1
|
Huang TL, Mei YW, Li Y, Chen X, Yu SX, Kuang YQ, Shu HF. Thrombospondin-2 promotes the proliferation and migration of glioma cells and contributes to the progression of glioma. Chin Neurosurg J 2022; 8:39. [PMID: 36476392 PMCID: PMC9728004 DOI: 10.1186/s41016-022-00308-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gliomas, especially high-grade gliomas, are highly malignant with a poor prognosis. Although existing treatments have improved the survival rate of patients with glioma, the recurrence and mortality rates are still not ideal. The molecular mechanisms involved in the occurrence and development of glioma are still poorly understood. We previously reported that thrombospondin-2 (TSP2) expression was increased in tumor specimens from rat models, promoting excitatory synapse formation. However, little is known about the effect of TSP2 on the biological characteristics of glioma. METHODS Glioma and cerebral cortex tissues were collected from 33 patients, and the expression of TSP2 in them was analyzed. Next, the proliferation and migration of TSP2 on glioma cells were analyzed in vitro. At last, a glioma transplantation model was constructed to explore the growth of TSP2 on glioma in vivo. RESULTS The expression of TSP2 in surgical glioma specimens was increased compared to that in the normal cortex. Interestingly, the TSP2 protein level was higher in high-grade glioma (HGG, World Health Organization (WHO) grades 3-4) than in low-grade glioma (LGG, WHO grades 1-2) tissues. Exogenous addition of the TSP2 protein at an appropriate concentration promoted the migration of glioma cells but did not significantly affect their proliferation. Surprisingly, overexpression of TSP2 promoted both the migration and proliferation of cultured glioma cells. Moreover, in vivo experimental data implied that overexpression of TSP2 in C6 cells promoted the malignant growth of gliomas, while knockout of TSP2 slowed glioma growth. CONCLUSIONS TSP2 promotes the migration and proliferation of glioma cells, which may provide new ideas for blocking glioma progression.
Collapse
Affiliation(s)
- Tian-Lan Huang
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, No.270 Rongdu Road, Jinniu District, 610083 Chengdu, China ,grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| | - Yi-Wen Mei
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, No.270 Rongdu Road, Jinniu District, 610083 Chengdu, China ,grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| | - Yang Li
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, No.270 Rongdu Road, Jinniu District, 610083 Chengdu, China ,grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| | - Xin Chen
- grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| | - Si-Xun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, No.270 Rongdu Road, Jinniu District, 610083 Chengdu, China ,grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| | - Yong-Qin Kuang
- grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| | - Hai-Feng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, No.270 Rongdu Road, Jinniu District, 610083 Chengdu, China ,grid.263901.f0000 0004 1791 7667College of Medicine, Southwest Jiaotong University, No. 111, North Section 1, Second Ring Road, 610031 Chengdu, China
| |
Collapse
|
2
|
Wang M, Wang J, Liu J, Zhu L, Ma H, Zou J, Wu W, Wang K. Systematic prediction of key genes for ovarian cancer by co-expression network analysis. J Cell Mol Med 2020; 24:6298-6307. [PMID: 32319226 PMCID: PMC7294139 DOI: 10.1111/jcmm.15271] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/18/2020] [Accepted: 03/28/2020] [Indexed: 01/18/2023] Open
Abstract
Ovarian cancer (OC) is the most lethal gynaecological malignancy, characterized by high recurrence and mortality. However, the mechanisms of its pathogenesis remain largely unknown, hindering the investigation of the functional roles. This study sought to identify key hub genes that may serve as biomarkers correlated with prognosis. Here, we conduct an integrated analysis using the weighted gene co‐expression network analysis (WGCNA) to explore the clinically significant gene sets and identify candidate hub genes associated with OC clinical phenotypes. The gene expression profiles were obtained from the MERAV database. Validations of candidate hub genes were performed with RNASeqV2 data and the corresponding clinical information available from The Cancer Genome Atlas (TCGA) database. In addition, we examined the candidate genes in ovarian cancer cells. Totally, 19 modules were identified and 26 hub genes were extracted from the most significant module (R2 = .53) in clinical stages. Through the validation of TCGA data, we found that five hub genes (COL1A1, DCN, LUM, POSTN and THBS2) predicted poor prognosis. Receiver operating characteristic (ROC) curves demonstrated that these five genes exhibited diagnostic efficiency for early‐stage and advanced‐stage cancer. The protein expression of these five genes in tumour tissues was significantly higher than that in normal tissues. Besides, the expression of COL1A1 was associated with the TAX resistance of tumours and could be affected by the autophagy level in OC cell line. In conclusion, our findings identified five genes could serve as biomarkers related to the prognosis of OC and may be helpful for revealing pathogenic mechanism and developing further research.
