1
|
Chen X, Ran X, Wei X, Zhu L, Chen S, Liao Z, Xu K, Xia W. Bioactive glass 1393 promotes angiogenesis and accelerates wound healing through ROS/P53/MMP9 signaling pathway. Regen Ther 2024; 26:132-144. [PMID: 38872979 PMCID: PMC11169082 DOI: 10.1016/j.reth.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
Compared to bioactive glass 45S5, bioactive glass 1393 has shown greater potential in activating tissue cells and promoting angiogenesis for bone repair. Nevertheless, the effect of bioactive glass 1393 in the context of wound healing remains extensively unexplored, and its mechanism in wound healing remains unclear. Considering that angiogenesis is a critical stage in wound healing, we hypothesize that bioactive glass 1393 may facilitate wound healing through the stimulation of angiogenesis. To validate this hypothesis and further explore the mechanisms underlying its pro-angiogenic effects, we investigated the impact of bioactive glass 1393 on wound healing angiogenesis through both in vivo and in vitro studies. The research demonstrated that bioactive glass 1393 accelerated wound healing by promoting the formation of granulation, deposition of collagen, and angiogenesis. The results of Western blot analysis and immunofluorescence staining revealed that bioactive glass 1393 up-regulated the expression of angiogenesis-related factors. Additionally, bioactive glass 1393 inhibited the expression of ROS and P53 to promote angiogenesis. Furthermore, bioactive glass 1393 stimulated angiogenesis through the P53 signaling pathway, as evidenced by P53 activation assays. Collectively, these findings indicate that bioactive glass 1393 accelerates wound healing by promoting angiogenesis via the ROS/P53/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Xuenan Chen
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
- College of Life and Environmental Sciences, Wenzhou University, Zhejiang, China
| | - Xinyu Ran
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuebo Wei
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lifei Zhu
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Shaodong Chen
- Department of Orthopaedics, Lishui People's Hospital, Zhejiang, China
| | - Zhiyong Liao
- College of Life and Environmental Sciences, Wenzhou University, Zhejiang, China
| | - Ke Xu
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
- College of Life and Environmental Sciences, Wenzhou University, Zhejiang, China
| | - Weidong Xia
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
2
|
Arnesen VS, Gras Navarro A, Chekenya M. Challenges and Prospects for Designer T and NK Cells in Glioblastoma Immunotherapy. Cancers (Basel) 2021; 13:4986. [PMID: 34638471 PMCID: PMC8507952 DOI: 10.3390/cancers13194986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is the most prevalent, aggressive primary brain tumour with a dismal prognosis. Treatment at diagnosis has limited efficacy and there is no standardised treatment at recurrence. New, personalised treatment options are under investigation, although challenges persist for heterogenous tumours such as GBM. Gene editing technologies are a game changer, enabling design of novel molecular-immunological treatments to be used in combination with chemoradiation, to achieve long lasting survival benefits for patients. Here, we review the literature on how cutting-edge molecular gene editing technologies can be applied to known and emerging tumour-associated antigens to enhance chimeric antigen receptor T and NK cell therapies for GBM. A tight balance of limiting neurotoxicity, avoiding tumour antigen loss and therapy resistance, while simultaneously promoting long-term persistence of the adoptively transferred cells must be maintained to significantly improve patient survival. We discuss the opportunities and challenges posed by the brain contexture to the administration of the treatments and achieving sustained clinical responses.
Collapse
Affiliation(s)
| | - Andrea Gras Navarro
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| |
Collapse
|
3
|
Kmiecik J, Gras Navarro A, Poli A, Planagumà JP, Zimmer J, Chekenya M. Combining NK cells and mAb9.2.27 to combat NG2-dependent and anti-inflammatory signals in glioblastoma. Oncoimmunology 2021; 3:e27185. [PMID: 24575382 PMCID: PMC3916357 DOI: 10.4161/onci.27185] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 11/13/2013] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma is a deadly brain cancer with limited treatment options. Targeting chondroitin sulfate proteoglycan 4 (CSPG4, best known as NG2) with the monoclonal antibody mAb9.2.27 and activated natural killer (NK) cells abrogated the tumor growth and prolonged the survival of glioblastoma-bearing animals by favoring the establishment of a pro-inflammatory microenvironment. The combination of NK cells and mAb9.2.27 recruited ED1+CCR2low macrophages that stimulated ED1+ED2lowMHCIIhigh microglial cells to exert robust cytotoxicity. Our findings demonstrate the therapeutic potential of targeting salient tumor associated-antigens.
Collapse
Affiliation(s)
- Justyna Kmiecik
- University of Bergen; Institute for Biomedicine; Bergen, Norway
| | | | - Aurelie Poli
- Laboratoire d'Immunogénétique-Allergologie; CRP-Santé, Luxembourg
| | | | - Jacques Zimmer
- Laboratoire d'Immunogénétique-Allergologie; CRP-Santé, Luxembourg
| | - Martha Chekenya
- University of Bergen; Institute for Biomedicine; Bergen, Norway ; University of Bergen, Institute for Clinical Dentistry; Bergen, Norway
| |
Collapse
|
4
|
Simon B, Uslu U. Fasten the seat belt: Increasing safety of CAR T-cell therapy. Exp Dermatol 2020; 29:1039-1045. [PMID: 32627228 DOI: 10.1111/exd.14131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
After the recent success and approvals of chimeric antigen receptor (CAR) T cells in haematological malignancies, its efficacy is currently evaluated in a broad spectrum of tumor entities including melanoma. However, severe and potentially life-threatening side effects like cytokine release syndrome, neurologic toxicities, and the competing risk of morbidity and mortality from the treatment itself are still a major limiting factor in the current CAR T-cell landscape. In addition, especially in solid tumors, the lack of ideal target antigens to avoid on-target/off-tumor toxicities also restricts its use. While various groups are working on strategies to boost CAR T-cell efficacy, mechanisms to increase engineered T-cell safety should not move out of focus. Thus, the aim of this article is to summarize and to discuss current and potential future strategies and mechanisms to increase CAR T-cell safety in order to enable the wide use of this promising approach in melanoma and other tumor entities.
Collapse
Affiliation(s)
- Bianca Simon
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Comprehensive Cancer Center Erlangen- European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.,Division of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ugur Uslu
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Comprehensive Cancer Center Erlangen- European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
5
|
Mellai M, Annovazzi L, Bisogno I, Corona C, Crociara P, Iulini B, Cassoni P, Casalone C, Boldorini R, Schiffer D. Chondroitin Sulphate Proteoglycan 4 (NG2/CSPG4) Localization in Low- and High-Grade Gliomas. Cells 2020; 9:E1538. [PMID: 32599896 PMCID: PMC7349878 DOI: 10.3390/cells9061538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Neuron glial antigen 2 or chondroitin sulphate proteoglycan 4 (NG2/CSPG4) is expressed by immature precursors/progenitor cells and is possibly involved in malignant cell transformation. The aim of this study was to investigate its role on the progression and survival of sixty-one adult gliomas and nine glioblastoma (GB)-derived cell lines. METHODS NG2/CSPG4 protein expression was assessed by immunohistochemistry and immunofluorescence. Genetic and epigenetic alterations were detected by molecular genetic techniques. RESULTS NG2/CSPG4 was frequently expressed in IDH-mutant/1p19q-codel oligodendrogliomas (59.1%) and IDH-wild type GBs (40%) and rarely expressed in IDH-mutant or IDH-wild type astrocytomas (14.3%). Besides tumor cells, NG2/CSPG4 immunoreactivity was found in the cytoplasm and/or cell membranes of reactive astrocytes and vascular pericytes/endothelial cells. In GB-derived neurospheres, it was variably detected according to the number of passages of the in vitro culture. In GB-derived adherent cells, a diffuse positivity was found in most cells. NG2/CSPG4 expression was significantly associated with EGFR gene amplification (p = 0.0005) and poor prognosis (p = 0.016) in astrocytic tumors. CONCLUSION The immunoreactivity of NG2/CSPG4 provides information on the timing of the neoplastic transformation and could have prognostic and therapeutic relevance as a promising tumor-associated antigen for antibody-based immunotherapy in patients with malignant gliomas.
Collapse
Affiliation(s)
- Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy; (M.M.); (R.B.)
- Centro Interdipartimentale di Ricerca Traslazionale sulle Malattie Autoimmuni e Allergiche (CAAD), Università del Piemonte Orientale (UPO), Corso Trieste 15A, 28100 Novara, Italy
- Fondazione Edo ed Elvo Tempia Valenta—ONLUS, Via Malta 3, 13900 Biella, Italy
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| | - Ilaria Bisogno
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Paola Crociara
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Barbara Iulini
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Paola Cassoni
- Dipartimento di Scienze Mediche, Università di Torino/Città della Salute e della Scienza, Via Santena 7, 10126 Torino, Italy;
| | - Cristina Casalone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Renzo Boldorini
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy; (M.M.); (R.B.)
| | - Davide Schiffer
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| |
Collapse
|
6
|
Liu Q, Yang Y, Fan X. Microvascular pericytes in brain-associated vascular disease. Biomed Pharmacother 2020; 121:109633. [DOI: 10.1016/j.biopha.2019.109633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 01/01/2023] Open
|
7
|
Harrer DC, Dörrie J, Schaft N. CSPG4 as Target for CAR-T-Cell Therapy of Various Tumor Entities-Merits and Challenges. Int J Mol Sci 2019; 20:ijms20235942. [PMID: 31779130 PMCID: PMC6928974 DOI: 10.3390/ijms20235942] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/21/2019] [Accepted: 11/23/2019] [Indexed: 12/18/2022] Open
Abstract
Targeting cancer cells using chimeric-antigen-receptor (CAR-)T cells has propelled adoptive T-cell therapy (ATT) to the next level. A plentitude of durable complete responses using CD19-specific CAR-T cells in patients suffering from various lymphoid malignancies resulted in the approval by the food and drug administration (FDA) of CD19-directed CAR-T cells for the treatment of acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). A substantial portion of this success in hematological malignancies can be traced back to the beneficial properties of the target antigen CD19, which combines a universal presence on target cells with no detectable expression on indispensable host cells. Hence, to replicate response rates achieved in ALL and DLBCL in the realm of solid tumors, where ideal target antigens are scant and CAR-T cells are still lagging behind expectations, the quest for appropriate target antigens represents a crucial task to expedite the next steps in the evolution of CAR-T-cell therapy. In this review, we want to highlight the potential of chondroitin sulfate proteoglycan 4 (CSPG4) as a CAR-target antigen for a variety of different cancer entities. In particular, we discuss merits and challenges associated with CSPG4-CAR-T cells for the ATT of melanoma, leukemia, glioblastoma, and triple-negative breast cancer.
