The insufficient effect of "gold standard" temozolomide-based concomitant radiochemotherapy may be due to the inability to prevent tumor cell invasion. Peritumoral infiltration depends mainly on the interaction between extracellular matrix (ECM) components and cell membrane receptors. Changes in invasive behaviour after oncotherapy can be evaluated at the molecular level by determining the RNA expression and protein levels of the invasion-related ECM components. The expression of nineteen ECM molecules was determined at both RNA and protein levels in thirty-one GBM samples. Fifteen GBM samples originated from the first surgical procedure on patients before oncotherapy, and sixteen GBM samples were collected at the second surgery due to local recurrence after concomitant chemoirradiation. RNA expressions were measured with qRT-PCR, and protein levels were determined by quantitative analysis of Western blots. Only MMP-9 RNA transcript level was reduced (p < 0.05) whereas at protein level, eight molecules showed changes concordant with RNA expression with significant decrease in brevican only. The results suggest that concomitant radiochemotherapy does not have sufficient impact on the expression of invasion-related ECM components of glioblastoma, oncotherapy does not significantly affect its invasive behavior. To avoid the spread of tumors into the brain parenchyma, supplementation of antiproliferative treatment with anti-invasive agents may be worth consideration in oncotherapy for glioblastoma.
Collapse
Affiliation(s)
- Gábor Hutóczki
- Department of Neurosurgery, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - László Bognár
- Department of Neurosurgery, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary.
| | - Judit Tóth
- Department of Oncology, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Beáta Scholtz
- Department of Biochemistry and Molecular Biology, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Gábor Zahuczky
- Department of Biochemistry and Molecular Biology, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary.,UD-Genomed Medical Genomic Technologies Ltd., Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Zoltán Hanzély
- National Institute of Clinical Neurosciences, Amerikai út 57, Budapest, 1145, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Judit Reményi-Puskár
- Department of Neurosurgery, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Gergő Kalló
- Department of Biochemistry and Molecular Biology, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Tibor Hortobágyi
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| | - Almos Klekner
- Department of Neurosurgery, University of Debrecen, Clinical Center, Nagyerdei krt. 98, Debrecen, 4032, Hungary
| |
Collapse
|
33
|
Expression pattern of invasion-related molecules in the peritumoral brain. Clin Neurol Neurosurg 2015; 139:138-43. [PMID: 26451999 DOI: 10.1016/j.clineuro.2015.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/22/2015] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The effectiveness of therapy of intracerebral neoplasms is mainly influenced by the invasive behaviour of the tumour. The peritumoral invasion depends on the interaction between the tumour cells and the extracellular matrix (ECM) of the surrounding brain. The invading tumour cells induce change in the activity of proteases, synthases and expression of ECM-components. These alterations in the peritumoral ECM are in connection to the highly different invasiveness of gliomas and metastatic brain tumours. To understand the fairly modified invasive potential of anaplastic intracerebral tumours of different origin, the effect of tumour on the peritumoral ECM and alterations of invasion related ECM components in the peritumoral brain were evaluated. METHODS For this reason the mRNA expression of 19 invasion-related molecules by quantitative reverse transcriptase polymerase chain reaction was determined in normal brain tissue (Norm), in the peritumoral brain tissue of glioblastoma (peri-GBM) and of intracerebral adenocarcinoma metastasis (peri-Met). To evaluate the translational expression of the investigated molecules protein levels were determined by targeted proteomic methods. RESULTS Establishing the invasion pattern of the investigated tissue samples 8 molecules showed concordant difference at mRNA and protein levels in the peri-GBM and peri-Met, 11 molecules in the peri-Met and normal brain and 12 in the peri-GBM and normal brain comparison. CONCLUSION Our results bring some ECM molecules into focus that probably play key role in arresting tumour cell invasion around the metastatic tumour, and also in the lack of impeding tumour cell migration in case of glioblastoma.