Collapse
Affiliation(s)
- Mingyuan Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Sepsis, Translational Medicine of Hunan, Central South University, Changsha, China.,Department of gynecology, Zhuzhou Central Hospital, Central South University, Zhuzhou, China
| | - Jinjin Wang
- Department of gynecology, Zhuzhou Central Hospital, Central South University, Zhuzhou, China
| | - Jinglan Liu
- Department of gynecology, Zhuzhou Central Hospital, Central South University, Zhuzhou, China
| | - Lili Zhu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Sepsis, Translational Medicine of Hunan, Central South University, Changsha, China
| | - Heng Ma
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Sepsis, Translational Medicine of Hunan, Central South University, Changsha, China
| | - Jiang Zou
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Sepsis, Translational Medicine of Hunan, Central South University, Changsha, China
| | - Wei Wu
- Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Sepsis, Translational Medicine of Hunan, Central South University, Changsha, China.,Department of Laboratory Animals, Hunan Key Laboratory of Animal Models for Human Diseases, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
3
|
Blaylock RL. Accelerated cancer aggressiveness by viral oncomodulation: New targets and newer natural treatments for cancer control and treatment. Surg Neurol Int 2019; 10:199. [PMID: 31768279 PMCID: PMC6826277 DOI: 10.25259/sni_361_2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022] Open
Abstract
An infectious etiology for a number of cancers has been entertained for over 100 years and modern studies have confirmed that a number of viruses are linked to cancer induction. While a large number of viruses have been demonstrated in a number of types of cancers, most such findings have been dismissed in the past as opportunistic infections, especially with persistent viruses with high rates of infectivity of the world’s populations. More recent studies have clearly shown that while not definitely causing these cancers, these viruses appear capable of affecting the biology of these tumors in such a way as to make them more aggressive and more resistant to conventional treatments. The term oncomodulatory viruses has been used to describe this phenomenon. A number of recent studies have shown a growing number of ways these oncomodulatory viruses can alter the pathology of these tumors by affecting cell-signaling, cell metabolism, apoptosis mechanisms, cell-cell communication, inflammation, antitumor immunity suppression, and angiogenesis. We are also learning that much of the behavior of tumors depends on cancer stem cells and stromal cells within the tumor microenvironment, which participate in extensive, dynamic crosstalk known to affect tumor behavior. Cancer stem cells have been found to be particularly susceptible to infection by human cytomegalovirus. In a number of studies, it has been shown that while only a select number of cells are actually infected with the virus, numerous viral proteins are released into cancer and stromal cells in the microenvironment and these viral proteins are known to affect tumor behavior and aggressiveness.
Collapse
|
4
|
Serum thrombospondin-2 is a candidate diagnosis biomarker for early non-small-cell lung cancer. Biosci Rep 2019; 39:BSR20190476. [PMID: 31296790 PMCID: PMC6658726 DOI: 10.1042/bsr20190476] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/04/2019] [Accepted: 06/28/2019] [Indexed: 12/18/2022] Open
Abstract
Thrombospondin-2 (THBS2) is a secreted protein overexpressed in numerous cancers and may function as a diagnostic tumor marker. The objective of the present study was to investigate the diagnostic performance of serum THBS2 in early stage non-small-cell lung cancer (NSCLC). Serum THBS2 and Cyfra21-1 level were evaluated in blood samples of 112 patients from NSCLC groups and 51 healthy control (HC) groups. Receiver operator characteristic (ROC) curves were used to evaluate the diagnostic significance. Serum THBS2 level was significantly up-regulated in NSCLC patients compared with healthy control subjects (P<0.0001), and the postoperative THBS2 level decreased significantly (P<0.0001). ROC curves analysis demonstrated that THBS2 was a comparable biomarker as Cyfra21-1 to distinguish early stage NSCLC or lung squamous cell carcinoma (SC) from healthy control subjects. And Cyfra21-1 was observed with significantly improved performances by the combination of THBS2 to distinguish early stage NSCLC (P<0.05) as well as SC (P<0.05) from the control subjects. In addition, THBS2 was estimated to perform well in the diagnosis of patients with Cyfra21-1-negative NSCLC (area under the curve [AUC] = 0.73). In summary, the present study suggested that serum THBS2 might be an early diagnostic biomarker for NSCLC.