Collapse
|
8
|
Tamburini E, Dallatomasina A, Quartararo J, Cortelazzi B, Mangieri D, Lazzaretti M, Perris R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality. FASEB J 2018; 33:3112-3128. [PMID: 30550356 DOI: 10.1096/fj.201801670r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The chondroitin sulfate proteoglycan 4 ( CSPG4) gene encodes a transmembrane proteoglycan (PG) constituting the largest and most structurally complex macromolecule of the human surfaceome. Its transcript shows an extensive evolutionary conservation and, due to the elaborated intracellular processing of the translated protein, it generates an array of glycoforms with the potential to exert variant-specific functions. CSPG4-mediated molecular events are articulated through the interaction with more than 40 putative ligands and the concurrent involvement of the ectodomain and cytoplasmic tail. Alternating inside-out and outside-in signal transductions may thereby be elicited through a tight functional connection of the PG with the cytoskeleton and its regulators. The potential of CSPG4 to influence both types of signaling mechanisms is also asserted by its lateral mobility along the plasma membrane and its intersection with microdomain-restricted internalization and endocytic trafficking. Owing to the multitude of molecular interplays that CSPG4 may engage, and thanks to a differential phosphorylation of its intracellular domain accounted by crosstalking signaling pathways, the PG stands out for its unique capability to affect numerous cellular phenomena, including those purporting pathologic conditions. We discuss here the progresses made in advancing our understanding about the structural-functional bases for the ability of CSPG4 to widely impact on cell behavior, such as to highlight how its multivalency may be exploited to interfere with disease progression.-Tamburini, E., Dallatomasina, A., Quartararo, J., Cortelazzi, B., Mangieri, D., Lazzaretti, M., Perris, R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality.
Collapse
Affiliation(s)
- Elisa Tamburini
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Alice Dallatomasina
- Division of Experimental Oncology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; and
| | - Jade Quartararo
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Barbara Cortelazzi
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | | | - Mirca Lazzaretti
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Roberto Perris
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| |
Collapse
|
9
|
Cenciarelli C, Marei HE, Felsani A, Casalbore P, Sica G, Puglisi MA, Cameron AJM, Olivi A, Mangiola A. PDGFRα depletion attenuates glioblastoma stem cells features by modulation of STAT3, RB1 and multiple oncogenic signals. Oncotarget 2018; 7:53047-53063. [PMID: 27344175 PMCID: PMC5288168 DOI: 10.18632/oncotarget.10132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022] Open
Abstract
Platelet derived growth factor receptors (PDGFRs) play an important role in tumor pathogenesis, and they are frequently overexpressed in glioblastoma (GBM). Earlier we have shown a higher protein expression of PDGFR isoforms (α and β) in peritumoral-tissue derived cancer stem cells (p-CSC) than in tumor core (c-CSC) of several GBM affected patients. In the current study, in order to assess the activity of PDGFRα/PDGF-AA signaling axis, we performed time course experiments to monitor the effects of exogenous PDGF-AA on the expression of downstream target genes in c-CSC vs p-CSC. Interestingly, in p-CSC we detected the upregulation of Y705-phosphorylated Stat3, concurrent with a decrement of Rb1 protein in its active state, within minutes of PDGF-AA addition. This finding prompted us to elucidate the role of PDGFRα in self-renewal, invasion and differentiation in p-CSC by using short hairpin RNA depletion of PDGFRα expression. Notably, in PDGFRα-depleted cells, protein analysis revealed attenuation of stemness-related and glial markers expression, alongside early activation of the neuronal marker MAP2a/b that correlated with the induction of tumor suppressor Rb1. The in vitro reduction of the invasive capacity of PDGFRα-depleted CSC as compared to parental cells correlated with the downmodulation of markers of epithelial-mesenchymal transition phenotype and angiogenesis. Surprisingly, we observed the induction of anti-apoptotic proteins and compensatory oncogenic signals such as EDN1, EDNRB, PRKCB1, PDGF-C and PDGF-D. To conclude, we hypothesize that the newly discovered PDGFRα/Stat3/Rb1 regulatory axis might represent a potential therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Carlo Cenciarelli
- Institute of Translational Pharmacology, Department of Biomedical Sciences-National Research Council (IFT-CNR), Rome, Italy
| | - Hany E Marei
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Armando Felsani
- Institute of Cell Biology and Neurobiology, Dept. of Biomedical Sciences-National Research Council (IBCN-CNR), Rome, Italy
| | - Patrizia Casalbore
- Institute of Cell Biology and Neurobiology, Dept. of Biomedical Sciences-National Research Council (IBCN-CNR), Rome, Italy
| | - Gigliola Sica
- Institute of Histology and Embryology, Catholic University-School of Medicine, Rome, Italy
| | | | - Angus J M Cameron
- Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Alessandro Olivi
- Institute of Neurosurgery, Department of Head and Neck, Catholic University-School of Medicine, Rome, Italy
| | - Annunziato Mangiola
- Institute of Neurosurgery, Department of Head and Neck, Catholic University-School of Medicine, Rome, Italy
| |
Collapse
|
10
|
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: 20.9] [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
|
11
|
Komori T. Pathology of oligodendroglia: An overview. Neuropathology 2017; 37:465-474. [PMID: 28548216 DOI: 10.1111/neup.12389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 11/29/2022]
Abstract
Oligodendroglia are cells responsible for creating myelin sheaths for axons in the CNS. However, pathologies of oligodendroglia other than demyelination are not well understood due to the lack of adequate methods of characterizing pathological conditions affecting oligodendroglia in human tissue. This review discusses three major topics with the aim of clarifying some of the controversies in the study of oligodendroglia. The oligodendroglioma, a relatively indolent form of diffuse gliomas thought to originate in oligodendrocytes, has never demonstrated myelin formation on electron microscopy nor shown a constant expression of myelin-related proteins. Oligodendrogliomas instead share an immune phenotype with oligodendrocyte progenitor cells (OPCs). Another type of cell that resembles OPCs are oligodendroglia-like cells (OLCs), which occur in many types of low-grade tumors and focal cortical dysplasia. In neurodegenerative disorders, oligodendroglia can be a target of abnormal aggregations of proteins such as tau. Tau-positive oligodendroglial inclusions in progressive supranuclear palsy and corticobasal generation differ from each other morphologically, ultrastructurally and biochemically, suggesting disparate underlying pathological processes despite significant overlapping of the clinical manifestations. To promote the study of oligodendroglia, novel methods for detecting OLCs in situ are urgently required.
Collapse
Affiliation(s)
- Takashi Komori
- Department of Laboratory Medicine and Pathology (Neuropathology), Tokyo Metropolitan Neurological Hospital, Fuchu, Japan
| |
Collapse
|
12
|
Lu L, Chai L, Wang W, Yuan X, Li S, Cao C. A Selenium-Enriched Ziyang Green Tea Polysaccharide Induces Bax-Dependent Mitochondrial Apoptosis and Inhibits TGF-β1-Stimulated Collagen Expression in Human Keloid Fibroblasts via NG2 Inactivation. Biol Trace Elem Res 2017; 176:270-277. [PMID: 27565798 DOI: 10.1007/s12011-016-0827-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/16/2016] [Indexed: 12/13/2022]
Abstract
Keloids are fibroproliferative disorders characterized by the overabundant deposition of extracellular matrix (ECM), especially collagen and overgrowth of scar tissue in response to cutaneous injury. In this study, we isolated a selenium (Se)-containing polysaccharide (Se-ZGTP-I) from Ziyang green tea and explored its potential therapeutic effects on keloid fibroblasts formation. 3-(4,5-Dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and annexin V/propidium iodide (PI) staining assays demonstrated that Se-ZGTP-I or neuron-glia 2 (NG2) short hairpin RNA (shRNA) significantly inhibited proliferation of human keloid fibroblasts via induction of apoptosis. Besides, the activation of caspase-3 and the subsequent cleavage of poly (ADP-ribose) polymerase (PARP) were observed in keloid fibroblasts following Se-ZGTP-I (200 and 400 μg/ml) or NG2 shRNA treatment. Moreover, Western blotting analysis showed that treatment of keloid fibroblasts with Se-ZGTP-I (200 and 400 μg/ml) or NG2 shRNA resulted in an increase of pro-apoptotic protein Bax expression and a decrease in expression levels of anti-apoptotic protein Bcl-2 and NG2. In addition, type I collagen biosynthesis and protein expression in keloid fibroblasts following TGF-β1 stimulation were decreased by Se-ZGTP-I (200 and 400 μg/ml) or NG2 shRNA management. Current findings imply that Se-ZGTP-I has a therapeutic potential to intervene and prevent keloid formation and other fibrotic diseases.
Collapse
Affiliation(s)
- Lele Lu
- The Third Military Medical University Southwest Plastic Surgery Hospital, Chongqing, 400037, People's Republic of China
| | - Linlin Chai
- The Third Military Medical University Southwest Plastic Surgery Hospital, Chongqing, 400037, People's Republic of China
| | - Wenping Wang
- The Third Military Medical University Southwest Plastic Surgery Hospital, Chongqing, 400037, People's Republic of China
| | - Xi Yuan
- The Third Military Medical University Southwest Plastic Surgery Hospital, Chongqing, 400037, People's Republic of China
| | - Shirong Li
- The Third Military Medical University Southwest Plastic Surgery Hospital, Chongqing, 400037, People's Republic of China.
| | - Chuan Cao
- The Third Military Medical University Southwest Plastic Surgery Hospital, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
13
|
de Souza Lins Borba FK, Felix GLQ, Costa EVL, Silva L, Dias PF, de Albuquerque Nogueira R. Fractal analysis of extra-embryonic vessels of chick embryos under the effect of glucosamine and chondroitin sulfates. Microvasc Res 2016; 105:114-8. [DOI: 10.1016/j.mvr.2016.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/07/2016] [Accepted: 02/08/2016] [Indexed: 11/30/2022]
|
14
|
Yadavilli S, Hwang EI, Packer RJ, Nazarian J. The Role of NG2 Proteoglycan in Glioma. Transl Oncol 2016; 9:57-63. [PMID: 26947882 PMCID: PMC4800061 DOI: 10.1016/j.tranon.2015.12.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/09/2015] [Accepted: 12/22/2015] [Indexed: 01/08/2023] Open
Abstract
Neuron glia antigen-2 ((NG2), also known as chondroitin sulphate proteoglycan 4, or melanoma-associated chondroitin sulfate proteoglycan) is a type-1 membrane protein expressed by many central nervous system (CNS) cells during development and differentiation and plays a critical role in proliferation and angiogenesis. ‘NG2’ often references either the protein itself or the highly proliferative and undifferentiated glial cells expressing high levels of NG2 protein. NG2 glia represent the fourth major type of neuroglia in the mammalian nervous system and are classified as oligodendrocyte progenitor cells by virtue of their committed oligodendrocyte generation in developing and adult brain. Here, we discuss NG2 glial cells as well as NG2 protein and its expression and role with regards to CNS neoplasms as well as its potential as a therapeutic target for treating childhood CNS cancers.
