1
|
Pournajaf S, Afsordeh N, Pourgholami MH. In vivo C6 glioma models: an update and a guide toward a more effective preclinical evaluation of potential anti-glioblastoma drugs. Rev Neurosci 2024; 35:183-195. [PMID: 37651618 DOI: 10.1515/revneuro-2023-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023]
Abstract
Glioblastoma multiform (GBM) is the most common primary brain tumor with a poor prognosis and few therapeutic choices. In vivo, tumor models are useful for enhancing knowledge of underlying GBM pathology and developing more effective therapies/agents at the preclinical level, as they recapitulate human brain tumors. The C6 glioma cell line has been one of the most widely used cell lines in neuro-oncology research as they produce tumors that share the most similarities with human GBM regarding genetic, invasion, and expansion profiles and characteristics. This review provides an overview of the distinctive features and the different animal models produced by the C6 cell line. We also highlight specific applications of various C6 in vivo models according to the purpose of the study and offer some technical notes for more convenient/repeatable modeling. This work also includes novel findings discovered in our laboratory, which would further enhance the feasibility of the model in preclinical GBM investigations.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| | - Nastaran Afsordeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| | | |
Collapse
|
2
|
Yadav N, Babu D, Madigubba S, Panigrahi M, Phanithi PB. Tyrphostin A9 attenuates glioblastoma growth by suppressing PYK2/EGFR-ERK signaling pathway. J Neurooncol 2023; 163:675-692. [PMID: 37415005 DOI: 10.1007/s11060-023-04383-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/24/2023] [Indexed: 07/08/2023]
Abstract
PURPOSE Glioblastoma (GBM) is a fatal primary brain tumor with extremely poor clinical outcomes. The anticancer efficiency of tyrosine kinase inhibitors (TKIs) has been shown in GBM and other cancer, with limited therapeutic outcomes. In the current study, we aimed to investigate the clinical impact of active proline-rich tyrosine kinase-2 (PYK2) and epidermal growth factor receptor (EGFR) in GBM and evaluate its druggability by a synthetic TKI-Tyrphostin A9 (TYR A9). METHODS The expression profile of PYK2 and EGFR in astrocytoma biopsies (n = 48) and GBM cell lines were evaluated through quantitative PCR, western blots, and immunohistochemistry. The clinical association of phospho-PYK2 and EGFR was analyzed with various clinicopathological features and the Kaplan-Meier survival curve. The phospho-PYK2 and EGFR druggability and subsequent anticancer efficacy of TYR A9 was evaluated in GBM cell lines and intracranial C6 glioma model. RESULTS Our expression data revealed an increased phospho-PYK2, and EGFR expression aggravates astrocytoma malignancy and is associated with patients' poor survival. The mRNA and protein correlation analysis showed a positive association between phospho-PYK2 and EGFR in GBM tissues. The in-vitro studies demonstrated that TYR A9 reduced GBM cell growth, cell migration, and induced apoptosis by attenuating PYK2/EGFR-ERK signaling. The in-vivo data showed TYR A9 treatment dramatically reduced glioma growth with augmented animal survival by repressing PYK2/EGFR-ERK signaling. CONCLUSION Altogether, this study report that increased phospho-PYK2 and EGFR expression in astrocytoma was associated with poor prognosis. The in-vitro and in-vivo evidence underlined translational implication of TYR A9 by suppressing PYK2/EGFR-ERK modulated signaling pathway. The schematic diagram displayed proof of concept of the current study indicating activated PYK2 either through the Ca2+/Calmodulin-dependent protein kinase II (CAMKII) signaling pathway or autophosphorylation at Tyr402 induces association to the SH2 domain of c-Src that leads to c-Src activation. Activated c-Src in turn activates PYK2 at other tyrosine residues that recruit Grb2/SOS complex and trigger ERK½ activation. Besides, PYK2 interaction with c-Src acts as an upstream of EGFR transactivator that can activate the ERK½ signaling pathway, which induces cell proliferation and cell survival by increasing anti-apoptotic proteins or inhibiting pro-apoptotic proteins. TYR A9 treatment attenuate GBM cell proliferation and migration; and induce GBM cell death by inhibiting PYK2 and EGFR-induced ERK activation.
