1
|
Senk A, Fazzari J, Djonov V. Vascular mimicry in zebrafish fin regeneration: how macrophages build new blood vessels. Angiogenesis 2024; 27:397-410. [PMID: 38546923 PMCID: PMC11303510 DOI: 10.1007/s10456-024-09914-y] [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: 01/15/2024] [Accepted: 03/10/2024] [Indexed: 08/07/2024]
Abstract
Vascular mimicry has been thoroughly investigated in tumor angiogenesis. In this study, we demonstrate for the first time that a process closely resembling tumor vascular mimicry is present during physiological blood vessel formation in tissue regeneration using the zebrafish fin regeneration assay. At the fin-regenerating front, vasculature is formed by mosaic blood vessels with endothelial-like cells possessing the morphological phenotype of a macrophage and co-expressing both endothelial and macrophage markers within single cells. Our data demonstrate that the vascular segments of the regenerating tissue expand, in part, through the transformation of adjacent macrophages into endothelial-like cells, forming functional, perfused channels and contributing to the de novo formation of microvasculature. Inhibiting the formation of tubular vascular-like structures by CVM-1118 prevents vascular mimicry and network formation resulting in a 70% shorter regeneration area with 60% reduced vessel growth and a complete absence of any signs of regeneration in half of the fin area. Additionally, this is associated with a significant reduction in macrophages. Furthermore, depleting macrophages using macrophage inhibitor PLX-3397, results in impaired tissue regeneration and blood vessel formation, namely a reduction in the regeneration area and vessel network by 75% in comparison to controls.
Collapse
Affiliation(s)
- Anita Senk
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | | | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
2
|
Cui J, Liu X, Dong W, Liu Y, Ruan X, Zhang M, Wang P, Liu L, Xue Y. SNORD17-mediated KAT6B mRNA 2'-O-methylation regulates vasculogenic mimicry in glioblastoma cells. Cell Biol Toxicol 2023; 39:2841-2860. [PMID: 37058271 DOI: 10.1007/s10565-023-09805-w] [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: 11/09/2022] [Accepted: 03/24/2023] [Indexed: 04/15/2023]
Abstract
Glioblastoma (GBM) is a primary tumor in the intracranial compartment. Vasculogenic mimicry (VM) is a process in which a pipeline of tumor cells that provide blood support to carcinogenic cells is formed, and studying VM could provide a new strategy for clinical targeted treatment of GBM. In the present study, we found that SNORD17 and ZNF384 were significantly upregulated and promoted VM in GBM, whereas KAT6B was downregulated and inhibited VM in GBM. RTL-P assays were performed to verify the 2'-O-methylation of KAT6B by SNORD17; IP assays were used to detect the acetylation of ZNF384 by KAT6B. In addition, the binding of ZNF384 to the promoter regions of VEGFR2 and VE-cadherin promoted transcription, as validated by chromatin immunoprecipitation and luciferase reporter assays. And finally, knockdown of SNORD17 and ZNF384 combined with KAT6B overexpression effectively reduced the xenograft tumor size, prolonged the survival time of nude mice and reduced the number of VM channels. This study reveals a novel mechanism of the SNORD17/KAT6B/ZNF384 axis in modulating VM development in GBM that may provide a new goal for the comprehensive treatment of GBM.
Collapse
Affiliation(s)
- Jingyi Cui
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Xiaobai Liu
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Weiwei Dong
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yunhui Liu
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Mengyang Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Ping Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|
3
|
Maddison K, Faulkner S, Graves MC, Fay M, Bowden NA, Tooney PA. Vasculogenic Mimicry Occurs at Low Levels in Primary and Recurrent Glioblastoma. Cancers (Basel) 2023; 15:3922. [PMID: 37568738 PMCID: PMC10417556 DOI: 10.3390/cancers15153922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Vasculogenic mimicry (VM), the ability of tumour cells to form functional microvasculature without an endothelial lining, may contribute to anti-angiogenic treatment resistance in glioblastoma. We aimed to assess the extent of VM formation in primary and recurrent glioblastomas and to determine whether VM vessels also express prostate-specific membrane antigen (PSMA), a pathological vessel marker. Formalin-fixed paraffin-embedded tissue from 35 matched pairs of primary and recurrent glioblastoma was immunohistochemically labelled for PSMA and CD34 and stained with periodic acid-Schiff (PAS). Vascular structures were categorised as endothelial vessels (CD34+/PAS+) or VM (CD34-/PAS+). Most blood vessels in both primary and recurrent tumours were endothelial vessels, and these significantly decreased in recurrent tumours (p < 0.001). PSMA was expressed by endothelial vessels, and its expression was also decreased in recurrent tumours (p = 0.027). VM was observed in 42.86% of primary tumours and 28.57% of recurrent tumours. VM accounted for only a small proportion of the tumour vasculature and VM density did not differ between primary and recurrent tumours (p = 0.266). The functional contribution of VM and its potential as a treatment target in glioblastoma require further investigation.
Collapse
Affiliation(s)
- Kelsey Maddison
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (K.M.); (S.F.); (P.A.T.)
- Mark Hughes Foundation Centre for Brain Cancer Research, The University of Newcastle, Callaghan, NSW 2308, Australia; (M.C.G.); (M.F.)
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Sam Faulkner
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (K.M.); (S.F.); (P.A.T.)
- Mark Hughes Foundation Centre for Brain Cancer Research, The University of Newcastle, Callaghan, NSW 2308, Australia; (M.C.G.); (M.F.)
| | - Moira C. Graves
- Mark Hughes Foundation Centre for Brain Cancer Research, The University of Newcastle, Callaghan, NSW 2308, Australia; (M.C.G.); (M.F.)
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Michael Fay
- Mark Hughes Foundation Centre for Brain Cancer Research, The University of Newcastle, Callaghan, NSW 2308, Australia; (M.C.G.); (M.F.)
- GenesisCare, Lake Macquarie Private Hospital, Gateshead, NSW 2290, Australia
| | - Nikola A. Bowden
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Paul A. Tooney
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (K.M.); (S.F.); (P.A.T.)
- Mark Hughes Foundation Centre for Brain Cancer Research, The University of Newcastle, Callaghan, NSW 2308, Australia; (M.C.G.); (M.F.)
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
4
|
Zhang A, Zhang S. Clinicopathological significance of vasculogenic mimicry and fetal hemoglobin expression in peripheral neuroblastic tumors in children. Am J Transl Res 2023; 15:4687-4698. [PMID: 37560203 PMCID: PMC10408510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023]
Abstract
PURPOSE Vasculogenic mimicry (VM) is present in a variety of malignant tumors, and is related to the degree of malignancy. Neuroblastoma (NB) can induce the expression of fetal hemoglobin (HB-F). The purpose of this study was to investigate the clinicopathological significance of the number of VMs and tumor cell expression of HB-F in children with peripheral neuroblastic tumors (pNTs). MATERIALS AND METHODS We collected tissue samples and clinical data from 101 children with pNTs; prepared serial sections of tissue wax blocks for hematoxylin and eosin staining, CD31/periodic acid-Schiff double staining, and HB-F immunohistochemical staining; and analyzed the experimental results. RESULTS There were significant differences in the number of VMs and HB-F expression in tumor cells according to the pathological classification of pNTs (P<0.001, collectively). Poorly differentiated NB had a median of 137 VMs and 25.5% HB-F expression. Differentiating NB had a median of 90.5 VMs and 8.5% HB-F expression. Ganglioneuroblastoma intermixed had a median of 6.0 VMs and 1.0% HB-F expression. Ganglioneuromas had no VM and a median of 0% HB-F expression. The number of VMs and the expression of HB-F were significantly higher in the poor prognosis group than the good prognosis group (P<0.001, collectively). There was a strong positive correlation between the number of VMs and HB-F expression in pNTs (r=0.891, P<0.001). CONCLUSION We confirmed VM and HB-F expression in pNTs. The number of VMs and HB-F expression were higher in poorly differentiated tumors. The number of VMs and level of HB-F expression in pNTs might be related to the prognosis.