Collapse
Affiliation(s)
- Sridevi Yadavilli
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Ave. NW, Washington, DC 20010, USA
| | - Eugene I Hwang
- Division of Oncology, Children's National Health System, 111 Michigan Ave. NW, Washington, DC 20010, USA
| | - Roger J Packer
- Brain Tumor Institute, Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC 20010, USA
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Ave. NW, Washington, DC 20010, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA.
| |
Collapse
|
15
|
Bansal P, Medhe S, Ganesh N, Srivastava MM. Antimelanoma Potential of Eruca sativa Seed Oil and its Bioactive Principles. Indian J Pharm Sci 2015; 77:208-17. [PMID: 26009655 PMCID: PMC4442471 DOI: 10.4103/0250-474x.156600] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 11/22/2014] [Accepted: 04/02/2015] [Indexed: 11/29/2022] Open
Abstract
The present communication reports the comparison of in vivo antioxidant, antimelanoma and antimutagenic activities of Eruca sativa seed oil and its bio principles (allyl isothiocyanate, phenylethyl isothiocyanate and sulphoraphane) against B16F10 melanoma cells induced in C57BL/6 mice model. Among the various treatments considered for the study, isothiocyanates combination (allyl isothiocyanate, phenylethyl isothiocyanate and sulphoraphane; 1:1:1; 10 µM) exhibited optimum antioxidant activity, 51.95±1.14 µM glutathione per mg protein compared to seed oil 25.91±1.26 µM. Lipid peroxidation value was 9.97±1.72 µM malondialdehyde per mg wet weight for isothiocyanates combination against seed oil, 28.45±1.87 µM and rendered significant protection against oxidative stress induced by melanoma in liver tissue. Isothiocyanates combination significantly suppressed various parameters, such as tumor growth, isothiocyanates combination by 36.36% while the seed oil by 15.23%; tumor weight, isothiocyanates combination by 45.9% and seed oil by 19.6%; tumor volume, isothiocyanates combination by 41.7% while the seed oil by 32.3%, measured for antimelanoma activity at a concentration of 10 µM. Isothiocyanates combination has been found to be more cytotoxic bioagent against B16F10 melanoma cells induced in C57BL/6 mice compared to naturally occurring Eruca sativa seed oil.
Collapse
Affiliation(s)
- Prachi Bansal
- Department of Chemistry, Dayalbagh Educational Institute, Dayalbagh, Agra-282 110, India
| | - S Medhe
- Department of Chemistry, Dayalbagh Educational Institute, Dayalbagh, Agra-282 110, India
| | - N Ganesh
- Department of Research, Jawaharlal Nehru Cancer Hospital & Research Centre, Bhopal-462 001, India
| | - M M Srivastava
- Department of Chemistry, Dayalbagh Educational Institute, Dayalbagh, Agra-282 110, India
| |
Collapse
|
16
|
Gras Navarro A, Björklund AT, Chekenya M. Therapeutic potential and challenges of natural killer cells in treatment of solid tumors. Front Immunol 2015; 6:202. [PMID: 25972872 PMCID: PMC4413815 DOI: 10.3389/fimmu.2015.00202] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/14/2015] [Indexed: 12/22/2022] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells that hold tremendous potential for effective immunotherapy for a broad range of cancers. Due to the mode of NK cell killing, requiring one-to-one target engagement and site-directed release of cytolytic granules, the therapeutic potential of NK cells has been most extensively explored in hematological malignancies. However, their ability to precisely kill antibody coated cells, cancer stem cells, and genotoxically altered cells, while maintaining tolerance to healthy cells makes them appealing therapeutic effectors for all cancer forms, including metastases. Due to their release of pro-inflammatory cytokines, NK cells may potently reverse the anti-inflammatory tumor microenvironment (TME) and augment adaptive immune responses by promoting differentiation, activation, and/or recruitment of accessory immune cells to sites of malignancy. Nevertheless, integrated and coordinated mechanisms of subversion of NK cell activity against the tumor and its microenvironment exist. Although our understanding of the receptor ligand interactions that regulate NK cell functionality has evolved remarkably, the diversity of ligands and receptors is complex, as is their mechanistic foundations in regulating NK cell function. In this article, we review the literature and highlight how the TME manipulates the NK cell phenotypes, genotypes, and tropism to evade tumor recognition and elimination. We discuss counter strategies that may be adopted to augment the efficacy of NK cell anti-tumor surveillance, the clinical trials that have been undertaken so far in solid malignancies, critically weighing the challenges and opportunities with this approach.
Collapse
Affiliation(s)
| | - Andreas T Björklund
- Karolinska University Hospital, Hematology Center and Karolinska Institute , Stockholm , Sweden
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen , Bergen , Norway
| |
Collapse
|
17
|
Nicolosi PA, Dallatomasina A, Perris R. Theranostic impact of NG2/CSPG4 proteoglycan in cancer. Theranostics 2015; 5:530-44. [PMID: 25767619 PMCID: PMC4350014 DOI: 10.7150/thno.10824] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/03/2014] [Indexed: 12/27/2022] Open
Abstract
NG2/CSPG4 is an unusual cell-membrane integral proteoglycan widely recognized to be a prognostic factor, a valuable tool for ex vivo and non-invasive molecular diagnostics and, by virtue of its tight association with malignancy, a tantalizing therapeutic target in several tumour types. Although the biology behind its involvement in cancer progression needs to be better understood, implementation of NG2/CSPG4 in the routine clinical practice is attainable and has the potential to contribute to an improved individualized management of cancer patients. In this context, its polymorphic nature seems to be particularly valuable in the effort to standardize informative diagnostic procedures and consolidate forcible immunotherapeutic treatment strategies. We discuss here the underpinnings for this potential and highlight the benefits of taking advantage of the intra-tumour and inter-patient variability in the regulation of NG2/CSPG4 expression. We envision that NG2/CSPG4 may effectively be exploited in therapeutic interventions aimed at averting resistance to target therapy agents and at interfering with secondary lesion formation and/or tumour recurrence.
Collapse
|
18
|
Rygh CB, Wang J, Thuen M, Gras Navarro A, Huuse EM, Thorsen F, Poli A, Zimmer J, Haraldseth O, Lie SA, Enger PØ, Chekenya M. Dynamic contrast enhanced MRI detects early response to adoptive NK cellular immunotherapy targeting the NG2 proteoglycan in a rat model of glioblastoma. PLoS One 2014; 9:e108414. [PMID: 25268630 PMCID: PMC4182474 DOI: 10.1371/journal.pone.0108414] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 08/27/2014] [Indexed: 01/05/2023] Open
Abstract
There are currently no established radiological parameters that predict response to immunotherapy. We hypothesised that multiparametric, longitudinal magnetic resonance imaging (MRI) of physiological parameters and pharmacokinetic models might detect early biological responses to immunotherapy for glioblastoma targeting NG2/CSPG4 with mAb9.2.27 combined with natural killer (NK) cells. Contrast enhanced conventional T1-weighted MRI at 7±1 and 17±2 days post-treatment failed to detect differences in tumour size between the treatment groups, whereas, follow-up scans at 3 months demonstrated diminished signal intensity and tumour volume in the surviving NK+mAb9.2.27 treated animals. Notably, interstitial volume fraction (ve), was significantly increased in the NK+mAb9.2.27 combination therapy group compared mAb9.2.27 and NK cell monotherapy groups (p = 0.002 and p = 0.017 respectively) in cohort 1 animals treated with 1 million NK cells. ve was reproducibly increased in the combination NK+mAb9.2.27 compared to NK cell monotherapy in cohort 2 treated with increased dose of 2 million NK cells (p<0.0001), indicating greater cell death induced by NK+mAb9.2.27 treatment. The interstitial volume fraction in the NK monotherapy group was significantly reduced compared to mAb9.2.27 monotherapy (p<0.0001) and untreated controls (p = 0.014) in the cohort 2 animals. NK cells in monotherapy were unable to kill the U87MG cells that highly expressed class I human leucocyte antigens, and diminished stress ligands for activating receptors. A significant association between apparent diffusion coefficient (ADC) of water and ve in combination NK+mAb9.2.27 and NK monotherapy treated tumours was evident, where increased ADC corresponded to reduced ve in both cases. Collectively, these data support histological measures at end-stage demonstrating diminished tumour cell proliferation and pronounced apoptosis in the NK+mAb9.2.27 treated tumours compared to the other groups. In conclusion, ve was the most reliable radiological parameter for detecting response to intralesional NK cellular therapy.
Collapse
Affiliation(s)
- Cecilie Brekke Rygh
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Cardiovascular Research Group, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| | - Jian Wang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Marte Thuen
- MI Lab, Department of Circulation and Medical Imaging, NTNU, Trondheim, Norway
| | | | - Else Marie Huuse
- MI Lab, Department of Circulation and Medical Imaging, NTNU, Trondheim, Norway
| | - Frits Thorsen
- Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Aurelie Poli
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Laboratoire d'Immunogénétique-Allergologie, CRP-Santé, Luxembourg City, Luxembourg
| | - Jacques Zimmer
- Laboratoire d'Immunogénétique-Allergologie, CRP-Santé, Luxembourg City, Luxembourg
| | - Olav Haraldseth
- MI Lab, Department of Circulation and Medical Imaging, NTNU, Trondheim, Norway
- Department of Medical Imaging, St. Olavs Hospital, Trondheim, Norway
| | - Stein Atle Lie
- Institute for Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Per Øyvind Enger
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Institute for Clinical Dentistry, University of Bergen, Bergen, Norway
| |
Collapse
|
19
|
Tumor bioengineering using a transglutaminase crosslinked hydrogel. PLoS One 2014; 9:e105616. [PMID: 25133673 PMCID: PMC4136878 DOI: 10.1371/journal.pone.0105616] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 07/25/2014] [Indexed: 11/19/2022] Open
Abstract
Development of a physiologically relevant 3D model system for cancer research and drug development is a current challenge. We have adopted a 3D culture system based on a transglutaminase-crosslinked gelatin gel (Col-Tgel) to mimic the tumor 3D microenvironment. The system has several unique advantages over other alternatives including presenting cell-matrix interaction sites from collagen-derived peptides, geometry-initiated multicellular tumor spheroids, and metabolic gradients in the tumor microenvironment. Also it provides a controllable wide spectrum of gel stiffness for mechanical signals, and technical compatibility with imaging based screening due to its transparent properties. In addition, the Col-Tgel provides a cure-in-situ delivery vehicle for tumor xenograft formation in animals enhancing tumor cell uptake rate. Overall, this distinctive 3D system could offer a platform to more accurately mimic in vivo situations to study tumor formation and progression both in vitro and in vivo.