Collapse
|
34
|
Abstract
Proteoglycans (PGs) regulate diverse functions in the central nervous system (CNS) by interacting with a number of growth factors, matrix proteins, and cell surface molecules. Heparan sulfate (HS) and chondroitin sulfate (CS) are two major glycosaminoglycans present in the PGs of the CNS. The functionality of these PGs is to a large extent dictated by the fine sulfation patterns present on their glycosaminoglycan (GAG) chains. In the past 15 years, there has been a significant expansion in our knowledge on the role of HS and CS chains in various neurological processes, such as neuronal growth, regeneration, plasticity, and pathfinding. However, defining the relation between distinct sulfation patterns of the GAGs and their functionality has thus far been difficult. With the emergence of novel tools for the synthesis of defined GAG structures, and techniques for their characterization, we are now in a better position to explore the structure-function relation of GAGs in the context of their sulfation patterns. In this review, we discuss the importance of GAGs on CNS development, injury, and disorders with an emphasis on their sulfation patterns. Finally, we outline several GAG-based therapeutic strategies to exploit GAG chains for ameliorating various CNS disorders.
Collapse
Affiliation(s)
- Vimal P Swarup
- Department of Bioengineering, University of Utah, Salt Lake City, 84112 UT , USA
| | | | | | | |
Collapse
|
35
|
Martino-Echarri E, Fernández-Rodríguez R, Bech-Serra JJ, Plaza-Calonge MDC, Vidal N, Casal C, Colomé N, Seoane J, Canals F, Rodríguez-Manzaneque JC. Relevance of IGFBP2 proteolysis in glioma and contribution of the extracellular protease ADAMTS1. Oncotarget 2015; 5:4295-304. [PMID: 24962328 PMCID: PMC4147324 DOI: 10.18632/oncotarget.2009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Expression of IGFBP2 (Insulin-like Growth Factor Binding Protein 2) has been positively correlated with glioma progression. Although the proteolysis of IGFBP2 has been widely recognized, with consequences as a major modulator of IGFII signaling, the relevance of this post-translational modification has not been well studied in tumors. Using an in vivo proteomic approach by Isotope-Coded Protein Label (ICPL), we identified IGFBP2 as a target of the extracellular protease ADAMTS1 (A Disintegrin And Metalloproteinase with ThromboSpondin motifs 1). Notably, the proteolytic pattern of IGFBP2 was also detected in human glioma culture cells and, more importantly, in all glioma samples evaluated. In addition, high expression of ADAMTS1 correlates with higher levels of cleaved IGFBP2 in glioblastoma multiforme cases. Using gene expression public databases, we confirmed that IGFBP2 is a poor prognosis marker for gliomas, and we also observed an important contribution of ADAMTS1.Finally, we showed the impact of ADAMTS1 on IGFII-mediated IGF1R phosphorylation and cellular migration. Our results support a functional interaction between IGFBP2 and ADAMTS1 and suggest the need to evaluate post-translational modifications of IGFBP2 in glioma, in order to approach new therapies.
Collapse
Affiliation(s)
- Estefanía Martino-Echarri
- GENYO. Centre for Genomics and Oncological Research: Pfizer/Universidad de Granada/Junta de Andalucía, Granada, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sainio A, Järveläinen H. Extracellular matrix macromolecules: potential tools and targets in cancer gene therapy. MOLECULAR AND CELLULAR THERAPIES 2014; 2:14. [PMID: 26056582 PMCID: PMC4452050 DOI: 10.1186/2052-8426-2-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/23/2014] [Indexed: 02/07/2023]
Abstract
Tumour cells create their own microenvironment where they closely interact with a variety of soluble and non-soluble molecules, different cells and numerous other components within the extracellular matrix (ECM). Interaction between tumour cells and the ECM is bidirectional leading to either progression or inhibition of tumourigenesis. Therefore, development of novel therapies targeted primarily to tumour microenvironment (TME) is highly rational. Here, we give a short overview of different macromolecules of the ECM and introduce mechanisms whereby they contribute to tumourigenesis within the TME. Furthermore, we present examples of individual ECM macromolecules as regulators of cell behaviour during tumourigenesis. Finally, we focus on novel strategies of using ECM macromolecules as tools or targets in cancer gene therapy in the future.