Collapse
Affiliation(s)
- Aihua Zhang
- Graduate School, Tianjin Medical UniversityTianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical CenterTianjin, China
| |
Collapse
|
5
|
Hatlen RR, Rajagopalan P. Investigating Trans-differentiation of Glioblastoma Cells in an In Vitro 3D Model of the Perivascular Niche. ACS Biomater Sci Eng 2023. [PMID: 37129167 DOI: 10.1021/acsbiomaterials.2c01310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Glioblastoma multiforme (GBM) is the deadliest form of brain cancer, responsible for over 50% of adult brain tumors. A specific region within the GBM environment is known as the perivascular niche (PVN). This area is defined as within approximately 100 μm of vasculature and plays an important role in the interactions between endothelial cells (ECs), astrocytes, GBM cells, and stem cells. We have designed a 3D in vitro model of the PVN comprising either collagen Type 1 or HyStem-C, human umbilical vein ECs (HUVECs), and LN229 (GBM) cells. HUVECs were encapsulated within the hydrogels to form vascular networks. After 7 days, LN229 cells were co-cultured to investigate changes in both cell types. Over a 14 day culture period, we measured alterations in HUVEC networks, the contraction of the hydrogels, trans-differentiation of LN229 cells, and the concentrations of two chemokines; CXCL12 and TGF-β. Increased cellular proliferation ranging from 10- to 16-fold was exhibited in co-cultures from days 8 to 14. This was accompanied with a decrease in the height of hydrogels of up to 68%. These changes in the biomaterial scaffold indicate that LN229-HUVEC interactions promote changes to the matrix. TGF-β and CXCL12 secretion increased approximately 2-2.6-fold each from day 8 to 14 in all co-cultures. The expression of CXCL12 correlated with cell colocalization, indicating a chemotactic role in enabling the migration of LN229 cells toward HUVECs in co-cultures. von Willebrand factor (vWF) was co-expressed with glial fibrillary acidic protein (GFAP) in up to 15% of LN229 cells after 24 h in co-culture. Additionally, when LN229 cells were co-cultured with human brain microvascular ECs, the percentages of GFAP+/vWF+ cells were up to 20% higher than that in co-cultures with HUVECs in collagen (2.2 mg/mL) and HyStem-C gels on day 14. The expression of vWF indicates the early stages of trans-differentiation of LN229 cells to an EC phenotype. Designing in vitro models of trans-differentiation may provide additional insights into how vasculature and cellular phenotypes are altered in GBM.
Collapse
Affiliation(s)
- Rosalyn R Hatlen
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
6
|
Liang Q, Peng J, Xu Z, Li Z, Jiang F, Ouyang L, Wu S, Fu C, Liu Y, Liu Y, Yan Y. Pan-cancer analysis of the prognosis and immunological role of AKAP12: A potential biomarker for resistance to anti-VEGF inhibitors. Front Genet 2022; 13:943006. [PMID: 36110213 PMCID: PMC9468827 DOI: 10.3389/fgene.2022.943006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
The primary or acquired resistance to anti-VEGF inhibitors remains a common problem in cancer treatment. Therefore, identifying potential biomarkers enables a better understanding of the precise mechanism. Through the GEO database, three profiles associated with bevacizumab (BV) resistance to ovarian cancer, glioma, and non-small-cell lung carcinoma, respectively, were collected for the screening process, and two genes were found. A-kinase anchor protein 12 (AKAP12), one of these two genes, correlates with tumorigenesis of some cancers. However, the role of AKAP12 in pan-cancer remains poorly defined. The present study first systematically analyzed the association of AKAP12 with anti-VEGF inhibitors’ sensitivity, clinical prognosis, DNA methylation, protein phosphorylation, and immune cell infiltration across various cancers via bioinformatic tools. We found that AKAP12 was upregulated in anti-VEGF therapy-resistant cancers, including ovarian cancer (OV), glioblastoma (GBM), lung cancer, and colorectal cancer (CRC). A high AKAP12 expression revealed dismal prognoses in OV, GBM, and CRC patients receiving anti-VEGF inhibitors. Moreover, AKAP12 expression was negatively correlated with cancer sensitivity towards anti-VEGF therapy. Clinical prognosis analysis showed that AKAP12 expression predicted worse prognoses of various cancer types encompassing colon adenocarcinoma (COAD), OV, GBM, and lung squamous cell carcinoma (LUSC). Gene mutation status may be a critical cause for the involvement of AKAP12 in resistance. Furthermore, lower expression of AKAP12 was detected in nearly all cancer types, and hypermethylation may explain its decreased expression. A decreased phosphorylation of T1760 was observed in breast cancer, clear-cell renal cell carcinoma, and lung adenocarcinoma. For the immunologic significance, AKAP12 was positively related to the abundance of pro-tumor cancer-associated fibroblasts (CAFs) in various types of cancer. The results of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis suggested that “cell junction organization” and “MAPK pathway” participated in the effect of AKAP12. Importantly, we discovered that AKAP12 expression was greatly associated with metastasis of lung adenocarcinoma as well as differential and angiogenesis of retinoblastoma through investigating the single-cell sequencing data. Our study showed that the dual role of AKAP12 in various cancers and AKAP12 could serve as a biomarker of anti-VEGF resistance in OV, GBM, LUSC, and COAD.
Collapse
Affiliation(s)
- Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhilan Li
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Feng Jiang
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Lingzi Ouyang
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Shangjun Wu
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Chencheng Fu
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Ying Liu
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yuanliang Yan,
| |
Collapse
|
7
|
Qi S, Deng S, Lian Z, Yu K. Novel Drugs with High Efficacy against Tumor Angiogenesis. Int J Mol Sci 2022; 23:6934. [PMID: 35805939 PMCID: PMC9267017 DOI: 10.3390/ijms23136934] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis is involved in physiological and pathological processes in the body. Tumor angiogenesis is a key factor associated with tumor growth, progression, and metastasis. Therefore, there is great interest in developing antiangiogenic strategies. Hypoxia is the basic initiating factor of tumor angiogenesis, which leads to the increase of vascular endothelial growth factor (VEGF), angiopoietin (Ang), hypoxia-inducible factor (HIF-1), etc. in hypoxic cells. The pathways of VEGF and Ang are considered to be critical steps in tumor angiogenesis. A number of antiangiogenic drugs targeting VEGF/VEGFR (VEGF receptor) or ANG/Tie2, or both, are currently being used for cancer treatment, or are still in various stages of clinical development or preclinical evaluation. This article aims to review the mechanisms of angiogenesis and tumor angiogenesis and to focus on new drugs and strategies for the treatment of antiangiogenesis. However, antitumor angiogenic drugs alone may not be sufficient to eradicate tumors. The molecular chaperone heat shock protein 90 (HSP90) is considered a promising molecular target. The VEGFR system and its downstream signaling molecules depend on the function of HSP90. This article also briefly introduces the role of HSP90 in angiogenesis and some HSP90 inhibitors.