Collapse
|
20
|
Stapor PC, Sweat RS, Dashti DC, Betancourt AM, Murfee WL. Pericyte dynamics during angiogenesis: new insights from new identities. J Vasc Res 2014; 51:163-74. [PMID: 24853910 DOI: 10.1159/000362276] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 03/11/2014] [Indexed: 12/13/2022] Open
Abstract
Therapies aimed at manipulating the microcirculation require the ability to control angiogenesis, defined as the sprouting of new capillaries from existing vessels. Blocking angiogenesis would be beneficial in many pathologies (e.g. cancer, retinopathies and rheumatoid arthritis). In others (e.g. myocardial infarction, stroke and hypertension), promoting angiogenesis would be desirable. We know that vascular pericytes elongate around endothelial cells (ECs) and are functionally associated with regulating vessel stabilization, vessel diameter and EC proliferation. During angiogenesis, bidirectional pericyte-EC signaling is critical for capillary sprout formation. Observations of pericytes leading capillary sprouts also implicate their role in EC guidance. As such, pericytes have recently emerged as a therapeutic target to promote or inhibit angiogenesis. Advancing our basic understanding of pericytes and developing pericyte-related therapies are challenged, like in many other fields, by questions regarding cell identity. This review article discusses what we know about pericyte phenotypes and the opportunity to advance our understanding by defining the specific pericyte cell populations involved in capillary sprouting.
Collapse
Affiliation(s)
- Peter C Stapor
- Department of Biomedical Engineering, Tulane University, Lindy Boggs Center, New Orleans, La., USA
| | | | | | | | | |
Collapse
|
21
|
Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia 2013; 59:1169-80. [PMID: 22379614 DOI: 10.1002/glia.21136] [Citation(s) in RCA: 372] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 12/08/2010] [Indexed: 02/06/2023]
Abstract
High-grade brain tumors are heterogeneous with respect to the composition of bona fide tumor cells and with respect to a range of intermingling parenchymal cells. Glioblastomas harbor multiple cell types, some with increased tumorigenicity and stem cell-like capacity. The stem-like cells maybe the cells of origin for tumor relapse. However, the tumor-associated parenchymal cells such as vascular cells,microglia, peripheral immune cells, and neural precursor cells also play a vital role in controlling the course of pathology.In this review, we describe the multiple interactions of bulk glioma cells and glioma stem cells with parenchymal cell populations and highlight the pathological impact as well as signaling pathways known for these types of cell-cell communication. The tumor-vasculature not only nourishes glioblastomas, but also provides a specialized niche for these stem-like cells. In addition, microglial cells,which can contribute up to 30% of a brain tumor mass,play a role in glioblastoma cell invasion. Moreover, non-neoplastic astrocytes can be converted into a reactive phenotype by the glioma microenvironment and can then secrete a number of factors which influences tumor biology. The young brain may have the capacity to inhibit gliomagenesis by the endogenous neural precursor cells, which secrete tumor suppressive factors. The factors, pathways, and interactions described in this review provide a new prospective on the cell biology of primary brain tumors, which may ultimately generate new treatment modalities. However, our picture of the multiple interactions between parenchymal and tumor cells is still incomplete.
Collapse
Affiliation(s)
- Nikki A Charles
- Brain Tumor Center and Department of Neurosurgery, Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | |
Collapse
|
22
|
Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia 2013; 60:502-14. [PMID: 22379614 DOI: 10.1002/glia.21264] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
High-grade brain tumors are heterogeneous with respect to the composition of bona fide tumor cells and with respect to a range of intermingling parenchymal cells. Glioblastomas harbor multiple cell types, some with increased tumorigenicity and stem cell-like capacity. The stem-like cells maybe the cells of origin for tumor relapse. However, the tumor-associated parenchymal cells such as vascular cells,microglia, peripheral immune cells, and neural precursor cells also play a vital role in controlling the course of pathology.In this review, we describe the multiple interactions of bulk glioma cells and glioma stem cells with parenchymal cell populations and highlight the pathological impact as well as signaling pathways known for these types of cell-cell communication. The tumor-vasculature not only nourishes glioblastomas, but also provides a specialized niche for these stem-like cells. In addition, microglial cells,which can contribute up to 30% of a brain tumor mass,play a role in glioblastoma cell invasion. Moreover, non-neoplastic astrocytes can be converted into a reactive phenotype by the glioma microenvironment and can then secrete a number of factors which influences tumor biology. The young brain may have the capacity to inhibit gliomagenesis by the endogenous neural precursor cells, which secrete tumor suppressive factors. The factors, pathways, and interactions described in this review provide a new prospective on the cell biology of primary brain tumors, which may ultimately generate new treatment modalities. However, our picture of the multiple interactions between parenchymal and tumor cells is still incomplete.
Collapse
Affiliation(s)
- Nikki A Charles
- Brain Tumor Center and Department of Neurosurgery, Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | |
Collapse
|
23
|
Kociński M, Klepaczko A, Materka A, Chekenya M, Lundervold A. 3D image texture analysis of simulated and real-world vascular trees. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2012; 107:140-154. [PMID: 21803438 DOI: 10.1016/j.cmpb.2011.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 05/04/2011] [Accepted: 06/06/2011] [Indexed: 05/31/2023]
Abstract
A method is proposed for quantitative description of blood-vessel trees, which can be used for tree classification and/or physical parameters indirect monitoring. The method is based on texture analysis of 3D images of the trees. Several types of trees were defined, with distinct tree parameters (number of terminal branches, blood viscosity, input and output flow). A number of trees were computer-simulated for each type. 3D image was computed for each tree and its texture features were calculated. Best discriminating features were found and applied to 1-NN nearest neighbor classifier. It was demonstrated that (i) tree images can be correctly classified for realistic signal-to-noise ratio, (ii) some texture features are monotonously related to tree parameters, (iii) 2D texture analysis is not sufficient to represent the trees in the discussed sense. Moreover, applicability of texture model to quantitative description of vascularity images was also supported by unsupervised exploratory analysis. Eventually, the experimental confirmation was done, with the use of confocal microscopy images of rat brain vasculature. Several classes of brain tissue were clearly distinguished based on 3D texture numerical parameters, including control and different kinds of tumours - treated with NG2 proteoglycan to promote angiogenesis-dependent growth of the abnormal tissue. The method, applied to magnetic resonance imaging e.g. real neovasculature or retinal images can be used to support noninvasive medical diagnosis of vascular system diseases.
Collapse
Affiliation(s)
- Marek Kociński
- Institute of Electronics, Technical University of Lodz, Lodz, Poland.
| | | | | | | | | |
Collapse
|
24
|
Xu JP, Zhao J, Li S. Roles of NG2 glial cells in diseases of the central nervous system. Neurosci Bull 2012; 27:413-21. [PMID: 22108818 DOI: 10.1007/s12264-011-1838-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
NG2 cells are a novel distinct class of central nervous system (CNS) glial cells, characterized by the expression of the chondroitin sulfate proteoglycan NG2. They have been detected in a variety of human CNS diseases. As morphological, physiological and biomolecular studies of NG2 cells have been conducted, their roles have been gradually revealed. Research on cellular and molecular mechanisms in the pathophysiological state was built on the preliminary findings of their physiological functions; and in turn, this helps to clarify their physiological roles and leads to the identification of novel therapeutic targets. This review summarizes recent findings regarding the potential roles of NG2 cells in traumatic brain injury, multiple sclerosis, glioma, epilepsy, Alzheimer's disease and electroconvulsive therapy for depression.
Collapse
Affiliation(s)
- Jian-Ping Xu
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | | | | |
Collapse
|
25
|
Dutoit V, Herold-Mende C, Hilf N, Schoor O, Beckhove P, Bucher J, Dorsch K, Flohr S, Fritsche J, Lewandrowski P, Lohr J, Rammensee HG, Stevanovic S, Trautwein C, Vass V, Walter S, Walker PR, Weinschenk T, Singh-Jasuja H, Dietrich PY. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. ACTA ACUST UNITED AC 2012; 135:1042-54. [PMID: 22418738 DOI: 10.1093/brain/aws042] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Peptides presented at the cell surface reflect the protein content of the cell; those on HLA class I molecules comprise the critical peptidome elements interacting with CD8 T lymphocytes. We hypothesize that peptidomes from ex vivo tumour samples encompass immunogenic tumour antigens. Here, we uncover >6000 HLA-bound peptides from HLA-A*02(+) glioblastoma, of which over 3000 were restricted by HLA-A*02. We prioritized in-depth investigation of 10 glioblastoma-associated antigens based on high expression in tumours, very low or absent expression in healthy tissues, implication in gliomagenesis and immunogenicity. Patients with glioblastoma showed no T cell tolerance to these peptides. Moreover, we demonstrated specific lysis of tumour cells by patients' CD8(+) T cells in vitro. In vivo, glioblastoma-specific CD8(+) T cells were present at the tumour site. Overall, our data show the physiological relevance of the peptidome approach and provide a critical advance for designing a rational glioblastoma immunotherapy. The peptides identified in our study are currently being tested as a multipeptide vaccine (IMA950) in patients with glioblastoma.
Collapse
Affiliation(s)
- Valérie Dutoit
- Laboratory of Tumour Immunology, Centre of Oncology, Geneva University Hospital, 1211 Geneva 14, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Le Jan S, Hayashi M, Kasza Z, Eriksson I, Bishop JR, Weibrecht I, Heldin J, Holmborn K, Jakobsson L, Söderberg O, Spillmann D, Esko JD, Claesson-Welsh L, Kjellén L, Kreuger J. Functional overlap between chondroitin and heparan sulfate proteoglycans during VEGF-induced sprouting angiogenesis. Arterioscler Thromb Vasc Biol 2012; 32:1255-63. [PMID: 22345168 DOI: 10.1161/atvbaha.111.240622] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Heparan sulfate proteoglycans regulate key steps of blood vessel formation. The present study was undertaken to investigate if there is a functional overlap between heparan sulfate proteoglycans and chondroitin sulfate proteoglycans during sprouting angiogenesis. METHODS AND RESULTS Using cultures of genetically engineered mouse embryonic stem cells, we show that angiogenic sprouting occurs also in the absence of heparan sulfate biosynthesis. Cells unable to produce heparan sulfate instead increase their production of chondroitin sulfate that binds key angiogenic growth factors such as vascular endothelial growth factor A, transforming growth factor β, and platelet-derived growth factor B. Lack of heparan sulfate proteoglycan production however leads to increased pericyte numbers and reduced adhesion of pericytes to nascent sprouts, likely due to dysregulation of transforming growth factor β and platelet-derived growth factor B signal transduction. CONCLUSIONS The present study provides direct evidence for a previously undefined functional overlap between chondroitin sulfate proteoglycans and heparan sulfate proteoglycans during sprouting angiogenesis. Our findings provide information relevant for potential future drug design efforts that involve targeting of proteoglycans in the vasculature.