Collapse
Affiliation(s)
- Neera Yadav
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Deepak Babu
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Sailaja Madigubba
- Department of Laboratory Medicine, Krishna Institute of Medical Sciences, Secunderabad, Telangana, 500 003, India
| | - Manas Panigrahi
- Department of Neurosurgery, Krishna Institute of Medical Sciences, Secunderabad, Telangana, 500 003, India
| | - Prakash Babu Phanithi
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
3
|
Zhu M, Lv B, Ge W, Cui Z, Zhao K, Feng Y, Yang X. Suppression of store-operated Ca 2+ entry regulated by silencing Orai1 inhibits C6 glioma cell motility via decreasing Pyk2 activity and promoting focal adhesion. Cell Cycle 2020; 19:3468-3479. [PMID: 33269647 DOI: 10.1080/15384101.2020.1843814] [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] [Indexed: 12/18/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) plays an important role in regulating Ca2+ influx, which participates in tumor cell survival and motility. We aim to elucidate the role of SOCE in the behavior of C6 glioma cells. Lentiviral vector inserted with the Orai1-targeting shRNA was used to inhibit SOCE in C6 glioma cells. The down-regulation of Orai1 was confirmed by western blot. The ability of shOrai1 or SOCE inhibitor (SKF96365) in regulating SOCE inhibition was evaluated by measuring Ca2+ concentration. Additionally, its effect on cell behavior was assessed using methyl thiazolyl tetrazolium (MTT) assay, wound healing assay, transwell assay, and adhesion assay. Focal adhesions were visualized by immunofluorescence assay. Further, the expression of proline-rich tyrosine kinase 2 (Pyk2) and phosphorylated Pyk2 (p-Pyk2) was analyzed using western blot. Both, SKF96365 treatment and the Orai1 down-regulation inhibited SOCE by perturbing Ca2+ influx. The inhibitory effects of shOrai1 on C6 cell proliferation, migration, and invasion were similar to that of SKF96365. Moreover, Orai1 inhibition enhanced C6 cell adhesion by increasing the size of focal adhesion plaques. The down-regulation of Pyk2 was observed in both SKF96365-treated and Orai1-silenced C6 cells. Additionally, Orai1 inhibition blocked AKT/mTOR, NFAT, and NF-κB pathways. The silencing of Orai1 inhibited the C6 glioma cell migration, invasion and contributed to focal adhesion.
Collapse
Affiliation(s)
- Meng Zhu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University , Qingdao, China.,Department of Neurosurgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Bingke Lv
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University , Qingdao, China
| | - Wenjing Ge
- Department of Radiology, Qingdao Municipal Hospital , Qingdao, China
| | - Zhenwen Cui
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University , Qingdao, China
| | - Kai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University , Qingdao, China
| | - Yugong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University , Qingdao, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital , Tianjin, China
| |
Collapse
|
4
|
Indovina P, Forte IM, Pentimalli F, Giordano A. Targeting SRC Family Kinases in Mesothelioma: Time to Upgrade. Cancers (Basel) 2020; 12:cancers12071866. [PMID: 32664483 PMCID: PMC7408838 DOI: 10.3390/cancers12071866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant mesothelioma (MM) is a deadly tumor mainly caused by exposure to asbestos. Unfortunately, no current treatment is able to change significantly the natural history of the disease, which has a poor prognosis in the majority of patients. The non-receptor tyrosine kinase SRC and other SRC family kinase (SFK) members are frequently hyperactivated in many cancer types, including MM. Several works have indeed suggested that SFKs underlie MM cell proliferation, survival, motility, and invasion, overall affecting multiple oncogenic pathways. Consistently, SFK inhibitors effectively counteracted MM cancerous features at the preclinical level. Dasatinib, a multi-kinase inhibitor targeting SFKs, was also assessed in clinical trials either as second-line treatment for patients with unresectable MM or, more recently, as a neoadjuvant agent in patients with resectable MM. Here, we provide an overview of the molecular mechanisms implicating SFKs in MM progression and discuss possible strategies for a more successful clinical application of SFK inhibitors. Our aim is to stimulate discussion and further consideration of these agents in better designed preclinical and clinical studies to make the most of another class of powerful antitumoral drugs, which too often are lost in translation when applied to MM.