Collapse
|
5
|
Alshabi AM, Vastrad B, Shaikh IA, Vastrad C. Identification of Crucial Candidate Genes and Pathways in Glioblastoma Multiform by Bioinformatics Analysis. Biomolecules 2019; 9:biom9050201. [PMID: 31137733 PMCID: PMC6571969 DOI: 10.3390/biom9050201] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to investigate the molecular mechanisms underlying glioblastoma multiform (GBM) and its biomarkers. The differentially expressed genes (DEGs) were diagnosed using the limma software package. The ToppGene (ToppFun) was used to perform pathway and Gene Ontology (GO) enrichment analysis of the DEGs. Protein-protein interaction (PPI) networks, extracted modules, miRNA-target genes regulatory network and TF-target genes regulatory network were used to obtain insight into the actions of DEGs. Survival analysis for DEGs was carried out. A total of 590 DEGs, including 243 up regulated and 347 down regulated genes, were diagnosed between scrambled shRNA expression and Lin7A knock down. The up-regulated genes were enriched in ribosome, mitochondrial translation termination, translation, and peptide biosynthetic process. The down-regulated genes were enriched in focal adhesion, VEGFR3 signaling in lymphatic endothelium, extracellular matrix organization, and extracellular matrix. The current study screened the genes in the PPI network, extracted modules, miRNA-target genes regulatory network, and TF-target genes regulatory network with higher degrees as hub genes, which included NPM1, CUL4A, YIPF1, SHC1, AKT1, VLDLR, RPL14, P3H2, DTNA, FAM126B, RPL34, and MYL5. Survival analysis indicated that the high expression of RPL36A and MRPL35 were predicting longer survival of GBM, while high expression of AP1S1 and AKAP12 were predicting shorter survival of GBM. High expression of RPL36A and AP1S1 were associated with pathogenesis of GBM, while low expression of ALPL was associated with pathogenesis of GBM. In conclusion, the current study diagnosed DEGs between scrambled shRNA expression and Lin7A knock down samples, which could improve our understanding of the molecular mechanisms in the progression of GBM, and these crucial as well as new diagnostic markers might be used as therapeutic targets for GBM.
Collapse
Affiliation(s)
- Ali Mohamed Alshabi
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran 61441, Saudi Arabia.
| | - Basavaraj Vastrad
- Department of Pharmaceutics, SET`S College of Pharmacy, Dharwad, Karnataka 580002, India.
| | - Ibrahim Ahmed Shaikh
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 61441, Saudi Arabia.
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India.
| |
Collapse
|
6
|
Clemessy M, Janzer RC, Lhermitte B, Gasc JM, Juillerat-Jeanneret L. Expression of dual angiogenic/neurogenic growth factors in human primary brain tumors. J Neurooncol 2011; 107:29-36. [PMID: 21979892 DOI: 10.1007/s11060-011-0715-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 09/16/2011] [Indexed: 12/20/2022]
Abstract
Brain tumors, benign or malignant, are characterized by a very high degree of vascularization. Recent accumulating evidence suggests that during development the neuronal wiring follows the same routes as the vasculature and that these two systems may share some of the same factors for guidance. Thus, expression of dual angiogenic/neurogenic growth factors was evaluated by in situ hybridization in human primary brain tumors of three different types, i.e., astrocytomas, oligodendrogliomas, and ependymomas, of increasing grades, in relation with the grade and type of the tumor. For this evaluation we selected vascular endothelial growth factor (VEGF-A) and its receptors VEGF-R1 and VEGF-R2 and the neuropilins 1 and 2 (NRP-1 and NRP-2), which have proangiogenic properties, platelet-derived growth factor (PDGF) receptor-beta (PDGF-Rβ), which is required for the functional maturation of blood vessels, the ephrins and their Eph receptors, angiotensinogen (AGT) and thrombospondin-2 (TSP-2), which have potential antiangiogenic properties, and netrin-1 (Net-1), which regulates vascular architecture. We show that the expression of the VEGF-NRP system, PDGF-Rβ, TSP-2, AGT, and Net-1 are differentially regulated, either increased or decreased, in relation with the type and grade of the tumor, whereas regulation of the ephrinB system does not seem to be relevant in these human brain tumors.
Collapse
Affiliation(s)
- Maud Clemessy
- INSERM U833, Collège-de-France, 11 place Marcelin Berthelot, Paris, France
| | | | | | | | | |
Collapse
|
7
|
Meng H, Zhang X, Hankenson KD, Wang MM. Thrombospondin 2 potentiates notch3/jagged1 signaling. J Biol Chem 2009; 284:7866-74. [PMID: 19147503 DOI: 10.1074/jbc.m803650200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Extracellular thrombospondins (TSP or THBS) and the Notch family of transmembrane receptors share a role in multiple, overlapping cellular functions and participate in developmental signaling and pathological reactions to tissue injury. We demonstrate that TSP2, but not TSP1, enhances the potency of Notch3 signal transduction. In addition, TSP2 reduces cancer cell proliferation in a Notch-ligand dependent fashion. The loss of TSP2 in knock-out mice reduces Notch target gene expression. TSP2 binds directly to Notch3 and Jagged1. TSP1 also binds to Notch3 and Jagged1; however, only TSP2 augments the interaction between Notch3 and Jagged1. These studies demonstrate that the diverse functions of TSP2 may also include a role as an intermediary protein that facilitates transcellular receptor-ligand interactions.