Collapse
Affiliation(s)
- Sébastien Le Jan
- Rudbeck Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Svendsen A, Verhoeff JJC, Immervoll H, Brøgger JC, Kmiecik J, Poli A, Netland IA, Prestegarden L, Planagumà J, Torsvik A, Kjersem AB, Sakariassen PØ, Heggdal JI, Van Furth WR, Bjerkvig R, Lund-Johansen M, Enger PØ, Felsberg J, Brons NHC, Tronstad KJ, Waha A, Chekenya M. Expression of the progenitor marker NG2/CSPG4 predicts poor survival and resistance to ionising radiation in glioblastoma. Acta Neuropathol 2011; 122:495-510. [PMID: 21863242 PMCID: PMC3185228 DOI: 10.1007/s00401-011-0867-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/15/2011] [Accepted: 08/15/2011] [Indexed: 12/05/2022]
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumour, where patients respond poorly to radiotherapy and exhibit dismal survival outcomes. The mechanisms of radioresistance are not completely understood. However, cancer cells with an immature stem-like phenotype are hypothesised to play a role in radioresistance. Since the progenitor marker neuron-glial-2 (NG2) has been shown to regulate several aspects of GBM progression in experimental systems, we hypothesised that its expression would influence the survival of GBM patients. Quantification of NG2 expression in 74 GBM biopsies from newly diagnosed and untreated patients revealed that 50% express high NG2 levels on tumour cells and associated vessels, being associated with significantly shorter survival. This effect was independent of age at diagnosis, treatment received and hypermethylation of the O(6)-methylguanine methyltransferase (MGMT) DNA repair gene promoter. NG2 was frequently co-expressed with nestin and vimentin but rarely with CD133 and the NG2 positive tumour cells harboured genetic aberrations typical for GBM. 2D proteomics of 11 randomly selected biopsies revealed upregulation of an antioxidant, peroxiredoxin-1 (PRDX-1), in the shortest surviving patients. Expression of PRDX-1 was associated with significantly reduced products of oxidative stress. Furthermore, NG2 expressing GBM cells showed resistance to ionising radiation (IR), rapidly recognised DNA damage and effectuated cell cycle checkpoint signalling. PRDX-1 knockdown transiently slowed tumour growth rates and sensitised them to IR in vivo. Our data establish NG2 as an important prognostic factor for GBM patient survival, by mediating resistance to radiotherapy through induction of ROS scavenging enzymes and preferential DNA damage signalling.
Collapse
Affiliation(s)
- Agnete Svendsen
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Joost J. C. Verhoeff
- Laboratory of Experimental Oncology and Radiobiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Heike Immervoll
- Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Jan C. Brøgger
- Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Justyna Kmiecik
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Aurelie Poli
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- Centre de Recherche de Public de la Santé, Luxembourg, Haukeland University Hospital, Bergen, Norway
| | - Inger A. Netland
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Lars Prestegarden
- Department of Dermatology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Jesús Planagumà
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Anja Torsvik
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | | | | | - Jan I. Heggdal
- Department of Oncology and Medical Physics, University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter R. Van Furth
- Department of Neurosurgery, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- Centre de Recherche de Public de la Santé, Luxembourg, Haukeland University Hospital, Bergen, Norway
| | | | - Per Ø. Enger
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- Department of Neurosurgery, Haukeland University Hospital, 5021 Bergen, Norway
| | - Joerg Felsberg
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nicolaas H. C. Brons
- Centre de Recherche de Public de la Santé, Luxembourg, Haukeland University Hospital, Bergen, Norway
| | - Karl J. Tronstad
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Andreas Waha
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- Translational Cancer Research Group, Jonas Lies vei 91, 5009 Bergen, Norway
| |
Collapse
|
28
|
Al-Mayhani MTF, Grenfell R, Narita M, Piccirillo S, Kenney-Herbert E, Fawcett JW, Collins VP, Ichimura K, Watts C. NG2 expression in glioblastoma identifies an actively proliferating population with an aggressive molecular signature. Neuro Oncol 2011; 13:830-45. [PMID: 21798846 PMCID: PMC3145476 DOI: 10.1093/neuonc/nor088] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 05/13/2011] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of primary brain tumor and a highly malignant and heterogeneous cancer. Current conventional therapies fail to eradicate or curb GBM cell growth. Hence, exploring the cellular and molecular basis of GBM cell growth is vital to develop novel therapeutic approaches. Neuroglia (NG)-2 is a transmembrane proteoglycan expressed by NG2+ progenitors and is strongly linked to cell proliferation in the normal brain. By using NG2 as a biomarker we identify a GBM cell population (GBM NG2+ cells) with robust proliferative, clonogenic, and tumorigenic capacity. We show that a significant proportion (mean 83%) of cells proliferating in the tumor mass express NG2 and that over 50% of GBM NG2+ cells are proliferating. Compared with the GBM NG2- cells from the same tumor, the GBM of NG2+ cells overexpress genes associated with aggressive tumorigenicity, including overexpression of Mitosis and Cell Cycling Module genes (e.g., MELK, CDC, MCM, E2F), which have been previously shown to correlate with poor survival in GBM. We also show that the coexpression pattern of NG2 with other glial progenitor markers in GBM does not recapitulate that described in the normal brain. The expression of NG2 by such an aggressive and actively cycling GBM population combined with its location on the cell surface identifies this cell population as a potential therapeutic target in a subset of patients with GBM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Colin Watts
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge (M.T.F.A-M., S.P., E.K-H., J.W.F., C.W.); MRC Laboratory of Molecular Biology, University of Cambridge (R.G.); CRUK Cancer Research Institute, University of Cambridge (M.N.); Division of Molecular Histopathology, Department of Pathology, University of Cambridge (V.P.C., K.I.); Department of Neurosurgery, University of Cambridge (C.W.), Cambridge, UK
| |
Collapse
|
29
|
Wang J, Svendsen A, Kmiecik J, Immervoll H, Skaftnesmo KO, Planagumà J, Reed RK, Bjerkvig R, Miletic H, Enger PØ, Rygh CB, Chekenya M. Targeting the NG2/CSPG4 proteoglycan retards tumour growth and angiogenesis in preclinical models of GBM and melanoma. PLoS One 2011; 6:e23062. [PMID: 21829586 PMCID: PMC3146530 DOI: 10.1371/journal.pone.0023062] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 07/11/2011] [Indexed: 02/07/2023] Open
Abstract
Aberrant expression of the progenitor marker Neuron-glia 2 (NG2/CSPG4) or melanoma proteoglycan on cancer cells and angiogenic vasculature is associated with an aggressive disease course in several malignancies including glioblastoma multiforme (GBM) and melanoma. Thus, we investigated the mechanism of NG2 mediated malignant progression and its potential as a therapeutic target in clinically relevant GBM and melanoma animal models. Xenografting NG2 overexpressing GBM cell lines resulted in increased growth rate, angiogenesis and vascular permeability compared to control, NG2 negative tumours. The effect of abrogating NG2 function was investigated after intracerebral delivery of lentivirally encoded shRNAs targeting NG2 in patient GBM xenografts as well as in established subcutaneous A375 melanoma tumours. NG2 knockdown reduced melanoma proliferation and increased apoptosis and necrosis. Targeting NG2 in two heterogeneous GBM xenografts significantly reduced tumour growth and oedema levels, angiogenesis and normalised vascular function. Vascular normalisation resulted in increased tumour invasion and decreased apoptosis and necrosis. We conclude that NG2 promotes tumour progression by multiple mechanisms and represents an amenable target for cancer molecular therapy.
Collapse
Affiliation(s)
- Jian Wang
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Agnete Svendsen
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Justyna Kmiecik
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Heike Immervoll
- Haukeland University Hospital, Department of Pathology, Bergen, Norway
- The Gades Institute, University of Bergen, Bergen, Norway
| | - Kai Ove Skaftnesmo
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jesús Planagumà
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Rolf Kåre Reed
- Heart and Circulatory Research Group, Haukeland University Hospital, Bergen, Norway
| | - Rolf Bjerkvig
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
- Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - Hrvoje Miletic
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
- Haukeland University Hospital, Department of Pathology, Bergen, Norway
| | - Per Øyvind Enger
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway
| | - Cecilie Brekke Rygh
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
- Heart and Circulatory Research Group, Haukeland University Hospital, Bergen, Norway
| | - Martha Chekenya
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
- * E-mail:
| |
Collapse
|
30
|
Abstract
The lack of effective conventional therapies for the treatment of advanced stage melanoma has stimulated interest in the development of novel strategies for the management of patients with malignant melanoma. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma. For many years, T cell-based immunotherapy has been emphasized in part because of the disappointing results of the monoclonal antibody (mAb)-based clinical trials conducted in the early 1980s and in part because of the postulated major role played by T cells in tumor growth control. More recently, mAb-based therapies have gained in popularity given their clinical and commercial success for a variety of malignant diseases. As a result, there has been increased interest in identifying and characterizing antibody-defined melanoma antigens. Among them, the chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen (HMW-MAA) or melanoma chondroitin sulfate proteoglycan (MCSP), has attracted much attention in recent years because of the growing experimental evidence that it fulfills two requirements for immunotherapy to be therapeutically effective: (1) targeting of cancer stem cells (CSC) and (2) development of combinatorial therapies to counteract the escape mechanisms driven by the genetic instability of tumor cells. With this in mind, in this chapter, we have reviewed recent information related to the distribution of CSPG4 on various types of tumors, including CSC, its expression on pericytes in the tumor microenvironment, its recognition by T cells, its role in cell biology as well as the potential mechanisms underlying the ability of CSPG4-specific immunity to control malignant cell growth.
Collapse
|
31
|
Terrile M, Appolloni I, Calzolari F, Perris R, Tutucci E, Malatesta P. PDGF-B-driven gliomagenesis can occur in the absence of the proteoglycan NG2. BMC Cancer 2010; 10:550. [PMID: 20939912 PMCID: PMC2964636 DOI: 10.1186/1471-2407-10-550] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 10/12/2010] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In the last years, the transmembrane proteoglycan NG2 has gained interest as a therapeutic target for the treatment of diverse tumor types, including gliomas, because increases of its expression correlate with dismal prognosis. NG2 has been shown to function as a co-receptor for PDGF ligands whose aberrant expression is common in gliomas. We have recently generated a glioma model based on the overexpression of PDGF-B in neural progenitors and here we investigated the possible relevance of NG2 during PDGF-driven gliomagenesis. METHODS The survival curves of NG2-KO mice overexpressing PDGF-B were compared to controls by using a Log-rank test. The characteristics of tumors induced in NG2-KO were compared to those of tumors induced in wild type mice by immunostaining for different cell lineage markers and by transplantation assays in adult mice. RESULTS We showed that the lack of NG2 does not appreciably affect any of the characterized steps of PDGF-driven brain tumorigenesis, such as oligodendrocyte progenitor cells (OPC) induction, the recruitment of bystander OPCs and the progression to full malignancy, which take place as in wild type animals. CONCLUSIONS Our analysis, using both NG2-KO mice and a miRNA based silencing approach, clearly demonstrates that NG2 is not required for PDGF-B to efficiently induce and maintain gliomas from neural progenitors. On the basis of the data obtained, we therefore suggest that the role of NG2 as a target molecule for glioma treatment should be carefully reconsidered.