Collapse
Affiliation(s)
- Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), I-80131 Naples, Italy
- Correspondence: (P.I.); (F.P.)
| | - Iris Maria Forte
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Naples, Italy;
| | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Naples, Italy;
- Correspondence: (P.I.); (F.P.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Department of Medical Biotechnologies, University of Siena, I-53100 Siena, Italy
| |
Collapse
|
5
|
Neamati F, Asemi Z. The effects of melatonin on signaling pathways and molecules involved in glioma. Fundam Clin Pharmacol 2019; 34:192-199. [PMID: 31808968 DOI: 10.1111/fcp.12526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/13/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022]
Abstract
Glioblastoma is one of the most common brain tumors with high invasion and malignancy. Despite extensive research in this area and the use of new and advanced therapies, the survival rate in this disease is very low. In addition, resistance to treatment has also been observed in this disease. One of the reasons for rapid progression and failure in treatment for this disease is the presence of a class of cells with high proliferation and high differentiation, a class called glioblastoma stem-like cells shown as being the source of glioblastoma tumors. It has been reported that several oncogenes are expressed in this disease. One important issue in recognizing the pathogenesis of this disease, and which could improve the treatment process, is the identification of involved oncogenes as well as molecules that affect the reduction of the expression of these oncogenes. Melatonin regulates the biological rhythm and inhibits the proliferation of malignant glioma cells due to antioxidant and anti-apoptotic effects. Melatonin has been considered in biological processes and in signaling pathways involved in the development of glioma. The aim of this review is to investigate the effects of melatonin on signaling pathways and molecules involved in the progression of glioma.
Collapse
Affiliation(s)
- Foroogh Neamati
- Department of Microbiology, Kashan University of Medical Sciences, Kashan, 87159-88141, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, 87159-88141, I.R. Iran
| |
Collapse
|
6
|
Abstract
Proline-rich tyrosine kinase 2 (Pyk2) plays essential roles in tumorigenesis and tumor progression. Pyk2 serves as a non-receptor tyrosine kinase regulating tumor cell survival, proliferation, migration, invasion, metastasis, and chemo-resistance, and is associated with poor prognosis and shortened survival in various cancer types. Thus, Pyk2 has been traditionally regarded as an oncogene and potential therapeutic target for cancers. However, a few studies have also demonstrated that Pyk2 exerts tumor-suppressive effects in some cancers, and anti-cancer treatment of Pyk2 inhibitors may only achieve marginal benefits in these cancers. Therefore, more detailed knowledge of the contradictory functions of Pyk2 is needed. In this review, we summarized the tissue distribution, expression, interactive molecules of Pyk2 in the signaling pathway, and roles of Pyk2 in cancers, and focused on regulation of the interconnectivity between Pyk2 and its downstream targets. The potential use of inhibitors of Pyk2 and its related pathways in cancer therapy is also discussed.
Collapse
Affiliation(s)
- Ting Shen
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, China (mainland).,Department of Gastroenterology, Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Qiang Guo
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, China (mainland).,Department of Gastroenterology, Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
7
|
Gagner JP, Sarfraz Y, Ortenzi V, Alotaibi FM, Chiriboga LA, Tayyib AT, Douglas GJ, Chevalier E, Romagnoli B, Tuffin G, Schmitt M, Lemercier G, Dembowsky K, Zagzag D. Multifaceted C-X-C Chemokine Receptor 4 (CXCR4) Inhibition Interferes with Anti-Vascular Endothelial Growth Factor Therapy-Induced Glioma Dissemination. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2080-2094. [PMID: 28734730 PMCID: PMC5809520 DOI: 10.1016/j.ajpath.2017.04.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/05/2017] [Indexed: 01/31/2023]
Abstract
Resistance to antiangiogenic therapy in glioblastoma (GBM) patients may involve hypoxia-induced expression of C-X-C motif chemokine receptor 4 (CXCR4) on invading tumor cells, macrophage/microglial cells (MGCs), and glioma stem cells (GSCs). We determined whether antagonizing CXCR4 with POL5551 disrupts anti-vascular endothelial growth factor (VEGF) therapy-induced glioma growth and dissemination. Mice bearing orthotopic CT-2A or GL261 gliomas received POL5551 and/or anti-VEGF antibody B20-4.1.1. Brain tissue was analyzed for tumor volume, invasiveness, hypoxia, vascular density, proliferation, apoptosis, GSCs, and MGCs. Glioma cells were evaluated for CXCR4 expression and polymorphism and POL5551's effects on CXCR4 ligand binding, cell viability, and migration. No CXCR4 mutations were identified. POL5551 inhibited CXCR4 binding to its ligand, stromal cell-derived factor-1α, and reduced hypoxia- and stromal cell-derived factor-1α-mediated migration dose-dependently but minimally affected cell viability. In vivo, B20-4.1.1 increased hypoxic foci and invasiveness, as seen in GBM patients receiving anti-VEGF therapy. Combination of POL5551 and B20-4.1.1 reduced both glioma invasiveness by 16% to 39% and vascular density compared to B20-4.1.1 alone in both glioma models. Reduced populations of GSCs and MGCs were also seen in CT-2A tumors. POL5551 concentrations, evaluated by mass spectrometry, were higher in tumors than in neighboring brain tissues, likely accounting for the results. Inhibition of CXCR4-regulated tumoral, stem cell, and immune mechanisms by adjunctive CXCR4 antagonists may help overcome antiangiogenic therapy resistance, benefiting GBM patients.