Collapse
Affiliation(s)
- He Meng
- Department of Neurology and Molecular, University of Michigan, Ann Arbor, Michigan 48109-5622, USA
| | | | | | | |
Collapse
|
8
|
Abstract
Angiogenesis, the sprouting of new blood vessels from preexisting blood vessels, is a hallmark of glioma progression. Malignant gliomas are among the most lethal tumors with a very dismal prognosis, despite advances in standard therapy, including surgery, radiation, and chemotherapy. The median survival of patients with malignant gliomas has changed little in the last few years and is still measured in months. In an attempt to develop new therapeutic strategies and identify the molecular mechanism involved in glioma growth and progression, there has been extraordinary scientific interest in the past 2 decades in angiogenic responses associated with gliomas. This chapter focuses on the molecular mechanism of glioma angiogenesis and summarizes some of the therapeutic approaches based on antiangiogenesis.
Collapse
Affiliation(s)
- Marcia Machein
- Department of Neurosurgery, University of Freiburg Medical School, Breisacher Str. 64, Freiburg 79106, Germany.
| | | |
Collapse
|
9
|
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
Affiliation(s)
- Joshua C Anderson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | |
Collapse
|
10
|
Fischer I, Gagner J, Law M, Newcomb EW, Zagzag D. Angiogenesis in gliomas: biology and molecular pathophysiology. Brain Pathol 2006; 15:297-310. [PMID: 16389942 PMCID: PMC8096031 DOI: 10.1111/j.1750-3639.2005.tb00115.x] [Citation(s) in RCA: 260] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is characterized by exuberant angiogenesis, a key event in tumor growth and progression. The pathologic mechanisms driving this change and the biological behavior of gliomas remain unclear. One mechanism may involve cooption of native blood vessels by glioma cells inducing expression of angiopoietin-2 by endothelial cells. Subsequently, vascular apoptosis and involution leads to necrosis and hypoxia. This in turn induces angiogenesis that is associated with expression of hypoxia-inducible factor (HIF)-1alpha and vascular endothelial growth factor (VEGF) in perinecrotic pseudopalisading glioma cells. Here we review the molecular and cellular mechanisms implicated in HIF-1-dependent and HIF-1-independent glioma-associated angiogenesis. In GBMs, both tumor hypoxia and genetic alterations commonly occur and act together to induce the expression of HIF-1. The angiogenic response of the tumor to HIF-1 is mediated by HIF-1-regulated target genes leading to the upregulation of several proangiogenic factors such as VEGF and other adaptive response molecules. Understanding the roles of these regulatory processes in tumor neovascularization, tumor growth and progression, and resistance to therapy will ultimately lead to the development of improved antiangiogenic therapies for GBMs.
Collapse
Affiliation(s)
- Ingeborg Fischer
- Microvascular and Molecular Neuro‐oncology Laboratory, New York University School of Medicine
- Department of Pathology, New York University School of Medicine
- Division of Neuropathology, New York University School of Medicine
| | - Jean‐Pierre Gagner
- Microvascular and Molecular Neuro‐oncology Laboratory, New York University School of Medicine
- Department of Pathology, New York University School of Medicine
- Division of Neuropathology, New York University School of Medicine
| | - Meng Law
- Department of Radiology, New York University School of Medicine
- Department of Neurosurgery, New York University School of Medicine
- New York University Cancer Institute, New York University School of Medicine
| | - Elizabeth W. Newcomb
- Department of Pathology, New York University School of Medicine
- New York University Cancer Institute, New York University School of Medicine
| | - David Zagzag
- Microvascular and Molecular Neuro‐oncology Laboratory, New York University School of Medicine
- Department of Pathology, New York University School of Medicine
- Division of Neuropathology, New York University School of Medicine
- Department of Neurosurgery, New York University School of Medicine
- New York University Cancer Institute, New York University School of Medicine
| |
Collapse
|
11
|
Wang D, Anderson JC, Gladson CL. The role of the extracellular matrix in angiogenesis in malignant glioma tumors. Brain Pathol 2005; 15:318-26. [PMID: 16389944 PMCID: PMC8095805 DOI: 10.1111/j.1750-3639.2005.tb00117.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Angiogenesis is a promising target for the development of effective strategies for the treatment of malignant brain tumors in that it has the potential to starve large tumors and prevent the regrowth of residual margins. Two critical steps in angiogenesis, the proliferation of activated endothelial cells and their migration into the perivascular space (sprouting), require adherence of the endothelial cells to the extracellular matrix (ECM). Thus, the availability of the appropriate ligands within the ECM contributes to the regulation of angiogenesis. In addition, several components of the ECM can act through other mechanisms to further promote angiogenesis or inhibit it. Current evidence suggests that the regulation of angiogenesis is a dynamic process in which the endothelial cells can promote angiogenesis by secreting proteases that remodel the ECM, tumor cells can further promote angiogenesis by secreting ECM components and actively remodeling their environment, and stromal cells may respond to angiogenesis associated with tumors and inflammatory reactions by secreting inhibitory molecules. Here, we provide a critical review of the protein and proteoglycan components of the ECM that have been implicated in angiogenesis with an emphasis on their role in promoting or inhibiting angiogenesis in brain tumors.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| | - Joshua C. Anderson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| | - Candece L. Gladson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| |