Collapse
Affiliation(s)
- Shiyu Qi
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Shoulong Deng
- National Health Commission (NHC) of China Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China;
| | - Zhengxing Lian
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Kun Yu
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
8
|
Deshors P, Arnauduc F, Boëlle B, Cohen-Jonathan Moyal E, Courtade-Saïdi M, Evrard SM. Impact of Regorafenib on Endothelial Transdifferentiation of Glioblastoma Stem-like Cells. Cancers (Basel) 2022; 14:1551. [PMID: 35326702 PMCID: PMC8946617 DOI: 10.3390/cancers14061551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastomas (GBM) are aggressive brain tumours with a poor prognosis despite heavy therapy that combines surgical resection and radio-chemotherapy. The presence of a subpopulation of GBM stem cells (GSC) contributes to tumour aggressiveness, resistance and recurrence. Moreover, GBM are characterised by abnormal, abundant vascularisation. Previous studies have shown that GSC are directly involved in new vessel formation via their transdifferentiation into tumour-derived endothelial cells (TDEC) and that irradiation (IR) potentiates the pro-angiogenic capacity of TDEC via the Tie2 signalling pathway. We therefore investigated the impact of regorafenib, a multikinase inhibitor with anti-angiogenic and anti-tumourigenic activity, on GSC and TDEC obtained from irradiated GSC (TDEC IR+) or non-irradiated GSC (TDEC). Regorafenib significantly decreases GSC neurosphere formation in vitro and inhibits tumour formation in the orthotopic xenograft model. Regorafenib also inhibits transdifferentiation by decreasing CD31 expression, CD31+ cell count, pseudotube formation in vitro and the formation of functional blood vessels in vivo of TDEC and TDEC IR+. All of these results confirm that regorafenib clearly impacts GSC tumour formation and transdifferentiation and may therefore be a promising therapeutic option in combination with chemo/radiotherapy for the treatment of highly aggressive brain tumours.
Collapse
Affiliation(s)
- Pauline Deshors
- Institut Claudius Regaud, IUCT Oncopole, 31059 Toulouse, France; (P.D.); (B.B.); (E.C.-J.M.)
| | - Florent Arnauduc
- Faculty of Medicine, Paul Sabatier University, Toulouse-3, 31062 Toulouse, France; (F.A.); (M.C.-S.)
- INSERM UMR 1037, Centre for Cancer Research of Toulouse, 31100 Toulouse, France
| | - Betty Boëlle
- Institut Claudius Regaud, IUCT Oncopole, 31059 Toulouse, France; (P.D.); (B.B.); (E.C.-J.M.)
| | - Elizabeth Cohen-Jonathan Moyal
- Institut Claudius Regaud, IUCT Oncopole, 31059 Toulouse, France; (P.D.); (B.B.); (E.C.-J.M.)
- Faculty of Medicine, Paul Sabatier University, Toulouse-3, 31062 Toulouse, France; (F.A.); (M.C.-S.)
- INSERM UMR 1037, Centre for Cancer Research of Toulouse, 31100 Toulouse, France
| | - Monique Courtade-Saïdi
- Faculty of Medicine, Paul Sabatier University, Toulouse-3, 31062 Toulouse, France; (F.A.); (M.C.-S.)
- INSERM UMR 1037, Centre for Cancer Research of Toulouse, 31100 Toulouse, France
- Pathology and Cytology Department, CHU Toulouse, IUCT Oncopole, 31059 Toulouse, France
| | - Solène M. Evrard
- Faculty of Medicine, Paul Sabatier University, Toulouse-3, 31062 Toulouse, France; (F.A.); (M.C.-S.)
- INSERM UMR 1037, Centre for Cancer Research of Toulouse, 31100 Toulouse, France
- Pathology and Cytology Department, CHU Toulouse, IUCT Oncopole, 31059 Toulouse, France
| |
Collapse
|
9
|
Ramalho MJ, Loureiro JA, Coelho MAN, Pereira MC. Transferrin Receptor-Targeted Nanocarriers: Overcoming Barriers to Treat Glioblastoma. Pharmaceutics 2022; 14:pharmaceutics14020279. [PMID: 35214012 PMCID: PMC8880499 DOI: 10.3390/pharmaceutics14020279] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and lethal type of brain tumor, and the clinically available approaches for its treatment are not curative. Despite the intensive research, biological barriers such as the blood–brain barrier (BBB) and tumor cell membranes are major obstacles to developing novel effective therapies. Nanoparticles (NPs) have been explored as drug delivery systems (DDS) to improve GBM therapeutic strategies. NPs can circumvent many of the biological barriers posed by this devastating disease, enhancing drug accumulation in the target site. This can be achieved by employing strategies to target the transferrin receptor (TfR), which is heavily distributed in BBB and GBM cells. These targeting strategies comprise the modification of NPs’ surface with various molecules, such as transferrin (Tf), antibodies, and targeting peptides. This review provides an overview and discussion on the recent advances concerning the strategies to target the TfR in the treatment of GBM, as their benefits and limitations.
Collapse
|
10
|
Brown CJ, Simon T, Cilibrasi C, Lynch PJ, Harries RW, Graf AA, Large MJ, Ogilvie SP, Salvage JP, Dalton AB, Giamas G, King AAK. Tuneable synthetic reduced graphene oxide scaffolds elicit high levels of three-dimensional glioblastoma interconnectivity in vitro. J Mater Chem B 2021; 10:373-383. [PMID: 34931630 DOI: 10.1039/d1tb01266e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Three-dimensional tissue scaffolds have utilised nanomaterials to great effect over the last decade. In particular, scaffold design has evolved to consider mechanical structure, morphology, chemistry, electrical properties, and of course biocompatibility - all vital to the performance of the scaffold and how successful they are in developing cell cultures. We have developed an entirely synthetic and tuneable three-dimensional scaffold of reduced graphene oxide (rGO) that shows good biocompatibility, and favourable mechanical properties as well as reasonable electrical conductivity. Importantly, the synthesis is scaleable and suitable for producing scaffolds of any desired geometry and size, and we observe a high level of biocompatibility and cell proliferation for multiple cell lines. In particular, one of the most devastating forms of malignant brain cancer, glioblastoma (GBM), grows especially well on our rGO scaffold in vitro, and without the addition of response-specific growth factors. We have observed that our scaffold elicits spontaneous formation of a high degree of intercellular connections across the GBM culture. This phenomenon is not well documented in vitro and nothing similar has been observed in synthetic scaffolds without the use of response-specific growth factors - which risk obscuring any potential phenotypic behaviour of the cells. The use of scaffolds like ours, which are not subject to the limitations of existing two-dimensional substrate technologies, provide an excellent system for further investigation into the mechanisms behind the rapid proliferation and success of cancers like GBM. These synthetic scaffolds can advance our understanding of these malignancies in the pursuit of improved theranostics against them.
Collapse
Affiliation(s)
- Christopher J Brown
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Thomas Simon
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chiara Cilibrasi
- Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Peter J Lynch
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Rhiannon W Harries
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Aline Amorim Graf
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Matthew J Large
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Sean P Ogilvie
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Jonathan P Salvage
- School of Pharmacy and Biomolecular Sciences, University of Brighton, BN2 4GJ, UK
| | - Alan B Dalton
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Alice A K King
- Department of Physics and Astronomy, University of Sussex, Brighton, BN1 9QH, UK.
| |
Collapse
|
11
|
Wang H, Guo J, Wang T, Wang K, Wu Z, Sun T. Efficacy and safety of bevacizumab in the treatment of adult gliomas: a systematic review and meta-analysis. BMJ Open 2021; 11:e048975. [PMID: 34857558 PMCID: PMC8640637 DOI: 10.1136/bmjopen-2021-048975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE To assess the efficacy and safety of bevacizumab (BEV) in patients with glioma. DESIGN Systematic review and meta-analysis. PARTICIPANTS Adults aged 18 years and above, whose histology was confirmed to be malignant glioma. PRIMARY AND SECONDARY OUTCOME MEASURES The main indicators included progression-free survival (PFS) rate and overall survival (OS) rate, and the secondary indicators were adverse reactions. RESULTS A total of 11 clinical centre trials were included in this study for meta-analysis, including 2392 patients. The results of the meta-analysis showed that the median PFS rate of the BEV group was significantly higher than that of the non-BEV group (p<0.00001). When comparing PFS between two groups, we found that the PFS in the BEV group was higher than that in the non-BEV group at 6 months (OR 3.31, 95% CI 2.74 to 4.00, p<0.00001), 12 months (OR 2.05, 95% CI 1.70 to 2.49, p<0.00001) and 18 months (OR 1.31, 95% CI 1.02 to 1.69, p=0.03). But at 24 months (OR 0.83, 95% CI 0.50 to 1.37, p=0.47), there was no significant difference between the two groups. At 30 months (OR 0.62, 95% CI 0.39 to 0.97, p=0.04), the PFS of the BEV group was lower than that of the non-BEV group. Moreover, The results showed that BEV had no significant effect on improving OS, but the adverse reaction in BEV group was significantly higher than that in non-BEV group. CONCLUSION The evidence suggests that BEV can significantly prolong the PFS of patients with glioma within 18 months and shorten the PFS of patients after 30 months. This limitation may be related to the subgroup of patients, the change of recurrence mode, the optimal dose of drug, the increase of hypoxia, the enhancement of invasiveness and so on. Therefore, it is necessary to carry out more samples and higher quality large-scale research in the future.