Collapse
Affiliation(s)
- Jean-Pierre Gagner
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University Langone Medical Center, New York, New York; Department of Pathology, New York University Langone Medical Center, New York, New York
| | - Yasmeen Sarfraz
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University Langone Medical Center, New York, New York; Department of Pathology, New York University Langone Medical Center, New York, New York
| | - Valerio Ortenzi
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University Langone Medical Center, New York, New York; Department of Pathology, New York University Langone Medical Center, New York, New York
| | - Fawaz M Alotaibi
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University Langone Medical Center, New York, New York; Department of Pathology, New York University Langone Medical Center, New York, New York
| | - Luis A Chiriboga
- Department of Pathology, New York University Langone Medical Center, New York, New York
| | - Awab T Tayyib
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University Langone Medical Center, New York, New York; Department of Pathology, New York University Langone Medical Center, New York, New York
| | | | | | | | | | | | | | | | - David Zagzag
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University Langone Medical Center, New York, New York; Department of Pathology, New York University Langone Medical Center, New York, New York; Division of Neuropathology, New York University Langone Medical Center, New York, New York; Department of Neurosurgery, New York University Langone Medical Center, New York, New York; New York University Langone Laura and Isaac Perlmutter Cancer Center, New York, New York.
| |
Collapse
|
8
|
Xu R, Han M, Xu Y, Zhang X, Zhang C, Zhang D, Ji J, Wei Y, Wang S, Huang B, Chen A, Zhang Q, Li W, Sun T, Wang F, Li X, Wang J. Coiled-coil domain containing 109B is a HIF1α-regulated gene critical for progression of human gliomas. J Transl Med 2017; 15:165. [PMID: 28754121 PMCID: PMC5534085 DOI: 10.1186/s12967-017-1266-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/19/2017] [Indexed: 02/08/2023] Open
Abstract
Background The coiled-coil domain is a structural motif found in proteins that participate in a variety of biological processes. Aberrant expression of such proteins has been shown to be associated with the malignant behavior of human cancers. In this study, we investigated the role of a specific family member, coiled-coil domain containing 109B (CCDC109B), in human gliomas. Methods and results We confirmed that CCDC109B was highly expressed in high grade gliomas (HGG; WHO III–IV) using immunofluorescence, western blot analysis, immunohistochemistry (IHC) and open databases. Through Cox regression analysis of The Cancer Genome Atlas (TCGA) database, we found that the expression levels of CCDC109B were inversely correlated with patient overall survival and it could serve as a prognostic marker. Then, a serious of cell functional assays were performed in human glioma cell lines, U87MG and U251, which indicated that silencing of CCDC109B attenuated glioma proliferation and migration/invasion both in vitro and in vivo. Notably, IHC staining in primary glioma samples interestingly revealed localization of elevated CCDC109B expression in necrotic areas which are typically hypoxic. Moreover, small interfering RNA (siRNA) and specific inhibiters of HIF1α led to decreased expression of CCDC109B in vitro and in vivo. Transwell assay further showed that CCDC109B is a critical factor in mediating HIF1α-induced glioma cell migration and invasion. Conclusion Our study elucidated a role for CCDC109B as an oncogene and a prognostic marker in human gliomas. CCDC109B may provide a novel therapeutic target for the treatment of human glioma. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1266-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ran Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Yangyang Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Chao Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Yuzhen Wei
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China.,Department of Neurosurgery, Jining No.1 People's Hospital, Jiankang Road, Jining, 272011, China
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Qing Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Feng Wang
- Ningxia Key Laboratory of Craniocerebral Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China.