Collapse
|
12
|
Rege TA, Fears CY, Gladson CL. Endogenous inhibitors of angiogenesis in malignant gliomas: nature's antiangiogenic therapy. Neuro Oncol 2005; 7:106-21. [PMID: 15831230 PMCID: PMC1871889 DOI: 10.1215/s115285170400119x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is necessary for tumor growth beyond a volume of approximately 2 mm(3). This observation, along with the accessibility of tumor vessels to therapeutic targeting, has resulted in a research focus on inhibitors of angiogenesis. A number of endogenous inhibitors of angiogenesis are found in the body. Some of these are synthesized by specific cells in different organs, and others are created by extracellular proteolytic cleavage of plasma-derived or extracellular matrix-localized proteins. In this review, we focus on angiostatin, endostatin, PEX, pigment epithelial-derived factor, and thrombospondin (TSP)-1 and -2, either because these molecules are expressed in malignant glioma biopsies or because animal studies in malignant glioma models have suggested that their therapeutic administration could be efficacious. We review the known mechanisms of action, potential receptors, expression in glioma biopsy samples, and studies testing their potential therapeutic efficacy in animal models of malignant glioma. Two conclusions can be made regarding the mechanisms of action of these inhibitors: (1) Several of these inhibitors appear to mediate their antiangiogenic effect through multiple protein-protein interactions that inhibit the function of proangiogenic molecules rather than through a specific receptor-mediated signaling event, and (2) TSP-1 and TSP-2 appear to mediate their antiangiogenic effect, at least in part, through a specific receptor, CD36, which initiates the antiangiogenic signal. Although not proven in gliomas, evidence suggests that expression of specific endogenous inhibitors of angiogenesis in certain organs may be part of a host antitumor response. The studies reviewed here suggest that new antiangiogenic therapies for malignant gliomas offer exciting promise as nontoxic, growth-inhibitory agents.
Collapse
Affiliation(s)
| | | | - Candece L. Gladson
- Address correspondence to Candece L. Gladson, The University of Alabama at Birmingham, LHRB 567, 701 South 19th Street, Birmingham, AL 35294, USA (
)
| |
Collapse
|
13
|
Abstract
Advances in our comprehension of cancer biology and metastasis formation have led to the development of new therapeutic approaches that target tumor biology. The survival and establishment of metastatic lesions depend on a shift in the normal balance of key regulatory factors in favor of angiogenesis. Although a number of these factors have been identified, the most potent regulator of angiogenesis is vascular endothelial growth factor (VEGF). In phase I trials, targeting VEGF with single-agent therapy did not produce clinical benefit for patients, despite promise in preclinical trials. However, the recent data showing that anti-VEGF therapy can enhance the effects of chemotherapy demonstrate the utility in targeting angiogenic factors as a component of antineoplastic regimens. A better understanding of the functions of VEGF allows the development of new hypotheses with regard to its mechanism of action. This article will highlight what is known about colorectal cancer angiogenesis, and will discuss how therapy targeting VEGF may enhance the effects of chemotherapy (and radiation therapy).
Collapse
Affiliation(s)
- Lee M Ellis
- Departments of Surgical Oncology and Cancer Biology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Box 444, Houston, TX 77030, USA
| |
Collapse
|
14
|
Horiguchi H, Jin L, Ruebel KH, Scheithauer BW, Lloyd RV. Regulation of VEGF-A, VEGFR-I, thrombospondin-1, -2, and -3 expression in a human pituitary cell line (HP75) by TGFbeta1, bFGF, and EGF. Endocrine 2004; 24:141-6. [PMID: 15347840 DOI: 10.1385/endo:24:2:141] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Revised: 06/21/2004] [Accepted: 06/21/2004] [Indexed: 02/07/2023]
Abstract
Pituitary tumors are highly vascular neoplasms, which suggest an important role of angiogenesis in pituitary tumor growth. We used the human pituitary cell line (HP75) to examine the effects of the growth factors TGFbeta1, bFGF, and EGF on cell growth, and on the regulation of the pro-angiogenic growth factor VEGF-A and the VEGFR-I and the anti-angiogenic molecules thrombospondin (TSP) TSP-1 and TSP-2 along with TSP-3. Real-time RT-PCR was used to measure mRNA levels, and Western blot was used to analyze TSP-1 and TSP-2 protein levels. TGFbeta1 treatment (1 x 10(-9) M) increased VEGF-A mRNA levels significantly (p < 0.05) after 4 and 24 h of treatment. TGF beta1 treatment decreased VEGF-R mRNA levels after 96 h of treatment (p < 0.05). After 96 h of treatment, TSP-1 and TSP-2 mRNA levels were significantly increased (p < 0.05) by TGFbeta1 treatment, which also inhibited HP75 cell growth. Basic FGF also increased TSP-1 mRNA levels after 96 h of treatment, but did not regulate growth of the pituitary tumor cells. Basic FGF and EGF did not modulate changes in VEGF-A mRNA levels after 4 and 24 h of treatment, but EGF increased VEGF-A significantly (p < 0.05) after 96 h of treatment. These results indicate that TGFbeta1 treatment may regulate angiogenesis in pituitary cells by initially increasing levels of pro-angiogenic VEGF-A and then stimulating the anti-angiogenic molecules TSP-1 and TSP-2 levels.