Collapse
Affiliation(s)
- Huan Wang
- Department of Medical Imaging, the first affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Jianxin Guo
- Department of Medical Imaging, the first affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Tianze Wang
- Major in Neurosurgery, Xi'an Jiaotong University, Xi'an, China
| | - Kai Wang
- Department of Neurosurgery, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhuojun Wu
- Department of Medical Imaging, the first affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Tianze Sun
- Department of Medical Imaging, the first affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Cruz Flores VA, Menghani H, Mukherjee PK, Marrero L, Obenaus A, Dang Q, Khoutorova L, Reid MM, Belayev L, Bazan NG. Combined Therapy With Avastin, a PAF Receptor Antagonist and a Lipid Mediator Inhibited Glioblastoma Tumor Growth. Front Pharmacol 2021; 12:746470. [PMID: 34630114 PMCID: PMC8498947 DOI: 10.3389/fphar.2021.746470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/06/2021] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive, highly proliferative, invasive brain tumor with a poor prognosis and low survival rate. The current standard of care for GBM is chemotherapy combined with radiation following surgical intervention, altogether with limited efficacy, since survival averages 18 months. Improvement in treatment outcomes for patients with GBM requires a multifaceted approach due to the dysregulation of numerous signaling pathways. Recently emerging therapies to precisely modulate tumor angiogenesis, inflammation, and oxidative stress are gaining attention as potential options to combat GBM. Using a mouse model of GBM, this study aims to investigate Avastin (suppressor of vascular endothelial growth factor and anti-angiogenetic treatment), LAU-0901 (a platelet-activating factor receptor antagonist that blocks pro-inflammatory signaling), Elovanoid; ELV, a novel pro-homeostatic lipid mediator that protects neural cell integrity and their combination as an alternative treatment for GBM. Female athymic nude mice were anesthetized with ketamine/xylazine, and luciferase-modified U87MG tumor cells were stereotactically injected into the right striatum. On post-implantation day 13, mice received one of the following: LAU-0901, ELV, Avastin, and all three compounds in combination. Bioluminescent imaging (BLI) was performed on days 13, 20, and 30 post-implantation. Mice were perfused for ex vivo MRI on day 30. Bioluminescent intracranial tumor growth percentage was reduced by treatments with LAU-0901 (43%), Avastin (77%), or ELV (86%), individually, by day 30 compared to saline treatment. In combination, LAU-0901/Avastin, ELV/LAU-0901, or ELV/Avastin had a synergistic effect in decreasing tumor growth by 72, 92, and 96%, respectively. Additionally, tumor reduction was confirmed by MRI on day 30, which shows a decrease in tumor volume by treatments with LAU-0901 (37%), Avastin (67%), or ELV (81.5%), individually, by day 30 compared to saline treatment. In combination, LAU-0901/Avastin, ELV/LAU-0901, or ELV/Avastin had a synergistic effect in decreasing tumor growth by 69, 78.7, and 88.6%, respectively. We concluded that LAU-0901 and ELV combined with Avastin exert a better inhibitive effect in GBM progression than monotherapy. To our knowledge, this is the first study that demonstrates the efficacy of these novel therapeutic regimens in a model of GBM and may provide the basis for future therapeutics in GBM patients.
Collapse
Affiliation(s)
- Valerie A Cruz Flores
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Pediatrics, Hematology-Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Hemant Menghani
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Pediatrics, Hematology-Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Pranab K Mukherjee
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Luis Marrero
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California Irvin, Irvine, CA, United States
| | - Quan Dang
- Department of Pediatrics, School of Medicine, University of California Irvin, Irvine, CA, United States
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Madigan M Reid
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Ludmila Belayev
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
13
|
Díaz-Flores L, Gutiérrez R, González-Gómez M, García MDP, Díaz-Flores L, González-Marrero I, Ávila J, Martín-Vasallo P. Disproportion in Pericyte/Endothelial Cell Proliferation and Mechanisms of Intussusceptive Angiogenesis Participate in Bizarre Vessel Formation in Glioblastoma. Cells 2021; 10:cells10102625. [PMID: 34685606 PMCID: PMC8534221 DOI: 10.3390/cells10102625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant tumor in the brain. In addition to the vascular pattern with thin-walled vessels and findings of sprouting angiogenesis, GBM presents a bizarre microvasculature (BM) formed by vascular clusters, vascular garlands, and glomeruloid bodies. The mechanisms in BM morphogenesis are not well known. Our objective was to assess the role of pericyte/endothelial proliferation and intussusceptive angiogenic mechanisms in the formation of the BM. For this purpose, we studied specimens of 66 GBM cases using immunochemistry and confocal microscopy. In the BM, the results showed (a) transitional forms between the BM patterns, mostly with prominent pericytes covering all the abluminal endothelial cell (EC) surface of the vessels, (b) a proliferation index high in the prominent pericytes and low in ECs (47.85 times higher in pericytes than in ECs), (c) intravascular pillars (hallmark of intussusceptive angiogenesis) formed by transcapillary interendothelial bridges, endothelial contacts of opposite vessel walls, and vessel loops, and (d) the persistence of these findings in complex glomeruloid bodies. In conclusion, disproportion in pericyte/EC proliferation and mechanisms of intussusceptive angiogenesis participate in BM formation. The contributions have morphogenic and clinical interest since pericytes and intussusceptive angiogenesis can condition antiangiogenic therapy in GBM.
Collapse
Affiliation(s)
- Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Ricardo Gutiérrez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Miriam González-Gómez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain;
| | - María-del-Pino García
- Department of Pathology, Eurofins Megalab–Hospiten Hospitals, 38100 Tenerife, Spain;
| | - Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Ibrahim González-Marrero
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Julio Ávila
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain;
- Department of Bioquímica, Microbiología, Biología Celular y Genética, University of La Laguna, 38206 Tenerife, Spain
| | - Pablo Martín-Vasallo
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain;
- Department of Bioquímica, Microbiología, Biología Celular y Genética, University of La Laguna, 38206 Tenerife, Spain
- Correspondence: ; Tel.: +34-922-318358; Fax: +34-922-319279
| |
Collapse
|
14
|
Pagano C, Navarra G, Pastorino O, Avilia G, Coppola L, Della Monica R, Chiariotti L, Florio T, Corsaro A, Torelli G, Caiazzo P, Gazzerro P, Bifulco M, Laezza C. N6-Isopentenyladenosine Hinders the Vasculogenic Mimicry in Human Glioblastoma Cells through Src-120 Catenin Pathway Modulation and RhoA Activity Inhibition. Int J Mol Sci 2021; 22:ijms221910530. [PMID: 34638872 PMCID: PMC8508824 DOI: 10.3390/ijms221910530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Vasculogenic mimicry (VM) is a functional microcirculation pattern formed by aggressive tumor cells. Thus far, no effective drugs have been developed to target VM. Glioblastoma (GBM) is the most malignant form of brain cancer and is a highly vascularized tumor. Vasculogenic mimicry represents a means whereby GBM can escape anti-angiogenic therapies. METHODS Here, using an in vitro tube formation assay on Matrigel, we evaluated the ability of N6-isopentenyladenosine (iPA) to interfere with vasculogenic mimicry (VM). RhoA activity was assessed using a pull-down assay, while the modulation of the adherens junctions proteins was analyzed by Western blot analysis. RESULTS We found that iPA at sublethal doses inhibited the formation of capillary-like structures suppressing cell migration and invasion of U87MG, U343MG, and U251MG cells, of patient-derived human GBM cells and GBM stem cells. iPA reduces the vascular endothelial cadherin (VE-cadherin) expression levels in a dose-dependent manner, impairs the vasculogenic mimicry network by modulation of the Src/p120-catenin pathway and inhibition of RhoA-GTPase activity. CONCLUSIONS Taken together, our results revealed iPA as a promising novel anti-VM drug in GBM clinical therapeutics.