Collapse
Affiliation(s)
- Marta Terrile
- National Institute for Cancer Research IST, IRCCS, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | | | | | | | | | | |
Collapse
|
32
|
Huang FJ, You WK, Bonaldo P, Seyfried TN, Pasquale EB, Stallcup WB. Pericyte deficiencies lead to aberrant tumor vascularizaton in the brain of the NG2 null mouse. Dev Biol 2010; 344:1035-46. [PMID: 20599895 DOI: 10.1016/j.ydbio.2010.06.023] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/16/2010] [Accepted: 06/21/2010] [Indexed: 01/12/2023]
Abstract
Tightly regulated crosstalk between endothelial cells and pericytes is required for formation and maintenance of functional blood vessels. When the NG2 proteoglycan is absent from pericyte surfaces, vascularization of syngeneic tumors growing in the C57Bl/6 mouse brain is aberrant in several respects, resulting in retardation of tumor progression. In the NG2 null mouse brain, pericyte investment of the tumor vascular endothelium is reduced, causing deficiencies in both pericyte and endothelial cell maturation, as well as reduced basal lamina assembly. While part of this deficit may be due to the previously-identified role of NG2 in beta1 integrin-dependent periyte/endothelial cell crosstalk, the ablation of NG2 also appears responsible for loss of collagen VI anchorage, in turn leading to reduced collagen IV deposition. Poor functionality of tumor vessels in NG2 null brain is reflected by reduced vessel patency and increased vessel leakiness, resulting in large increases in tumor hypoxia. These findings demonstrate the importance of NG2-dependent pericyte/endothelial cell interaction in the development and maturation of tumor blood vessels, identifying NG2 as a potential target for anti-angiogenic cancer therapy.
Collapse
Affiliation(s)
- Feng-Ju Huang
- Sanford-Burnham Medical Research Institute, Cancer Center, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
33
|
Prestegarden L, Svendsen A, Wang J, Sleire L, Skaftnesmo KO, Bjerkvig R, Yan T, Askland L, Persson A, Sakariassen PØ, Enger PØ. Glioma Cell Populations Grouped by Different Cell Type Markers Drive Brain Tumor Growth. Cancer Res 2010; 70:4274-9. [DOI: 10.1158/0008-5472.can-09-3904] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Angiogenesis inhibitor DC101 delays growth of intracerebral glioblastoma but induces morbidity when combined with irradiation. Cancer Lett 2009; 285:39-45. [DOI: 10.1016/j.canlet.2009.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 04/28/2009] [Accepted: 04/29/2009] [Indexed: 11/15/2022]
|
35
|
Verhoeff JJC, Stalpers LJA, Claes A, Hovinga KE, Musters GD, Peter Vandertop W, Richel DJ, Leenders WPJ, van Furth WR. Tumour control by whole brain irradiation of anti-VEGF-treated mice bearing intracerebral glioma. Eur J Cancer 2009; 45:3074-80. [PMID: 19734041 DOI: 10.1016/j.ejca.2009.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 07/23/2009] [Accepted: 08/06/2009] [Indexed: 11/18/2022]
Abstract
AIM OF THE STUDY Tumour angiogenesis and invasion are key features of glioblastoma multiforme (GBM). Angiogenesis inhibitors increase progression-free survival (PFS) of recurrent GBM patients. VEGF inhibition controls the bulk tumour growth by inhibition of angiogenesis, but does not inhibit the invasive tumour component. We investigated if invasive tumour growth can be controlled by combining anti-VEGF treatment with irradiation of tumour plus surrounding brain in an orthotopic murine model for GBM. METHODS AND MATERIALS GBM cell line U251-NG2 was inoculated through a guide screw in the right frontal lobe of 53 athymic nude mice. Pegaptanib (a slow-releasing aptamer against VEGF) was injected in the tumour bed either or not followed by irradiation treatment with implanted I-125 seeds. Pegaptanib and/or irradiation were compared with sham-treated controls, resulting in four groups of 10-15 mice each. After 6 weeks of treatment, histological analysis was performed on all brains. RESULTS VEGF inhibition by locally deposited pegaptanib decreased tumour blood vessel density, and increased tumour hypoxia. Pegaptanib treatment still allowed the formation of tumour satellites. Irradiation decreased tumour size and suppressed formation of satellites. Combined pegaptanib plus irradiation further increased PFS. Tumour size directly correlated with PFS. CONCLUDING STATEMENT The anti-tumour effects of local VEGF inhibition are partially circumvented by the formation of invasive tumour satellites. Additional irradiation is effective in slowing down proliferation of these invasive tumour components.
Collapse
Affiliation(s)
- Joost J C Verhoeff
- Department of Radiation Oncology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Tate MC, Aghi MK. Biology of angiogenesis and invasion in glioma. Neurotherapeutics 2009; 6:447-57. [PMID: 19560735 PMCID: PMC5084181 DOI: 10.1016/j.nurt.2009.04.001] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 03/30/2009] [Accepted: 04/09/2009] [Indexed: 01/12/2023] Open
Abstract
Treatment of adult brain tumors, in particular glioblastoma, remains a significant clinical challenge, despite modest advances in surgical technique, radiation, and chemotherapeutics. The formation of abnormal, dysfunctional tumor vasculature and glioma cell invasion along white matter tracts are believed to be major components of the inability to treat these tumors effectively. Recent insight into the fundamental processes governing glioma angiogenesis and invasion provide a renewed hope for development of novel strategies aimed at reducing the morbidity of this uniformly fatal disease. In this review, we discuss background biology of the blood brain barrier and its pertinence to blood vessel formation and tumor invasion. We will then focus our attention on the biology of glioma angiogenesis and invasion, and the key mediators of these processes. Last, we will briefly discuss recent and ongoing clinical trials targeting mediators of angiogenesis or invasion in glioma patients. The findings provide a renewed hope for those endeavoring to improve treatment of patients with glioma by providing a novel set of rational targets for translational drug discovery.
Collapse
Affiliation(s)
- Matthew C. Tate
- grid.266102.10000000122976811Department of Neurological Surgery, University of California, 505 Parnassus Avenue, Room M779, 94143-0112 San Francisco, CA
| | - Manish K. Aghi
- grid.266102.10000000122976811Department of Neurological Surgery, University of California, 505 Parnassus Avenue, Room M779, 94143-0112 San Francisco, CA
| |
Collapse
|
37
|
Erfurt C, Müller E, Emmerling S, Klotz C, Hertl M, Schuler G, Schultz ES. Melanoma-associated chondroitin sulphate proteoglycan as a new target antigen for CD4+ T cells in melanoma patients. Int J Cancer 2009; 124:2341-6. [PMID: 19173283 DOI: 10.1002/ijc.24235] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Melanoma-associated chondroitin sulfate proteoglycan (MCSP) (also known as high molecular weight-melanoma-associated antigen) represents an interesting target antigen for cancer immunotherapy which is expressed on human melanomas and other tumors such as breast carcinomas, gliomas, neuroblastomas and acute leukemias. MCSP seems to play an important functional role in melanoma as it is involved in tumor cell migration, invasion and angiogenesis. In this study, we isolated CD4(+) T helper cells from the blood of a healthy donor, recognizing a peptide from the MCSP core protein presented by HLA-DBR1*1101 molecules. T cell reactivity against the identified peptide could be detected in the blood of healthy donors and melanoma patients. MCSP specific T cells from the blood of a patient could be readily expanded by repeated peptide stimulation and recognized MCSP and HLA-DR expressing tumor cells. Our findings suggest that vaccination against MCSP helper T cell epitopes might be a promising approach to fight melanoma.
Collapse
Affiliation(s)
- Cornelia Erfurt
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Paul AK, Ciesielski MJ, Sajjad M, Wang X, Ferrone S, Abdel-Nabi H, Fenstermaker RA. Expression of HMP/AN2, a melanoma associated antigen, in murine cerebral gliomas: potential for radioimmunotargeting. J Neurooncol 2009; 94:21-30. [PMID: 19152070 DOI: 10.1007/s11060-009-9798-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Accepted: 12/08/2008] [Indexed: 11/28/2022]
Abstract
Human melanoma proteoglycan (HMP), a melanoma-associated antigen, is expressed in both human melanomas and gliomas. We used HMP-specific monoclonal antibody (mAb) VT68.2 to investigate whether murine GL261 cerebral gliomas express the HMP homologue AN2 and to determine whether AN2 could be targeted for selective delivery of radiation in vivo. HMP-specific mAb VT68.2 stained murine GL261 glioma cells grown in culture and intracerebrally in syngeneic C57BL/6 mice. Positron emission tomography with radiolabeled mAb VT68.2 showed high-contrast, positive images of gliomas against a negative background. At 96 h after injection, glioma uptake of radiolabeled mAb VT68.2 was 10x greater than that of the isotype control mAb and 20x greater than that detected in normal cerebral tissue. Our results show murine GL261 cerebral gliomas express AN2 and HMP-specific mAb VT68.2 accumulates selectively and specifically at high concentration and is retained within murine cerebral gliomas. Thus, HMP is a potential target for antibody-mediated selective delivery of radiation to cerebral gliomas in vivo.
Collapse
Affiliation(s)
- Asit K Paul
- Department of Nuclear Medicine, School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Antiangiogenesis approaches have the potential to be particularly effective in the treatment of glioblastoma tumours. These tumours exhibit extremely high levels of neovascularisation, which may contribute to their extremely aggressive behaviour, not only by providing oxygenation and nutrition, but also by establishing a leaky vasculature that lacks a blood-brain barrier. This leaky vasculature enables migration of tumour cells, as well as the build up of fluid, which exacerbates tissue damage due to increased intracranial pressure. Here, we discuss the considerable progress that has been made in the identification of the pro- and antiangiogenic factors produced by glioblastoma tumours and the effects of these molecules in animal models of the disease. The safety and efficacy of some of these approaches have now been demonstrated in clinical trials. However, the ability of tumours to overcome these therapies and to re-establish angiogenesis requires further clinical research regarding potential multimodality therapies, as well as basic research into the regulation of angiogenesis by as yet unidentified factors. Optimisation of noninvasive procedures for monitoring of angiogenesis would greatly facilitate such research.