Collapse
Affiliation(s)
- Hidehisa Horiguchi
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, 55901, USA
| | | | | | | | | |
Collapse
|
15
|
Cinatl J, Vogel JU, Kotchetkov R, Wilhelm Doerr H. Oncomodulatory signals by regulatory proteins encoded by human cytomegalovirus: a novel role for viral infection in tumor progression. FEMS Microbiol Rev 2004; 28:59-77. [PMID: 14975530 DOI: 10.1016/j.femsre.2003.07.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2003] [Revised: 07/29/2003] [Accepted: 07/31/2003] [Indexed: 01/06/2023] Open
Abstract
A high frequency of human cytomegalovirus (HCMV) genome and antigens in tumor samples of patients with different malignancies is now well documented, although the causative role for HCMV in the development of the neoplasias remains to be established. HCMV infection can modulate multiple cellular regulatory and signalling pathways in a manner similar to that of oncoproteins of small DNA tumor viruses such as human papilloma virus or adenoviruses. However, in contrast to these DNA tumor viruses, HCMV infection fails to transform susceptible normal human cells. There is now growing evidence that tumor cells with disrupted regulatory and signalling pathways enable HCMV to modulate their properties including stimulation of cell proliferation, survival, invasion, production of angiogenic factors, and immunogenic properties. In contrast to previously suggested "hit and run" transformation we suggest that persistence in tumor cells is essential for HCMV to fully express its oncomodulatory effects. These effects are observed particularly in persistent HCMV infection and are mediated mainly by activity of HCMV regulatory proteins. In persistently HCMV-infected tumor cell lines - a selection of novel, slowly growing virus variants with changes in coding sequences for virus regulatory proteins takes place. As a result, oncomodulatory effects of HCMV infection may lead to a shift to more malignant phenotype of tumor cells contributing to tumor progression.
Collapse
Affiliation(s)
- Jindrich Cinatl
- Zentrum der Hygiene, Institut für Medizinische Virologie, Klinikum der Johann Wolfgang Goethe-Universität, Paul-Ehrlich-Str. 40, 60596 Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
16
|
Radovanovic I, D'Angelo MG, Aguzzi A. Angiogenesis in transgenic models of multistep angiogenesis. Cancer Treat Res 2004; 117:97-114. [PMID: 15015554 DOI: 10.1007/978-1-4419-8871-3_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The histopathology and the epidemiology of human cancers, as well as studies of animal models of tumorigenesis, have led to a widely accepted notion that multiple genetic and epigenetic changes have to accumulate for progression to malignancy. Formation of new blood vessels (tumor angiogenesis) has been recognized, in addition to proliferative capabilities and ability to down-modulate cell death (apoptosis), as essential for the progressive growth and expansion of solid tumors. Mice overexpressing activated forms of oncogenes or carrying targeted mutations in tumor suppressor genes have proven extremely useful for linking the function of these genes with specific tumor features such as continuous proliferation, escape from apoptosis, invasion and neo-angiogenesis. The interbreeding of these mice allows for studying the extent of cooperativity between different genetic lesions in disease progression, leading to a greater understanding of multi-stage nature of tumorigenesis.