Collapse
Affiliation(s)
- Cristina Pagano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
| | - Giovanna Navarra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
| | - Olga Pastorino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
| | - Giorgio Avilia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
| | - Laura Coppola
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
| | - Rosa Della Monica
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
- CEINGE—Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Lorenzo Chiariotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
- CEINGE—Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Tullio Florio
- Department of Internal Medicine, University of Genova, Viale Benedetto XV 2, 16136 Genova, Italy; (T.F.); (A.C.)
- IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Alessandro Corsaro
- Department of Internal Medicine, University of Genova, Viale Benedetto XV 2, 16136 Genova, Italy; (T.F.); (A.C.)
| | - Giovanni Torelli
- Neurosurgery Unit A.O. San Giovanni di Dio e Ruggi d’Aragona—Salerno’s School of Medicine Largo Città di Ippocrate, 84131 Salerno, Italy;
- Neurosurgery, Unit A.O. “A.Cardarelli”, 80145 Naples, Italy;
| | | | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Fisciano, 84084 Salerno, Italy;
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80145 Naples, Italy; (C.P.); (G.N.); (O.P.); (G.A.); (L.C.); (R.D.M.); (L.C.)
- Correspondence: (M.B.); or (C.L.)
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), 80145 Naples, Italy
- Correspondence: (M.B.); or (C.L.)
| |
Collapse
|
15
|
Mahjoum S, Rufino-Ramos D, Pereira de Almeida L, Broekman MLD, Breakefield XO, van Solinge TS. Living Proof of Activity of Extracellular Vesicles in the Central Nervous System. Int J Mol Sci 2021; 22:ijms22147294. [PMID: 34298912 PMCID: PMC8303915 DOI: 10.3390/ijms22147294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 12/27/2022] Open
Abstract
The central nervous system (CNS) consists of a heterogeneous population of cells with highly specialized functions. For optimal functioning of the CNS, in disease and in health, intricate communication between these cells is vital. One important mechanism of cellular communication is the release and uptake of extracellular vesicles (EVs). EVs are membrane enclosed particles actively released by cells, containing a wide array of proteins, lipids, RNA, and DNA. These EVs can be taken up by neighboring or distant cells, and influence a wide range of processes. Due to the complexity and relative inaccessibility of the CNS, our current understanding of the role of EVs is mainly derived in vitro work. However, recently new methods and techniques have opened the ability to study the role of EVs in the CNS in vivo. In this review, we discuss the current developments in our understanding of the role of EVs in the CNS in vivo.
Collapse
Affiliation(s)
- Shadi Mahjoum
- Program in Neuroscience, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02119, USA; (S.M.); (M.L.D.B.); (X.O.B.)
| | - David Rufino-Ramos
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.R.-R.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.R.-R.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Marike L. D. Broekman
- Program in Neuroscience, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02119, USA; (S.M.); (M.L.D.B.); (X.O.B.)
- Department of Neurosurgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Neurosurgery, Haaglanden Medical Center, 2512 VA The Hague, The Netherlands
| | - Xandra O. Breakefield
- Program in Neuroscience, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02119, USA; (S.M.); (M.L.D.B.); (X.O.B.)
| | - Thomas S. van Solinge
- Program in Neuroscience, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02119, USA; (S.M.); (M.L.D.B.); (X.O.B.)
- Department of Neurosurgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Correspondence:
| |
Collapse
|
16
|
Advances in Chemokine Signaling Pathways as Therapeutic Targets in Glioblastoma. Cancers (Basel) 2021; 13:cancers13122983. [PMID: 34203660 PMCID: PMC8232256 DOI: 10.3390/cancers13122983] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023] Open
Abstract
With a median patient survival of 15 months, glioblastoma (GBM) is still one of the deadliest malign tumors. Despite immense efforts, therapeutic regimens fail to prolong GBM patient overall survival due to various resistance mechanisms. Chemokine signaling as part of the tumor microenvironment plays a key role in gliomagenesis, proliferation, neovascularization, metastasis and tumor progression. In this review, we aimed to investigate novel therapeutic approaches targeting various chemokine axes, including CXCR2/CXCL2/IL-8, CXCR3/CXCL4/CXCL9/CXCL10, CXCR4/CXCR7/CXCL12, CXCR6/CXCL16, CCR2/CCL2, CCR5/CCL5 and CX3CR1/CX3CL1 in preclinical and clinical studies of GBM. We reviewed targeted therapies as single therapies, in combination with the standard of care, with antiangiogenic treatment as well as immunotherapy. We found that there are many antagonist-, antibody-, cell- and vaccine-based therapeutic approaches in preclinical and clinical studies. Furthermore, targeted therapies exerted their highest efficacy in combination with other established therapeutic applications. The novel chemokine-targeting therapies have mainly been examined in preclinical models. However, clinical applications are auspicious. Thus, it is crucial to broadly investigate the recently developed preclinical approaches. Promising preclinical applications should then be investigated in clinical studies to create new therapeutic regimens and to overcome therapy resistance to GBM treatment.
Collapse
|
17
|
The Carbonic Anhydrase Inhibitor E7070 Sensitizes Glioblastoma Cells to Radio- and Chemotherapy and Reduces Tumor Growth. Mol Neurobiol 2021; 58:4520-4534. [PMID: 34085182 DOI: 10.1007/s12035-021-02437-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/18/2021] [Indexed: 01/13/2023]
Abstract
Glioblastomas (GBMs), the most common and lethal primary brain tumor, show inherent infiltrative nature and high molecular heterogeneity that make complete surgical resection unfeasible and unresponsive to conventional adjuvant therapy. Due to their fast growth rate even under hypoxic and acidic conditions, GBM cells can conserve the intracellular pH at physiological range by overexpressing membrane-bound carbonic anhydrases (CAs). The synthetic sulfonamide E7070 is a potent inhibitor of CAs that harbors putative anticancer properties; however, this drug has still not been tested in GBMs. The present study aimed to evaluate the effects of E7070 on CA9 and CA12 enzymes in GBM cells as well as in the tumor cell growth, migration, invasion, and resistance to radiotherapy and chemotherapy. We found that E7070 treatment significantly reduced tumor cell growth and increased radio- and chemotherapy efficacy against GBM cells under hypoxia. Our data suggests that E7070 has therapeutic potential as a radio-chemo-sensitizing in drug-resistant GBMs, representing an attractive strategy to improve the adjuvant therapy. We showed that CA9 and CA12 represent potentially valuable therapeutic targets that should be further investigated as useful diagnostic and prognostic biomarkers for GBM tailored therapy.