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China. .,Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| |
Collapse
|
9
|
Couto M, Mastandrea I, Cabrera M, Cabral P, Teixidor F, Cerecetto H, Viñas C. Small-Molecule Kinase-Inhibitors-Loaded Boron Cluster as Hybrid Agents for Glioma-Cell-Targeting Therapy. Chemistry 2017; 23:9233-9238. [PMID: 28605114 DOI: 10.1002/chem.201701965] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Indexed: 12/30/2022]
Abstract
The reported new anilinoquinazoline-icosahedral borane hybrids have been evaluated as glioma targeting for potential use in cancer therapy. Their anti-glioma activity depends on hybrids' lipophilicity; the most powerful compound against glioma cells, a 1,7-closo-derivative, displayed at least 3.3 times higher activity than the parent drug erlotinib. According to the cytotoxic effects on normal glia cells, the hybrids were selective for epidermal growth factor receptor (EGFR)-overexpressed tumor cells. These boron carriers could be used to enrich glioma cancer cells with boron for cancer therapy.
Collapse
Affiliation(s)
- Marcos Couto
- Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400, Montevideo, Uruguay.,Institut de Ciències dels Materials de Barcelona, ICMAB-CSIC Campus UAB, 08193, Bellaterra, Spain.,Área de Radiofarmacia, Centro de Investigaciones Nucleares Facultad de Ciencias, Universidad de la República, Mataojo 2055, 11400, Montevideo, Uruguay
| | - Ignacio Mastandrea
- Centro Universitario Paysandú. CenUR Litoral Norte, Universidad de la República., 60000, Paysandú, Uruguay.,Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Mauricio Cabrera
- Centro Universitario Paysandú. CenUR Litoral Norte, Universidad de la República., 60000, Paysandú, Uruguay
| | - Pablo Cabral
- Área de Radiofarmacia, Centro de Investigaciones Nucleares Facultad de Ciencias, Universidad de la República, Mataojo 2055, 11400, Montevideo, Uruguay
| | - Francesc Teixidor
- Institut de Ciències dels Materials de Barcelona, ICMAB-CSIC Campus UAB, 08193, Bellaterra, Spain
| | - Hugo Cerecetto
- Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400, Montevideo, Uruguay.,Área de Radiofarmacia, Centro de Investigaciones Nucleares Facultad de Ciencias, Universidad de la República, Mataojo 2055, 11400, Montevideo, Uruguay
| | - Clara Viñas
- Institut de Ciències dels Materials de Barcelona, ICMAB-CSIC Campus UAB, 08193, Bellaterra, Spain
| |
Collapse
|
10
|
Simon T, Gagliano T, Giamas G. Direct Effects of Anti-Angiogenic Therapies on Tumor Cells: VEGF Signaling. Trends Mol Med 2017; 23:282-292. [DOI: 10.1016/j.molmed.2017.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/04/2017] [Accepted: 01/09/2017] [Indexed: 12/18/2022]
|
11
|
van Beijnum JR, Nowak-Sliwinska P, Huijbers EJM, Thijssen VL, Griffioen AW. The great escape; the hallmarks of resistance to antiangiogenic therapy. Pharmacol Rev 2015; 67:441-61. [PMID: 25769965 DOI: 10.1124/pr.114.010215] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The concept of antiangiogenic therapy in cancer treatment has led to the approval of different agents, most of them targeting the well known vascular endothelial growth factor pathway. Despite promising results in preclinical studies, the efficacy of antiangiogenic therapy in the clinical setting remains limited. Recently, awareness has emerged on resistance to antiangiogenic therapies. It has become apparent that the intricate complex interplay between tumors and stromal cells, including endothelial cells and associated mural cells, allows for escape mechanisms to arise that counteract the effects of these targeted therapeutics. Here, we review and discuss known and novel mechanisms that contribute to resistance against antiangiogenic therapy and provide an outlook to possible improvements in therapeutic approaches.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands (J.R.v.B., E.J.M.H., V.L.T., A.W.G.); and Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology, Lausanne, Switzerland (P.N.-S.)
| | - Patrycja Nowak-Sliwinska
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands (J.R.v.B., E.J.M.H., V.L.T., A.W.G.); and Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology, Lausanne, Switzerland (P.N.-S.)