Collapse
Affiliation(s)
- Ivan Radovanovic
- Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland
| | | | | |
Collapse
|
17
|
Abstract
Quantitative determination of the degree of vascularity has been shown to be independently prognostically significant in many human tumor types. In particular, tumor vascularity has known importance in astrocytomas, in which endothelial proliferation is a criterion for anaplasia in many grading schemes. This chapter summarizes the known associations of quantitated microvessel parameters obtained from histologic sections of human brain tumors with clinical outcome, or other pathobiologic factors that have been examined. Among the conclusions are 1) brain tumors have the unique feature of complex "glomeruloid" vessels, as well as heterogeneity of microvascular distribution and caliber; 2) lower-grade astrocytomas incorporate pre-existing vessels, while glioblastomas develop new vessels; 3) quantitation may have additional independent prognostic value over and above routine histologic grade in low-grade astrocytomas with low tumor cell proliferative indices. These findings have implications for the appropriateness of antiangiogenic therapies
Collapse
Affiliation(s)
- Rebecca D Folkerth
- Departments of Pathology (Neuropathology), Brigham and Women 's Hospital and Children 's Hospital, and Harvard Medical School, Boston MA 02115, USA
| |
Collapse
|
18
|
Kishi M, Nakamura M, Nishimine M, Ishida E, Shimada K, Kirita T, Konishi N. Loss of heterozygosity on chromosome 6q correlates with decreased thrombospondin-2 expression in human salivary gland carcinomas. Cancer Sci 2003; 94:530-5. [PMID: 12824879 PMCID: PMC11160221 DOI: 10.1111/j.1349-7006.2003.tb01478.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2002] [Revised: 04/03/2003] [Accepted: 04/10/2003] [Indexed: 11/28/2022] Open
Abstract
Since loss of heterozygosity (LOH) on the long arm of chromosome 6q is frequently observed in salivary gland carcinomas, we examined 28 salivary gland carcinomas using 24 microsat- ellite markers mapping to 6q15-27 to identify the commonly deleted region that we felt might contain one or more tumor suppressor genes. LOH was detected in at least one locus in 10 of 28 tumors (35.7%). The most frequently deleted regions occurred between D6S1581 and D6S305 (LOH cluster region 1 (LCR1) and between D6S297 and D6S1590 (LCR2). LOH was observed in 60% of adenoid cystic carcinomas (ACC) and in 57.1% of mucoepidermoid carcinomas (MEC), but was not observed in any locus in any other histological subtypes studied. The gene encoding for thrombospondin-2 (TSP-2) is located in LCR2 and 8 of 9 tumors demonstrating LOH in this region also showed significantly decreased TSP-2 expression by immunohistochemistry. As TSP-2 is a potent inhibitor of tumor growth and angiogenesis, we examined whether TSP-2 expression correlated to microvascular angiogenesis in these tumors and discovered that microvessel counts were significantly higher in lesions with decreased TSP-2 expression (P = 0.02). Our results suggest that 6q LOH may be a significant event in salivary gland carcinogenesis, particularly in ACC and MEC, and that the correlated decrease of TSP-2 expression also plays a critical role.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Adenoid Cystic/chemistry
- Carcinoma, Adenoid Cystic/genetics
- Carcinoma, Adenoid Cystic/pathology
- Carcinoma, Mucoepidermoid/chemistry
- Carcinoma, Mucoepidermoid/genetics
- Carcinoma, Mucoepidermoid/pathology
- Carcinoma, Squamous Cell/chemistry
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Chromosomes, Human, Pair 6
- DNA, Neoplasm/genetics
- Female
- Humans
- Loss of Heterozygosity
- Male
- Microsatellite Repeats
- Middle Aged
- Mutation
- Polymerase Chain Reaction
- Salivary Gland Neoplasms/chemistry
- Salivary Gland Neoplasms/genetics
- Salivary Gland Neoplasms/pathology
- Thrombospondins/genetics
Collapse
Affiliation(s)
- Munehiro Kishi
- Department of Pathology and Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Abstract
Many genetic alterations that contribute to CNS tumorigenesis and progression have been identified. One goal of such studies is to identify loci that would serve as diagnostic prognostic markers or both. A significant advance is the observation that chromosome 1p loss identified anaplastic oligodendroglioma and a subset of high-grade glioma patients who responded to chemotherapy and had longer survival times. Combined 1p and 19q loss was a predictor of prolonged survival of patients having pure oligodendrogliomas. Such markers eventually may be used to identify patients upfront who would benefit from treatment, while sparing patients who would not benefit. Although many molecular participants involved in the biologic pathways that promote proliferation, angiogenesis, and invasion have been elucidated, there are still many gaps in clinicians' knowledge. It is expected that the use of the human genome project information and databases such as SAGEmap, in combination with techniques such as cDNA arrays and proteomics, will facilitate greatly the identification of novel genes that contribute to CNS tumors. cDNA arrays and tissue arrays will permit the construction of CNS-specific screening tools that will permit the identification of tumor-specific mutations and alterations so that patient-specific therapies can be designed.