Collapse
|
18
|
The complexity of tumour angiogenesis based on recently described molecules. Contemp Oncol (Pozn) 2021; 25:33-44. [PMID: 33911980 PMCID: PMC8063899 DOI: 10.5114/wo.2021.105075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Tumour angiogenesis is a crucial factor associated with tumour growth, progression, and metastasis. The whole process is the result of an interaction between a wide range of different molecules, influencing each other. Herein we summarize novel discoveries related to the less known angiogenic molecules such as galectins, pentraxin-3, Ral-interacting protein of 76 kDa (RLIP76), long non-coding RNAs (lncRNAs), B7-H3, and delta-like ligand-4 (DLL-4) and their role in the process of tumour angiogenesis. These molecules influence the most important molecular pathways involved in the formation of blood vessels in cancer, including the vascular endothelial growth factor (VEGF)-vascular endothelial growth factor receptor interaction (VEGFR), HIF1-a activation, or PI3K/Akt/mTOR and JAK-STAT signalling pathways. Increased expression of galectins, RLIP76, and B7H3 has been proven in several malignancies. Pentraxin-3, which appears to inhibit tumour angiogenesis, shows reduced expression in tumour tissues. Anti-angiogenic treatment based mainly on VEGF inhibition has proved to be of limited effectiveness, leading to the development of drug resistance. The newly discovered molecules are of great interest as a potential source of new anti-cancer therapies. Their role as targets for new drugs and as prognostic markers in neoplasms is discussed in this review.
Collapse
|
19
|
Parmar D, Apte M. Angiopoietin inhibitors: A review on targeting tumor angiogenesis. Eur J Pharmacol 2021; 899:174021. [PMID: 33741382 DOI: 10.1016/j.ejphar.2021.174021] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is the process of formation of new blood vessels from existing ones. Vessels serve the purpose of providing oxygen, nutrients and removal of waste from the cells. The physiological angiogenesis is a normal process and is required in the embryonic development, wound healing, menstrual cycle. For homeostasis, balance of pro angiogenic factors and anti angiogenic factors like is important. Their imbalance causes a process known as "angiogenic switch" which leads to various pathological conditions like inflammation, tumor and restenosis. Like normal cells, tumor cells also require oxygen and nutrients to grow which is provided by tumor angiogenesis. Hence angiogenic process can be inhibited to prevent tumor growth. This gives rise to study of anti angiogenic drugs. Currently approved anti angiogenic drugs are mostly VEGF inhibitors, but VEGF inhibitors have certain limitations like toxicity, low progression free survival (PFS), and resistance to anti VEGF therapy. This article focuses on angiopoietins as alternative and potential targets for anti angiogenic therapy. Angiopoietins are ligands of Tie receptor and play a crucial role in angiogenesis, their inhibition can prevent many tumor growths even on later stages of development. We present current clinical and preclinical stages of angiopoietin inhibitors. Drugs studied in the article are selective as well as non-selective inhibitors of angiopoietin 2 like Trebananib (AMG 386), AMG 780, REGN 910, CVX 060, MEDI 3617 and dual inhibitors of angiopoietin 2 and VEGF like Vanucizumab and RG7716. The angiopoietin inhibitors show promising results alone and in combination with VEGF inhibitors in various malignancies.
Collapse
Affiliation(s)
- Digna Parmar
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Maharashtra, India.
| | - Madhavi Apte
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Maharashtra, India.
| |
Collapse
|
20
|
The CXCL2/IL8/CXCR2 Pathway Is Relevant for Brain Tumor Malignancy and Endothelial Cell Function. Int J Mol Sci 2021; 22:ijms22052634. [PMID: 33807899 PMCID: PMC7961945 DOI: 10.3390/ijms22052634] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/30/2022] Open
Abstract
We aimed to evaluate the angiogenic capacity of CXCL2 and IL8 affecting human endothelial cells to clarify their potential role in glioblastoma (GBM) angiogenesis. Human GBM samples and controls were stained for proangiogenic factors. Survival curves and molecule correlations were obtained from the TCGA (The Cancer Genome Atlas) database. Moreover, proliferative, migratory and angiogenic activity of peripheral (HUVEC) and brain specific (HBMEC) primary human endothelial cells were investigated including blockage of CXCR2 signaling with SB225502. Gene expression analyses of angiogenic molecules from endothelial cells were performed. Overexpression of VEGF and CXCL2 was observed in GBM patients and associated with a survival disadvantage. Molecules of the VEGF pathway correlated but no relation for CXCR1/2 and CXCL2/IL8 was found. Interestingly, receptors of endothelial cells were not induced by addition of proangiogenic factors in vitro. Proliferation and migration of HUVEC were increased by VEGF, CXCL2 as well as IL8. Their sprouting was enhanced through VEGF and CXCL2, while IL8 showed no effect. In contrast, brain endothelial cells reacted to all proangiogenic molecules. Additionally, treatment with a CXCR2 antagonist led to reduced chemokinesis and sprouting of endothelial cells. We demonstrate the impact of CXCR2 signaling on endothelial cells supporting an impact of this pathway in angiogenesis of glioblastoma.
Collapse
|
21
|
Ali S, Borin TF, Piranlioglu R, Ara R, Lebedyeva I, Angara K, Achyut BR, Arbab AS, Rashid MH. Changes in the tumor microenvironment and outcome for TME-targeting therapy in glioblastoma: A pilot study. PLoS One 2021; 16:e0246646. [PMID: 33544755 PMCID: PMC7864405 DOI: 10.1371/journal.pone.0246646] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is a hypervascular and aggressive primary malignant tumor of the central nervous system. Recent investigations showed that traditional therapies along with antiangiogenic therapies failed due to the development of post-therapy resistance and recurrence. Previous investigations showed that there were changes in the cellular and metabolic compositions in the tumor microenvironment (TME). It can be said that tumor cell-directed therapies are ineffective and rethinking is needed how to treat GBM. It is hypothesized that the composition of TME-associated cells will be different based on the therapy and therapeutic agents, and TME-targeting therapy will be better to decrease recurrence and improve survival. Therefore, the purpose of this study is to determine the changes in the TME in respect of T-cell population, M1 and M2 macrophage polarization status, and MDSC population following different treatments in a syngeneic model of GBM. In addition to these parameters, tumor growth and survival were also studied following different treatments. The results showed that changes in the TME-associated cells were dependent on the therapeutic agents, and the TME-targeting therapy improved the survival of the GBM bearing animals. The current GBM therapies should be revisited to add agents to prevent the accumulation of bone marrow-derived cells in the TME or to prevent the effect of immune-suppressive myeloid cells in causing alternative neovascularization, the revival of glioma stem cells, and recurrence. Instead of concurrent therapy, a sequential strategy would be better to target TME-associated cells.
Collapse
Affiliation(s)
- Sehar Ali
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Thaiz F. Borin
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Raziye Piranlioglu
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Roxan Ara
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Iryna Lebedyeva
- Department of Chemistry and Physics, Augusta University, Augusta, Georgia, United States of America
| | - Kartik Angara
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Bhagelu R. Achyut
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| | - Ali Syed Arbab
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
- * E-mail: (ASA); (MHR)
| | - Mohammad H. Rashid
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail: (ASA); (MHR)
| |
Collapse
|
22
|
Hu C, Hui K, Jiang X. Effects of microRNA regulation on antiangiogenic therapy resistance in non-small cell lung cancer. Biomed Pharmacother 2020; 131:110557. [PMID: 32836072 DOI: 10.1016/j.biopha.2020.110557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 12/28/2022] Open
Abstract
Antiangiogenic drugs have become a standard therapeutic regimen for advanced non-small cell lung cancer (NSCLC); however, many issues remain to be solved. Identifying specific markers to predict patient response to antiangiogenic drugs to ensure therapeutic efficacy would increase their clinical benefit. MicroRNAs (miRNAs) are involved in the process of resistance to antiangiogenic therapy, as they regulate various key signaling pathways. Therefore, miRNAs may be used as targets for reversing tumor resistance to antiangiogenic therapy. This article reviews the molecular mechanisms of antiangiogenic therapy resistance and the specific mechanisms of miRNA regulation of resistance. Signal transducer and activator of transcription 3 (STAT3) is one of multiple target genes of miRNAs, and is closely related to antiangiogenic research. Thus, it is described separately in this review article.