Collapse
|
40
|
Stallcup WB, Huang FJ. A role for the NG2 proteoglycan in glioma progression. Cell Adh Migr 2008; 2:192-201. [PMID: 19262111 PMCID: PMC2634088 DOI: 10.4161/cam.2.3.6279] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 05/13/2008] [Indexed: 01/18/2023] Open
Abstract
Many human gliomas carry markers characteristic of oligodendrocyte progenitor cells (such as Olig-2, PDGF alpha receptor and NG2 proteoglycan), suggesting these progenitors as the cells of origin for glioma initiation. This review considers the potential roles of the NG2 proteoglycan in glioma progression. NG2 is expressed not only by glioma cells and by oligodendrocyte progenitors, but also by pericytes associated with the tumor microvasculature. The proteoglycan may therefore promote tumor vascularization and recruitment of normal progenitors to the tumor mass, in addition to mediating expansion of the transformed cell population. Along with potentiating growth factor signaling and serving as a cell surface receptor for extracellular matrix components, NG2 also has the ability to mediate activation of beta-1 integrins. These molecular interactions allow the proteoglycan to contribute to critical processes such as cell proliferation, cell motility and cell survival.
Collapse
Affiliation(s)
- William B Stallcup
- Burnham Institute for Medical Research, Cancer Research Center, La Jolla, California 92037, USA.
| | | |
Collapse
|
41
|
Chekenya M, Krakstad C, Svendsen A, Netland IA, Staalesen V, Tysnes BB, Selheim F, Wang J, Sakariassen PØ, Sandal T, Lønning PE, Flatmark T, Enger PØ, Bjerkvig R, Sioud M, Stallcup WB. The progenitor cell marker NG2/MPG promotes chemoresistance by activation of integrin-dependent PI3K/Akt signaling. Oncogene 2008; 27:5182-94. [PMID: 18469852 DOI: 10.1038/onc.2008.157] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chemoresistance represents a major problem in the treatment of many malignancies. Overcoming this obstacle will require improved understanding of the mechanisms responsible for this phenomenon. The progenitor cell marker NG2/melanoma proteoglycan (MPG) is aberrantly expressed by various tumors, but its role in cell death signaling and its potential as a therapeutic target are largely unexplored. We have assessed cytotoxic drug-induced cell death in glioblastoma spheroids from 15 patients, as well as in five cancer cell lines that differ with respect to NG2/MPG expression. The tumors were treated with doxorubicin, etoposide, carboplatin, temodal, cisplatin and tumor necrosis factor (TNF)alpha. High NG2/MPG expression correlated with multidrug resistance mediated by increased activation of alpha3beta1 integrin/PI3K signaling and their downstream targets, promoting cell survival. NG2/MPG knockdown with shRNAs incorporated into lentiviral vectors attenuated beta1 integrin signaling revealing potent antitumor effects and further sensitized neoplastic cells to cytotoxic treatment in vitro and in vivo. Thus, as a novel regulator of the antiapoptotic response, NG2/MPG may represent an effective therapeutic target in several cancer subtypes.
Collapse
Affiliation(s)
- M Chekenya
- Norlux Neuro-Oncology Group, Department of Biomedicine, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Li JL, Sainson RCA, Shi W, Leek R, Harrington LS, Preusser M, Biswas S, Turley H, Heikamp E, Hainfellner JA, Harris AL. Delta-like 4 Notch ligand regulates tumor angiogenesis, improves tumor vascular function, and promotes tumor growth in vivo. Cancer Res 2008; 67:11244-53. [PMID: 18056450 DOI: 10.1158/0008-5472.can-07-0969] [Citation(s) in RCA: 291] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The vascular endothelial growth factor (VEGF) plays a key role in tumor angiogenesis. However, clinical trials targeting the VEGF pathway are often ineffective, suggesting that other factors/pathways are also important in tumor angiogenesis. We have previously shown that the Notch ligand Delta-like 4 (DLL4) is up-regulated in tumor vasculature. Here, we show that DLL4, when expressed in tumor cells, functions as a negative regulator of tumor angiogenesis by reducing the number of blood vessels in all five types of xenografts, but acts as a positive driver for tumor growth in two of them (human glioblastoma and prostate cancer). The growth of in vivo models was not related to the effects on growth in vitro. DLL4 expressed in the tumor cells activated Notch signaling in host stromal/endothelial cells, increased blood vessel size, and improved vascular function within tumors. The promotion of tumor growth was, to some extent, due to a reduction of tumor hypoxia and apoptosis. DLL4-expressing tumor cells responded to anti-VEGF therapy with bevacizumab. A soluble form of DLL4 (D4ECD-Fc) blocked tumor growth in both bevacizumab-sensitive and bevacizumab-resistant tumors by disrupting vascular function despite increased tumor vessel density. In addition, we show that DLL4 is up-regulated in tumor cells and tumor endothelial cells of human glioblastoma. Our findings provide a rational basis for the development of novel antiangiogenic strategies via blockade of DLL4/Notch signaling and suggest that combined approaches for interrupting both DLL4 and VEGF pathways may improve antiangiogenic therapy.
Collapse
Affiliation(s)
- Ji-Liang Li
- Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wang J, Sakariassen PØ, Tsinkalovsky O, Immervoll H, Bøe SO, Svendsen A, Prestegarden L, Røsland G, Thorsen F, Stuhr L, Molven A, Bjerkvig R, Enger PØ. CD133 negative glioma cells form tumors in nude rats and give rise to CD133 positive cells. Int J Cancer 2008; 122:761-8. [PMID: 17955491 DOI: 10.1002/ijc.23130] [Citation(s) in RCA: 399] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CD133 is a cell surface marker expressed on progenitors of haematopoietic and endothelial cell lineages. Moreover, several studies have identified CD133 as a marker of brain tumor-initiating cells. In this study, human glioblastoma multiforme biopsies were engrafted intracerebrally into nude rats. The resulting tumors were serially passaged in vivo, and monitored by magnetic resonance imaging. CD133 expression was analyzed at various passages. Tumors initiated directly from the biopsies expressed little or no CD133, and showed no contrast enhancement suggesting an intact blood-brain barrier. During passaging, the tumors gradually displayed more contrast enhancement, increased angiogenesis and a shorter survival. Real-time qPCR and immunoblots showed that this was accompanied by increased CD133 expression. Primary biopsy spheroids and xenograft tumors were subsequently dissociated and flow sorted into CD133 negative and CD133 positive cell populations. Both populations incorporated BrdU in cell culture, and expressed the neural precursor marker nestin. Notably, CD133 negative cells derived from 6 different patients were tumorgenic when implanted into the rat brains. For 3 of these patients, analysis showed that the resulting tumors contained CD133 positive cells. In conclusion, we show that CD133 negative glioma cells are tumorgenic in nude rats, and that CD133 positive cells can be obtained from these tumors. Upon passaging of the tumors in vivo, CD133 expression is upregulated, coinciding with the onset of angiogenesis and a shorter survival. Thus, our findings do not suggest that CD133 expression is required for brain tumor initiation, but that it may be involved during brain tumor progression.
Collapse
Affiliation(s)
- Jian Wang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Malignant primary brain tumors, gliomas, often overexpress both platelet-derived growth factor (PDGF) ligands and receptors providing an autocrine and/or paracrine boost to tumor growth. Glioblastoma multiforme (GBM) is the most frequent glioma. Its aggressive and infiltrative growth renders it extremely difficult to treat. Median survival after diagnosis is currently only 12-14 months. The present review describes the use of retroviral tagging to identify candidate cancer-causing genes that cooperate with PDGF in brain tumor formation. Newborn mice injected intracerebrally with a Moloney murine leukemia retrovirus carrying the sis/PDGF-B oncogene and a replication competent helper virus developed brain tumors with many characteristics of human gliomas. Analysis of proviral integrations in the brain tumors identified almost 70 common insertion sites (CISs). These CISs were named brain tumor loci and harbored known but also putative novel cancer-causing genes. Microarray analysis identified differentially expressed genes in the mouse brain tumors compared to normal brain. Known tumor genes and markers of immature cells were upregulated in the tumors. Tumors developed 13-42 weeks after injection and short latency tumors were further distinguished as fast growing and GBM-like. Long latency tumors resembled slow-growing oligodendrogliomas and contained significantly less integrations as compared to short latency tumors. Several candidate genes tagged in this retroviral screen have known functions in neoplastic transformation and oncogenesis. Some candidates with a previously unknown function in tumorigenesis were found and their putative role in brain tumor formation will be discussed in this review. The results show that proviral tagging may be a useful tool in the search for candidate glioma genes.
Collapse
|
45
|
Sheng-Hua C, Yan-Bin M, Zhi-An Z, Hong Z, Dong-Fu F, Zhi-Qiang L, Xian-Hou Y. RETRACTED: Radiation-enhanced hepatocyte growth factor secretion in malignant glioma cell lines. ACTA ACUST UNITED AC 2007; 68:610-613. [PMID: 17765959 DOI: 10.1016/j.surneu.2006.12.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Accepted: 12/06/2006] [Indexed: 10/22/2022]
Abstract
BACKGROUND Postoperative radiotherapy is the standard treatment for patients with a malignant glioma. However, a malignant glioma is radioresistant and almost always recurs, even after a high dose of radiation. A malignant glioma is characterized by its proliferation, invasion and neoangiogenesis, which can be attributed to the high levels of HGF. The scope of this study is to investigate HGF secretion by malignant glioma cells with different radiosensitivity after irradiation. METHODS Three human malignant glioma cell lines (U251, U251-NG2, and BT325) were irradiated with single doses of 0, 5, 10, and 20 grays of gamma-rays from a (137)Cs source. Hepatocyte growth factor levels in medium were measured by ELISA at 24, 48, and 72 hours after radiation. Cell survival was measured by the proliferation-based assay (XTT assay) 7 days after irradiation. RESULTS After a single dose radiation, the HGF levels showed a dose-dependent increase in U251, U251-NG2, and BT325 glioma cells. Both baseline and radiation-enhanced HGF levels were about 10-fold higher in BT325 compared to U251 and U251-NG2 cells. In addition, in the XTT assay, the BT325 was more radioresistant than both U251 and U251-NG2 cell lines (dose modifying factor = 1.5 and 1.6, respectively). CONCLUSION Irradiation-enhanced HGF secretion in all 3 tested glioma cell lines (up to 7 times basal levels). It is tempting to associate the radiation-enhanced HGF secretion with an increased angiogenic potential of the tumor, which may be a factor in radioresistance.