Collapse
Affiliation(s)
- Annele Sainio
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Hannu Järveläinen
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland ; Division of Endocrinology, Department of Medicine, Turku University Hospital, Kiinamyllynkatu 4-8, Fl-20520 Turku, Finland
| |
Collapse
|
37
|
Silver DJ, Silver J. Contributions of chondroitin sulfate proteoglycans to neurodevelopment, injury, and cancer. Curr Opin Neurobiol 2014; 27:171-8. [PMID: 24762654 DOI: 10.1016/j.conb.2014.03.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/25/2014] [Accepted: 03/25/2014] [Indexed: 01/09/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are a diverse family of extracellular matrix (ECM) molecules that make significant contributions to the patterning and routing of migrating neural cells and extending axons. Three distinct modes of migration mediation result from the relative abundance and positioning of expressed CSPGs, the profile of CSPG receptors expressed by the motile cell types, and the overall way in which the CSPGs integrate into and stabilize the neural ECM. Here we discuss recent findings that help to clarify the molecular mechanisms that underlie these distinct migration-regulating properties as they pertain to neural development, CNS injury, and gliomagenesis.
Collapse
Affiliation(s)
- Daniel J Silver
- Human Biology Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Mailstop C3-168, Seattle, WA 98109, United States
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, 2109 Adelbert Road Rm E-658, Cleveland, OH 44106, United States.
| |
Collapse
|
38
|
Chondroitin sulfate proteoglycans potently inhibit invasion and serve as a central organizer of the brain tumor microenvironment. J Neurosci 2013; 33:15603-17. [PMID: 24068827 DOI: 10.1523/jneurosci.3004-12.2013] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) remains the most pervasive and lethal of all brain malignancies. One factor that contributes to this poor prognosis is the highly invasive character of the tumor. GBM is characterized by microscopic infiltration of tumor cells throughout the brain, whereas non-neural metastases, as well as select lower grade gliomas, develop as self-contained and clearly delineated lesions. Illustrated by rodent xenograft tumor models as well as pathological human patient specimens, we present evidence that one fundamental switch between these two distinct pathologies--invasion and noninvasion--is mediated through the tumor extracellular matrix. Specifically, noninvasive lesions are associated with a rich matrix containing substantial amounts of glycosylated chondroitin sulfate proteoglycans (CSPGs), whereas glycosylated CSPGs are essentially absent from diffusely infiltrating tumors. CSPGs, acting as central organizers of the tumor microenvironment, dramatically influence resident reactive astrocytes, inducing their exodus from the tumor mass and the resultant encapsulation of noninvasive lesions. Additionally, CSPGs induce activation of tumor-associated microglia. We demonstrate that the astrogliotic capsule can directly inhibit tumor invasion, and its absence from GBM presents an environment favorable to diffuse infiltration. We also identify the leukocyte common antigen-related phosphatase receptor (PTPRF) as a putative intermediary between extracellular glycosylated CSPGs and noninvasive tumor cells. In all, we present CSPGs as critical regulators of brain tumor histopathology and help to clarify the role of the tumor microenvironment in brain tumor invasion.