Collapse
Affiliation(s)
- Chenxi Hu
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, No.182, Tongguan Road, Lianyungang City 222002, Jiangsu Province, China
| | - Kaiyuan Hui
- Department of Oncology, Lianyungang Clinical Medical College of Nanjing Medical University, No.182, Tongguan Road, Lianyungang City 222002, Jiangsu Province, China.
| | - Xiaodong Jiang
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, No.182, Tongguan Road, Lianyungang City 222002, Jiangsu Province, China; Department of Oncology, Lianyungang Clinical Medical College of Nanjing Medical University, No.182, Tongguan Road, Lianyungang City 222002, Jiangsu Province, China.
| |
Collapse
|
23
|
Targeting Glioblastoma: Advances in Drug Delivery and Novel Therapeutic Approaches. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000124] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Cai H, Liu W, Liu X, Li Z, Feng T, Xue Y, Liu Y. Advances and Prospects of Vasculogenic Mimicry in Glioma: A Potential New Therapeutic Target? Onco Targets Ther 2020; 13:4473-4483. [PMID: 32547078 PMCID: PMC7247597 DOI: 10.2147/ott.s247855] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
Abstract
Vasculogenic mimicry (VM) is the formation of a “vessel-like” structure without endothelial cells. VM exists in vascular-dependent solid tumors and is a special blood supply source involved in the highly invasive tumor progression. VM is observed in a variety of human malignant tumors and is closely related to tumor proliferation, invasion, and recurrence. Here, we review the mechanism, related signaling pathways, and molecular regulation of VM in glioma and discuss current research problems and the potential future applications of VM in glioma treatment. This review may provide a new viewpoint for glioma therapy.
Collapse
Affiliation(s)
- Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Wenjing Liu
- Department of Geriatrics, First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Zhiqing Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Tianda Feng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| |
Collapse
|
25
|
Xue W, Zhang J, Tong H, Xie T, Chen X, Zhou B, Wu P, Zhong P, Du X, Guo Y, Yang Y, Liu H, Fang J, Wang S, Wu H, Xu K, Zhang W. Effects of BMPER, CXCL10, and HOXA9 on Neovascularization During Early-Growth Stage of Primary High-Grade Glioma and Their Corresponding MRI Biomarkers. Front Oncol 2020; 10:711. [PMID: 32432046 PMCID: PMC7214627 DOI: 10.3389/fonc.2020.00711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/15/2020] [Indexed: 02/02/2023] Open
Abstract
Neovascularization is required in high-grade glioma (HGG). The objective of this study was to explore neovascularization-related genes and their corresponding MRI biomarkers during the early-growth stage of HGG. Tumor tissues from 30 HGG patients underwent perfusion MRI scanning prior to surgery were used to establish orthotopic xenograft models, pathologically analyze the tumor vasculature and perform transcriptome sequencing. The cases were divided into two groups based on whether the xenograft was successfully established. Microvascular density and BMPER, CXCL10, and HOXA9 expression of surgical specimens in the xenograft-forming group was significantly elevated and the microvascular diameter was significantly reduced, in vitro inhibition of BMPER, CXCL10, or HOXA9 in the glioma stem cell significantly suppressed its tube formation abilities. The in vivo experiment showed that BMPER was highly expressed in the early tumor growth phase (20 days), CXCL10 and HOXA9 expression was elevated with tumor progress, and spatially associated with tumor vasculature. Perfusion weighted MRI (PWI-MRI) derived parameters, rCBV, rCBF, Ktrans, and Vp, were also increased in the xenograft-forming group. In conclusion BMPER, CXCL10, and HOXA9 promote early tumor growth and progression by stimulating neovascularization of primary HGG. The rCBV, rCBF, Ktrans, and Vp can be used as imaging biomarkers to predict the expression statuses of these genes.
Collapse
Affiliation(s)
- Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Pengfei Wu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xuesong Du
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Youyuan Yang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Heng Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Wu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Kai Xu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Clinical Research Centre of Imaging and Nuclear Medicine, Chongqing, China
| |
Collapse
|
26
|
Zhu Y, Liu X, Zhao P, Zhao H, Gao W, Wang L. Celastrol Suppresses Glioma Vasculogenic Mimicry Formation and Angiogenesis by Blocking the PI3K/Akt/mTOR Signaling Pathway. Front Pharmacol 2020; 11:25. [PMID: 32116702 PMCID: PMC7025498 DOI: 10.3389/fphar.2020.00025] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis and vasculogenic mimicry (VM) are thought to be the predominant processes ensuring tumor blood supply during the growth and metastasis of glioblastoma (GBM). Celastrol has potential anti-glioma effects, however the mechanisms underlying these effects remain unclarified. Recent studies have shown that the PI3K/Akt/mTOR signaling pathway is closely related to angiogenesis and VM formation. In the present study, we have demonstrated, for the first time, that celastrol eliminated VM formation by blocking this signaling pathway in glioma cells. By the treatment of celastrol, tumor growth was suppressed, tight junction and basal lamina structures in tumor microvasculature were disarranged in U87 glioma orthotopic xenografts in nude mice. Periodic acid Schiff (PAS)-CD31 staining revealed that celastrol inhibited both VM and angiogenesis in tumor tissues. Additionally, celastrol reduced the expression levels of the angiogenesis-related proteins CD31, vascular endothelial growth factor receptor (VEGFR) 2, angiopoietin (Ang) 2 and VEGFA, VM-related proteins ephrin type-A receptor (EphA) 2, and vascular endothelial (VE)-cadherin. Hypoxia inducible factor (HIF)-1α, phosphorylated PI3K, Akt, and mTOR were also downregulated by treatment with celastrol. In vitro, we further demonstrated that celastrol inhibited the growth, migration, and invasion of U87 and U251 cells, disrupted VM formation, and blocked the activity of PI3K, Akt, and mTOR. Collectively, our data suggest that celastrol inhibits VM formation and angiogenesis likely by regulating the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yingjun Zhu
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Xihong Liu
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Henan, China
| | - Peiyuan Zhao
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Henan, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China.,School of Pharmaceutical Sciences, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Vengoji R, Ponnusamy MP, Rachagani S, Mahapatra S, Batra SK, Shonka N, Macha MA. Novel therapies hijack the blood-brain barrier to eradicate glioblastoma cancer stem cells. Carcinogenesis 2019; 40:2-14. [PMID: 30475990 DOI: 10.1093/carcin/bgy171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 10/12/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is amongst the most aggressive brain tumors with a dismal prognosis. Despite significant advances in the current multimodality therapy including surgery, postoperative radiotherapy (RT) and temozolomide (TMZ)-based concomitant and adjuvant chemotherapy (CT), tumor recurrence is nearly universal with poor patient outcomes. These limitations are in part due to poor drug penetration through the blood-brain barrier (BBB) and resistance to CT and RT by a small population of cancer cells recognized as tumor-initiating cells or cancer stem cells (CSCs). Though CT and RT kill the bulk of the tumor cells, they fail to affect CSCs, resulting in their enrichment and their development into more refractory tumors. Therefore, identifying the mechanisms of resistance and developing therapies that specifically target CSCs can improve response, prevent the development of refractory tumors and increase overall survival of GBM patients. Small molecule inhibitors that can breach the BBB and selectively target CSCs are emerging. In this review, we have summarized the recent advancements in understanding the GBM CSC-specific signaling pathways, the CSC-tumor microenvironment niche that contributes to CT and RT resistance and the use of novel combination therapies of small molecule inhibitors that may be used in conjunction with TMZ-based chemoradiation for effective management of GBM.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sidharth Mahapatra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
28
|
Ionizing radiation induces endothelial transdifferentiation of glioblastoma stem-like cells through the Tie2 signaling pathway. Cell Death Dis 2019; 10:816. [PMID: 31659157 PMCID: PMC6817826 DOI: 10.1038/s41419-019-2055-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022]
Abstract
Glioblastomas (GBM) are brain tumors with a poor prognosis despite treatment that combines surgical resection and radio-chemotherapy. These tumors are characterized by abundant vascularization and significant cellular heterogeneity including GBM stem-like cells (GSC) which contribute to tumor aggressiveness, resistance, and recurrence. Recent data has demonstrated that GSC are directly involved in the formation of new vessels via their transdifferentiation into Tumor Derived Endothelial Cells (TDEC). We postulate that cellular stress such as ionizing radiation (IR) could enhance the transdifferentiation of GSC into TDEC. GSC neurospheres isolated from 3 different patients were irradiated or not and were then transdifferentiated into TDEC. In fact, TDEC obtained from irradiated GSC (TDEC IR+) migrate more towards VEGF, form more pseudotubes in MatrigelTM in vitro and develop more functional blood vessels in MatrigelTM plugs implanted in Nude mice than TDEC obtained from non-irradiated GSC. Transcriptomic analysis allows us to highlight an overexpression of Tie2 in TDEC IR+. All IR-induced effects on TDEC were abolished by using a Tie2 kinase inhibitor, which confirms the role of the Tie2 signaling pathway in this process. Finally, by analyzing Tie2 expression in patient GBMs by immunohistochemistry, we demonstrated that the number of Tie2+ vessels increases in recurrent GBM compared with matched untreated tumors. In conclusion, we demonstrate that IR potentiates proangiogenic features of TDEC through the Tie2 signaling pathway, which indicates a new pathway of treatment-induced tumor adaptation. New therapeutic strategies that associate standard treatment and a Tie2 signaling pathway inhibitor should be considered for future trials.