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands (J.R.v.B., E.J.M.H., V.L.T., A.W.G.); and Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology, Lausanne, Switzerland (P.N.-S.)
| | - Victor L Thijssen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands (J.R.v.B., E.J.M.H., V.L.T., A.W.G.); and Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology, Lausanne, Switzerland (P.N.-S.)
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands (J.R.v.B., E.J.M.H., V.L.T., A.W.G.); and Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology, Lausanne, Switzerland (P.N.-S.)
| |
Collapse
|
12
|
Meguenani M, Miljkovic-Licina M, Fagiani E, Ropraz P, Hammel P, Aurrand-Lions M, Adams RH, Christofori G, Imhof BA, Garrido-Urbani S. Junctional adhesion molecule B interferes with angiogenic VEGF/VEGFR2 signaling. FASEB J 2015; 29:3411-25. [PMID: 25911611 DOI: 10.1096/fj.15-270223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/16/2015] [Indexed: 12/22/2022]
Abstract
De novo formation of blood vessels is a pivotal mechanism during cancer development. During the past few years, antiangiogenic drugs have been developed to target tumor vasculature. However, because of limitations and adverse effects observed with current therapies, there is a strong need for alternative antiangiogenic strategies. Using specific anti-junctional adhesion molecule (JAM)-B antibodies and Jam-b-deficient mice, we studied the role in antiangiogenesis of JAM-B. We found that antibodies against murine JAM-B, an endothelium-specific adhesion molecule, inhibited microvessel outgrowth from ex vivo aortic rings and in vitro endothelial network formation. In addition, anti-JAM-B antibodies blocked VEGF signaling, an essential pathway for angiogenesis. Moreover, increased aortic ring branching was observed in aortas isolated from Jam-b-deficient animals, suggesting that JAM-B negatively regulates proangiogenic pathways. In mice, JAM-B expression was detected in de novo-formed blood vessels of tumors, but anti-JAM-B antibodies unexpectedly did not reduce tumor growth. Accordingly, JAM-B deficiency in vivo had no impact on blood vessel formation, suggesting that targeting JAM-B in vivo may be offset by other proangiogenic mechanisms. In conclusion, despite the promising effects observed in vitro, targeting JAM-B during tumor progression seems to be inefficient as a stand-alone antiangiogenesis therapy.
Collapse
Affiliation(s)
- Mehdi Meguenani
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Marijana Miljkovic-Licina
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Ernesta Fagiani
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Patricia Ropraz
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Philippe Hammel
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Michel Aurrand-Lions
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Ralf H Adams
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Gerhard Christofori
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Beat A Imhof
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| | - Sarah Garrido-Urbani
- *Department of Pathology and Immunology, Medical Faculty, University Medical Center, University of Geneva, Geneva, Switzerland; Department of Biomedicine, Institute of Biochemistry and Genetics, University of Basel, Basel, Switzerland; Unité Mixte de Recherche 1068, Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Marseille Université, Marseille, France; Unité Mixte de Recherche 7258, Centre National de la Recherche Scientifique, Marseille, France; Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Münster, Germany; and Faculty of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
13
|
Xu CS, Wang ZF, Huang XD, Dai LM, Cao CJ, Li ZQ. Involvement of ROS-alpha v beta 3 integrin-FAK/Pyk2 in the inhibitory effect of melatonin on U251 glioma cell migration and invasion under hypoxia. J Transl Med 2015; 13:95. [PMID: 25889845 PMCID: PMC4371719 DOI: 10.1186/s12967-015-0454-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Melatonin, a well-known antioxidant, has been shown to possess anti-invasive properties for glioma. However, little is known about the effect of melatonin on glioma cell migration and invasion under hypoxia, which is a crucial microenvironment for tumor progress. In addition, focal adhesion kinase (FAK) and proline-rich tyrosine kinase 2 (Pyk2) are closely associated with cell migration and