Collapse
Affiliation(s)
- Chu Sheng-Hua
- Department of Neurosurgery, No. 3 People's Hospital Affiliated to Shanghai Jiao Tong University College of Medicine, Shanghai 201900, ROC China.
| | - Ma Yan-Bin
- Department of Neurosurgery, No. 3 People's Hospital Affiliated to Shanghai Jiao Tong University College of Medicine, Shanghai 201900, ROC China
| | - Zhu Zhi-An
- Department of Neurosurgery, No. 3 People's Hospital Affiliated to Shanghai Jiao Tong University College of Medicine, Shanghai 201900, ROC China
| | - Zhang Hong
- Department of Neurosurgery, No. 3 People's Hospital Affiliated to Shanghai Jiao Tong University College of Medicine, Shanghai 201900, ROC China
| | - Feng Dong-Fu
- Department of Neurosurgery, No. 3 People's Hospital Affiliated to Shanghai Jiao Tong University College of Medicine, Shanghai 201900, ROC China
| | - Li Zhi-Qiang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, ROC China
| | - Yuan Xian-Hou
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, ROC China
| |
Collapse
|
46
|
Dörrie J, Birkholz K, Schaft N. Strategies for immune intervention; from bench to bedside: Symposium 2006 of the Collaborative Research Center "Strategies of cellular immune intervention" July 17th-18th, Erlangen, Germany. Cancer Immunol Immunother 2007; 56:1677-85. [PMID: 17393161 PMCID: PMC11030158 DOI: 10.1007/s00262-007-0311-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Accepted: 03/03/2007] [Indexed: 10/23/2022]
Affiliation(s)
- Jan Dörrie
- Department Dermatology, University Hospital Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Katrin Birkholz
- Department Dermatology, University Hospital Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Niels Schaft
- Department Dermatology, University Hospital Erlangen, Hartmannstrasse 14, 91052 Erlangen, Germany
| |
Collapse
|
47
|
Verhoeff JJC, Stalpers LJA, Coumou AW, Koedooder K, Lavini C, Van Noorden CJF, Haveman J, Vandertop WP, van Furth WR. Experimental iodine-125 seed irradiation of intracerebral brain tumors in nude mice. Radiat Oncol 2007; 2:38. [PMID: 17897452 PMCID: PMC2174502 DOI: 10.1186/1748-717x-2-38] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Accepted: 09/26/2007] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND High-dose radiotherapy is standard treatment for patients with brain cancer. However, in preclinical research external beam radiotherapy is limited to heterotopic murine models- high-dose radiotherapy to the murine head is fatal due to radiation toxicity. Therefore, we developed a stereotactic brachytherapy mouse model for high-dose focal irradiation of experimental intracerebral (orthotopic) brain tumors. METHODS Twenty-one nude mice received a hollow guide-screw implanted in the skull. After three weeks, 5 x 105 U251-NG2 human glioblastoma cells were injected. Five days later, a 2 mCi iodine-125 brachytherapy seed was inserted through the guide-screw in 11 randomly selected mice; 10 mice received a sham seed. Mice were euthanized when severe neurological or physical symptoms occurred. The cumulative irradiation dose 5 mm below the active iodine-125 seeds was 23.0 Gy after 13 weeks (BEDtumor = 30.6 Gy). RESULTS In the sham group, 9/10 animals (90%) showed signs of lethal tumor progression within 6 weeks. In the experimental group, 2/11 mice (18%) died of tumor progression within 13 weeks. Acute side effects in terms of weight loss or neurological symptoms were not observed in the irradiated animals. CONCLUSION The intracerebral implantation of an iodine-125 brachytherapy seed through a stereotactic guide-screw in the skull of mice with implanted brain tumors resulted in a significantly prolonged survival, caused by high-dose irradiation of the brain tumor that is biologically comparable to high-dose fractionated radiotherapy- without fatal irradiation toxicity. This is an excellent mouse model for testing orthotopic brain tumor therapies in combination with radiation therapy.
Collapse
Affiliation(s)
- Joost JC Verhoeff
- Department of Radiation Oncology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Lukas JA Stalpers
- Department of Radiation Oncology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Annet W Coumou
- Department of Neurosurgery, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Kees Koedooder
- Department of Radiation Oncology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Cristina Lavini
- Department of Radiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Cornelis JF Van Noorden
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jaap Haveman
- Department of Radiation Oncology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - William P Vandertop
- Department of Neurosurgery, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Wouter R van Furth
- Department of Neurosurgery, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
48
|
Lim SD, Stallcup W, Lefkove B, Govindarajan B, Au KS, Northrup H, Lang D, Fisher DE, Patel A, Amin MB, Arbiser JL. Expression of the neural stem cell markers NG2 and L1 in human angiomyolipoma: are angiomyolipomas neoplasms of stem cells? Mol Med 2007. [PMID: 17592550 DOI: 10.2119/2006-00070.lim] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Angiomyolipomas are benign tumors of the kidney which express phenotypes of smooth muscle, fat, and melanocytes. These tumors appear with increased frequency in the autosomal dominant disorder tuberous sclerosis and are the leading cause of morbidity in adults with tuberous sclerosis. While benign, these tumors are capable of provoking life threatening hemorrhage and replacement of the kidney parenchyma, resulting in renal failure. The histogenesis of these tumors is currently unclear, although currently, we believe these tumors arise from "perivascular epithelioid cells" of which no normal counterpart has been convincingly demonstrated. Recently, stem cell precursors have been recognized that can give rise to smooth muscle and melanocytes. These precursors have been shown to express the neural stem cell marker NG2 and L1. In order to determine whether angiomyolipomas, which exhibit smooth muscle and melanocytic phenotypes, express NG2 and L1, we performed immunocytochemistry on a cell line derived from a human angiomyolipoma, and found that these cells are uniformly positive. Immunohistochemistry of human angiomyolipoma specimens revealed uniform staining of tumor cells, while renal cell carcinomas revealed positivity only of angiogenic vessels. These results support a novel histogenesis of angiomyolipoma as a defect in differentiation of stem cell precursors.
Collapse
Affiliation(s)
- So Dug Lim
- Department of Urology, Emory University School of Medicine, and Atlanta VA Medical Center, Georgia 30322, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Erfurt C, Sun Z, Haendle I, Schuler-Thurner B, Heirman C, Thielemans K, van der Bruggen P, Schuler G, Schultz ES. Tumor-reactive CD4+ T cell responses to the melanoma-associated chondroitin sulphate proteoglycan in melanoma patients and healthy individuals in the absence of autoimmunity. THE JOURNAL OF IMMUNOLOGY 2007; 178:7703-9. [PMID: 17548607 DOI: 10.4049/jimmunol.178.12.7703] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To avoid immune escape by down-regulation or loss of Ag by the tumor cells, target Ags are needed, which are important for the malignant phenotype and survival of the tumor. We could identify a CD4(+) T cell epitope derived from the human melanoma-associated chondroitin sulfate proteoglycan (MCSP) (also known as high m.w.-melanoma-associated Ag), which is strongly expressed on >90% of human melanoma lesions and is important for the motility and invasion of melanoma cells. However, MCSP is not strictly tumor specific, because it is also expressed in a variety of normal tissues. Therefore, self tolerance should prevent the induction of strong T cell responses against these Ags by vaccination strategies. In contrast, breaking self tolerance to this Ag by effectively manipulating the immune system might mediate antitumor responses, although it would bear the risk of autoimmunity. Surprisingly, we could readily isolate CD4(+) Th cells from the blood of a healthy donor-recognizing peptide MCSP(693-709) on HLA-DR11-expressing melanoma cells. Broad T cell reactivity against this Ag could be detected in the peripheral blood of both healthy donors and melanoma patients, without any apparent signs of autoimmune disease. In some patients, a decline of T cell reactivity was observed upon tumor progression. Our data indicate that CD4(+) T cells are capable of recognizing a membrane glycoprotein that is important in melanoma cell function, and it may be possible that the sizable reactivity to this Ag in most normal individuals contributes to immune surveillance against cancer.
Collapse
Affiliation(s)
- Cornelia Erfurt
- Department of Dermatology, University Hospital of Erlangen, Hartmannstrasse 14, Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lim SD, Stallcup W, Lefkove B, Govindarajan B, Au KS, Northrup H, Lang D, Fisher DE, Patel A, Amin MB, Arbiser JL. Expression of the neural stem cell markers NG2 and L1 in human angiomyolipoma: are angiomyolipomas neoplasms of stem cells? MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2007; 13:160-5. [PMID: 17592550 PMCID: PMC1892760 DOI: 10.2119/2006–00070.lim] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 02/22/2007] [Indexed: 11/06/2022]
Abstract
Angiomyolipomas are benign tumors of the kidney which express phenotypes of smooth muscle, fat, and melanocytes. These tumors appear with increased frequency in the autosomal dominant disorder tuberous sclerosis and are the leading cause of morbidity in adults with tuberous sclerosis. While benign, these tumors are capable of provoking life threatening hemorrhage and replacement of the kidney parenchyma, resulting in renal failure. The histogenesis of these tumors is currently unclear, although currently, we believe these tumors arise from "perivascular epithelioid cells" of which no normal counterpart has been convincingly demonstrated. Recently, stem cell precursors have been recognized that can give rise to smooth muscle and melanocytes. These precursors have been shown to express the neural stem cell marker NG2 and L1. In order to determine whether angiomyolipomas, which exhibit smooth muscle and melanocytic phenotypes, express NG2 and L1, we performed immunocytochemistry on a cell line derived from a human angiomyolipoma, and found that these cells are uniformly positive. Immunohistochemistry of human angiomyolipoma specimens revealed uniform staining of tumor cells, while renal cell carcinomas revealed positivity only of angiogenic vessels. These results support a novel histogenesis of angiomyolipoma as a defect in differentiation of stem cell precursors.
Collapse
Affiliation(s)
- So Dug Lim
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William Stallcup
- Atlanta VA Medical Center, The Burnham Institute for Medical Research, Cancer Research Center, La Jolla, California, USA
| | - Benjamin Lefkove
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Baskaran Govindarajan
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kit Sing Au
- Division of Medical Genetics, Department of Pediatrics, University of Texas Medical School at Houston, Houston, Texas, USA; Cardiovascular Division, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Hope Northrup
- Division of Medical Genetics, Department of Pediatrics, University of Texas Medical School at Houston, Houston, Texas, USA; Cardiovascular Division, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Deborah Lang
- Division of Medical Genetics, Department of Pediatrics, University of Texas Medical School at Houston, Houston, Texas, USA; Cardiovascular Division, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - David E Fisher
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Avani Patel
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mahul B Amin
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
- Address correspondence and reprint requests to Jack L Arbiser, Department of Dermatology, Emory University School of Medicine, 1639 Pierce Drive, WMB 5309, Atlanta, GA 30322. Phone: 404-727-5063; Fax: 404-727-5878; E-mail:
| |
Collapse
|