Collapse
Affiliation(s)
- S A Rempel
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA.
| |
Collapse
|
21
|
Bouton CM, Hossain MA, Frelin LP, Laterra J, Pevsner J. Microarray analysis of differential gene expression in lead-exposed astrocytes. Toxicol Appl Pharmacol 2001; 176:34-53. [PMID: 11578147 DOI: 10.1006/taap.2001.9274] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The toxic metal lead is a widespread environmental health hazard that can adversely affect human health. In an effort to better understand the cellular and molecular consequences of lead exposure, we have employed cDNA microarrays to analyze the effects of acute lead exposure on large-scale gene expression patterns in immortalized rat astrocytes. Our studies identified many genes previously reported to be differentially regulated by lead exposure. Additionally, we have identified novel putative targets of lead-mediated toxicity, including members of the family of calcium/phospholipid binding annexins, the angiogenesis-inducing thrombospondins, collagens, and tRNA synthetases. We demonstrate the ability to distinguish lead-exposed samples from control or sodium samples solely on the basis of large-scale gene expression patterns using two complementary clustering methods. We have confirmed the altered expression of candidate genes and their encoded proteins by RT-PCR and Western blotting, respectively. Finally, we show that the calcium-dependent phospholipid binding protein annexin A5, initially identified as a differentially regulated gene by our microarray analysis, is directly bound and activated by nanomolar concentrations of lead. We conclude that microarray technology is an effective tool for the identification of lead-induced patterns of gene expression and molecular targets of lead.
Collapse
Affiliation(s)
- C M Bouton
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
22
|
Abstract
The thrombospondins (TSPs) are a family of five secreted proteins that are widely distributed in the extracellular matrix of numerous tissues. TSPs are multimodular and each domain specifies a distinct biological function through interaction with a specific receptor. TSP1 and TSP2 have anti-angiogenic activity, which, at least for TSP1, involves interaction with the microvascular endothelial cell receptor CD36. Expression of TSP1 and TSP2 is modulated by hypoxia and by oncogenes. In several tumors (thyroid, colon, bladder carcinomas), TSP1 expression is inversely correlated with tumor grade and survival rate, whereas in others (e.g. breast carcinomas), it is correlated with the stromal response and is of little prognostic value. Recent studies suggest that TSPs or TSP-derived peptides retaining biological activity could be developed into promising new therapeutic strategies for the anti-angiogenic treatment of solid tumors.
Collapse
Affiliation(s)
- F de Fraipont
- INSERM EMI 0105, Dept of Molecular and Structural Biology, Commissariat à l'Energie Atomique, Grenoble, France
| | | | | | | |
Collapse
|
23
|
|
24
|
Abstract
Quantitative determination of the degree of vascularity has been shown to be independently prognostically significant in many human tumor types. In particular, tumor vascularity has known importance in astrocytomas, in which endothelial proliferation is a criterion for anaplasia in many grading schemes. This review analyzes reports of microvessel quantification performed on histologic sections of human brain tumors, and in which correlations with clinical outcome, or other pathobiologic factors have been made. Among the conclusions are: (1) brain tumors have the unique feature of complex 'glomeruloid' vessels, as well as heterogeneity of microvascular distribution and caliber; (2) lower-grade astrocytomas may incorporate pre-existing vessels, while glioblastomas may develop new vessels; (3) quantification may have additional independent prognostic value over and above routine histologic grade in low-grade astrocytomas with low tumor cell proliferative indices. These findings have implications for the appropriateness of antiangiogenic therapies.
Collapse
Affiliation(s)
- R D Folkerth
- Department of Pathology (Neuropathology), Brigham and Women's Hospital and Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Abstract
One of the most recent and exciting approaches in cancer gene therapy is the ability to target the developing blood supply of the tumor. An appealing feature of antiangiogenic gene therapy is that the tumor vasculature is a readily accessible target, particularly when the carrier and its gene are administered systemically. This is in contrast to several other gene therapy approaches in which the tumor vasculature represents a major obstacle to achieving high levels of transfection of the tumor cells. Several gene-based viral or non-viral therapies that target tumor angiogenesis have shown efficacy in pre-clinical models. Genes that encode antiangiogenic polypeptides such as angiostatin and endostatin have significantly inhibited tumor growth, inducing a microscopic dormant state. The products of these genes are thought to act extracellularly to inhibit angiogenesis. An alternative approach that investigators have used successfully in tumor-bearing mice is to target angiogenic growth factors or their receptors that are essential for tumor growth. Levels of angiogenic factors such as vascular endothelial growth factor (VEGF) have been reduced by either antisense methods or the use of genes encoding truncated angiogenic decoy receptors. Despite these promising findings of tumor reduction with antiangiogenic gene therapy, advances in the viral and/or non-viral delivery systems are essential for this therapy to have clinical utility. In this review, we will discuss the mechanisms of angiogenesis/antiangiogenesis, and the current status and future directions of antiangiogenic gene therapy.
Collapse
Affiliation(s)
- L Zhang
- Department of Pathology, University of Maryland at Baltimore, MD 21201
| | - Q R Chen
- Department of Pathology, University of Maryland at Baltimore, MD 21201
| | - A J Mixson
- Department of Pathology, University of Maryland at Baltimore, MD 21201
| |
Collapse
|