invasion. Therefore, we investigated the possible role of these kinases and its related signaling in the regulation of human U251 glioma cells behavior by melatonin under hypoxia. METHODS The abilities of migration and invasion of U251 glioma cells were determined by wound healing and transwell assay in vitro. The intracellular production of reactive oxygen species (ROS) was measured by using the fluorescent probe 6-carboxy-2', 7'-dichorodihydrofluorescein diacetate (DCFH-DA). Immunofluorescence experiments and western blotting analysis were used to detect the expression level of protein. Small interfering RNAs (siRNA) was used to silence specific gene expression. RESULTS The pharmacologic concentration (1 mM) of melatonin significantly inhibited the migration and invasion of human U251 glioma cells under hypoxia. The inhibitory effect of melatonin was accompanied with the reduced phosphorylation of FAK and Pyk2, and decreased expression of alpha v beta 3 (αvβ3) integrin. Additionally, inhibition of αvβ3 integrin by siRNA reduced the phosphorylation of FAK/Pyk2 and demonstrated the similar anti-tumor effects as melatonin, suggesting the involvement of αvβ3 integrin- FAK/Pyk2 pathway in the anti-migratory and anti-invasive effect of melatonin. It was also found that melatonin treatment decreased the ROS levels in U251 glioma cells cultured under hypoxia. ROS inhibitor apocynin not only inhibited αvβ3 integrin expression and the phosphorylation levels of FAK and Pyk2, but also suppressed the migratory and invasive capacity of U251 glioma cells under hypoxia. CONCLUSIONS These results suggest that melatonin exerts anti-migratory and anti-invasive effects on glioma cells in response to hypoxia via ROS-αvβ3 integrin-FAK/Pyk2 signaling pathways. This provides evidence that melatonin may be a potential therapeutic molecule targeting the hypoxic microenvironment of glioma.
Collapse
Affiliation(s)
- Cheng-Shi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Ze-Fen Wang
- Department of Physiology, School of basic medical science, Wuhan University, Wuhan, 430071, PR China.
| | - Xiao-Dong Huang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China. .,Department of Neurosurgery, Taihe Hospital of Shiyan, Shiyan, 442000, PR China.
| | - Li-Ming Dai
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Chang-Jun Cao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China. .,Laboratory of Neuro-oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
14
|
Wang P, Cheng H, Wu J, Yan A, Zhang L. STK33 plays an important positive role in the development of human large cell lung cancers with variable metastatic potential. Acta Biochim Biophys Sin (Shanghai) 2015; 47:214-23. [PMID: 25662617 DOI: 10.1093/abbs/gmu136] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Serine/threonine kinase 33 (STK33) is a novel protein that has attracted considerable interest in recent years. Previous research has revealed that STK33 expression plays a special role in cancer cell proliferation. However, the mechanisms of STK33 induction of cancer cells remain largely unknown. In this study, it is demonstrated that STK33 expression varies in NL9980 and L9981 cells which are homogeneous cell lines with similar genetic backgrounds. STK33 can promote cell migration and invasion and suppress p53 gene expression in the NL9980 and L9981 cells. In addition, this protein also promotes epithelial-mesenchymal transition (EMT). Moreover, STK33 knockdown decreases tumor-related gene expression and inhibits cell migration, invasion, and EMT, suggesting that STK33 may be a mediator of signaling pathways that are involved in cancer. In conclusion, our results suggest that STK33 may be an important prognostic marker and a therapeutic target for the metastatic progression of human lung cancer.
Collapse
Affiliation(s)
- Ping Wang
- Department of Thoracic Surgery, First People's Hospital of Yunnan Province, Kunming 650031, China
| | - Hongzhong Cheng
- Department of Thoracic Surgery, First People's Hospital of Yunnan Province, Kunming 650031, China
| | - Jianqiang Wu
- Department of Thoracic Surgery, First People's Hospital of Yunnan Province, Kunming 650031, China
| | - Anrun Yan
- Department of Thoracic Surgery, First People's Hospital of Yunnan Province, Kunming 650031, China
| | - Libin Zhang
- Department of Thoracic Surgery, First People's Hospital of Yunnan Province, Kunming 650031, China
| |
Collapse
|