Collapse
|
39
|
Thelin MA, Bartolini B, Axelsson J, Gustafsson R, Tykesson E, Pera E, Oldberg Å, Maccarana M, Malmstrom A. Biological functions of iduronic acid in chondroitin/dermatan sulfate. FEBS J 2013; 280:2431-46. [PMID: 23441919 PMCID: PMC3717172 DOI: 10.1111/febs.12214] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 02/17/2013] [Accepted: 02/18/2013] [Indexed: 01/08/2023]
Abstract
The presence of iduronic acid in chondroitin/dermatan sulfate changes the properties of the polysaccharides because it generates a more flexible chain with increased binding potentials. Iduronic acid in chondroitin/dermatan sulfate influences multiple cellular properties, such as migration, proliferation, differentiation, angiogenesis and the regulation of cytokine/growth factor activities. Under pathological conditions such as wound healing, inflammation and cancer, iduronic acid has diverse regulatory functions. Iduronic acid is formed by two epimerases (i.e. dermatan sulfate epimerase 1 and 2) that have different tissue distribution and properties. The role of iduronic acid in chondroitin/dermatan sulfate is highlighted by the vast changes in connective tissue features in patients with a new type of Ehler–Danlos syndrome: adducted thumb-clubfoot syndrome. Future research aims to understand the roles of the two epimerases and their interplay with the sulfotransferases involved in chondroitin sulfate/dermatan sulfate biosynthesis. Furthermore, a better definition of chondroitin/dermatan sulfate functions using different knockout models is needed. In this review, we focus on the two enzymes responsible for iduronic acid formation, as well as the role of iduronic acid in health and disease.
Collapse
Affiliation(s)
- Martin A Thelin
- Department of Experimental Medical Science, BMC, Lund University, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kultti A, Li X, Jiang P, Thompson CB, Frost GI, Shepard HM. Therapeutic targeting of hyaluronan in the tumor stroma. Cancers (Basel) 2012; 4:873-903. [PMID: 24213471 PMCID: PMC3712709 DOI: 10.3390/cancers4030873] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/28/2012] [Accepted: 08/31/2012] [Indexed: 12/12/2022] Open
Abstract
The tumor stroma, consisting of non-malignant cells and the extracellular matrix, undergoes significant quantitative and qualitative changes throughout malignant transformation and tumor progression. With increasing recognition of the role of the tumor microenvironment in disease progression, stromal components of the tumor have become attractive targets for therapeutic intervention. Stromal accumulation of the glycosaminoglycan hyaluronan occurs in many tumor types and is frequently associated with a negative disease prognosis. Hyaluronan interacts with other extracellular molecules as well as cellular receptors to form a complex interaction network influencing physicochemical properties, signal transduction, and biological behavior of cancer cells. In preclinical animal models, enzymatic removal of hyaluronan is associated with remodeling of the tumor stroma, reduction of tumor interstitial fluid pressure, expansion of tumor blood vessels and facilitated delivery of chemotherapy. This leads to inhibition of tumor growth and increased survival. Current evidence shows that abnormal accumulation of hyaluronan may be an important stromal target for cancer therapy. In this review we highlight the role of hyaluronan and hyaluronan-mediated interactions in cancer, and discuss historical and recent data on hyaluronidase-based therapies and the effect of hyaluronan removal on tumor growth.
Collapse
Affiliation(s)
- Anne Kultti
- Department of Research, Halozyme Therapeutics, 11388 Sorrento Valley Road, San Diego, CA 92121, USA; E-Mails: (H.M.S.)
| | - Xiaoming Li
- Department of Pharmacology and Safety Assessment, Halozyme Therapeutics, 11388 Sorrento Valley Road, San Diego, CA 92121, USA; E-Mails: (X.L.); (P.J.); (C.B.T.)
| | - Ping Jiang
- Department of Pharmacology and Safety Assessment, Halozyme Therapeutics, 11388 Sorrento Valley Road, San Diego, CA 92121, USA; E-Mails: (X.L.); (P.J.); (C.B.T.)
| | - Curtis B. Thompson
- Department of Pharmacology and Safety Assessment, Halozyme Therapeutics, 11388 Sorrento Valley Road, San Diego, CA 92121, USA; E-Mails: (X.L.); (P.J.); (C.B.T.)
| | - Gregory I. Frost
- Department of General and Administrative, Halozyme Therapeutics, 11388 Sorrento Valley Road, San Diego, CA 92121, USA; E-Mail: (G.I.F.)
| | - H. Michael Shepard
- Department of Research, Halozyme Therapeutics, 11388 Sorrento Valley Road, San Diego, CA 92121, USA; E-Mails: (H.M.S.)
| |
Collapse
|