Collapse
|
29
|
Neuroimaging findings of cerebral syphilitic gumma. Exp Ther Med 2019; 18:4185-4192. [PMID: 31772624 PMCID: PMC6861868 DOI: 10.3892/etm.2019.8089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
Cerebral syphilitic gumma is a rarely reported disease of the central nervous system. Magnetic resonance imaging (MRI) is an important diagnostic method for syphilitic gumma. The present study aimed to describe and characterize neuroimaging results from 6 patients with pathologically diagnosed cerebral syphilitic gumma. The 6 patients (age, 32–61 years) underwent brain CT and MRI, with 1 patient also undergoing whole-body 2-deoxy-2-(fluorine-18)fluoro-D-glucose-positron emission tomography/CT (18F-FDG PET/CT). Non-enhanced CT, conventional T1 weighted imaging (T1WI) and T2WI, diffusion weighted imaging (DWI) and gadolinium-enhanced T1WI images were acquired for all patients. The CT and MRI scans were retrospectively reviewed by two experienced radiologists for consensus on the location, number, size, T1WI, T2WI and DWI signal intensity characteristics, extent of vasogenic oedema, and enhancement patterns. In total, the 6 patients exhibited 10 lesions, nine of which were located in the cerebral hemisphere, primarily in the grey matter. The remaining lesion was located in the fourth ventricle, leading to mild-to-moderate hydrocephalus. The diameters of the identified 10 lesions ranged from 0.9–6.5 cm, with a mean diameter of 3.9 cm. The main feature observed in CT was low density and in MRI the features were T1WI and DWI hypointensity and T2WI hyperintensity. A single case exhibited syphilis gumma with massive haemorrhage. Ring-like or strip-like signs (n=5), accompanied by the dural tail sign (n=2) and homogeneous enhancement (n=1), were noted on T1WI with gadolinium. The 18F-FDG PET/CT performed in one patient of a cerebral syphilis gumma revealed low uptake and metabolism. The present study indicated that gadolinium-enhanced MRI combined with 18F-FDG PET/CT and laboratory examinations are helpful in distinguishing cerebral syphilitic gumma from brain tumors and infectious diseases, therefore avoiding unnecessary surgery.
Collapse
|
30
|
Yadav VN, Altshuler D, Kadiyala P, Zamler D, Comba A, Appelman H, Dunn P, Koschmann C, Castro MG, Löwenstein PR. Molecular ablation of tumor blood vessels inhibits therapeutic effects of radiation and bevacizumab. Neuro Oncol 2019; 20:1356-1367. [PMID: 29660022 DOI: 10.1093/neuonc/noy055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Glioblastoma (GBM) is an aggressive and highly vascular tumor with median survival below 2 years. Despite advances in surgery, radiotherapy, and chemotherapy, survival has improved modestly. To combat glioma vascular proliferation, anti-angiogenic agents targeting vascular endothelial growth factor (VEGF) were introduced. Preclinically these agents were effective, yet they did not improve overall survival in phase III trials. We tested the hypothesis that ganciclovir (GCV)-mediated killing of proliferating endothelial cells expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK) would have direct antitumor effects, and whether vessel ablation would affect the antitumor activity of anti-VEGF antibodies and radiotherapy. Methods Proliferating endothelial cells were eliminated using GCV-mediated killing of proliferating endothelial cells expressing HSV1-TK (in Tie2-TK-IRES-GFP mice). Syngeneic NRAS/p53 (NP) gliomas were implanted into the brains of Tie2-TK-IRES-GFP mice. Endothelial proliferation activates the Tie2 promoter and HSV1-TK expression. Administration of GCV kills proliferating tumor endothelial cells and slows tumor growth. The effects of endothelial cell ablation on anti-angiogenic therapy were examined using anti-VEGF antibodies or irradiation. Results GCV administration reduced tumor growth and vascular density, increased tumor apoptosis, and prolonged survival. Anti-VEGF antibodies or irradiation also prolonged survival. Surprisingly, combining GCV with irradiation, or with anti-VEGF antibodies, reduced their individual therapeutic effects. Conclusion GCV-mediated killing of proliferating endothelial cells expressing HSV1-TK, anti-VEGF antibodies, or irradiation all reduced growth of a murine glioma. However, elimination of microvascular proliferation decreased the efficacy of anti-VEGF or irradiation therapy. We conclude that, in our model, the integrity of proliferating vessels is necessary for the antiglioma effects of anti-VEGF and radiation therapy.
Collapse
Affiliation(s)
- Viveka Nand Yadav
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - David Altshuler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel Zamler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Henry Appelman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Patrick Dunn
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Carl Koschmann
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Pediatric Hematology/Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Pedro R Löwenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
31
|
Single-agent Bevacizumab in Recurrent Glioblastoma After Second-line Chemotherapy With Fotemustine: The Experience of the Italian Association of Neuro-Oncology. Am J Clin Oncol 2019; 41:1272-1275. [PMID: 29782366 DOI: 10.1097/coc.0000000000000464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Bevacizumab is an anti-vascular endothelial growth factor antibody used in the treatment of recurrent glioblastoma (GBM). Despite the large number of studies carried out in patients with recurrent GBM, little is known about the administration of this angiogenesis inhibitor after the failure of the second-line chemotherapy. MATERIALS AND METHODS In this retrospective multicenter study, on behalf of the Italian Association of Neuro-Oncology, we reported the results obtained in 51 patients with recurrent GBM treated with single-agent bevacizumab after the failure of second-line chemotherapy with fotemustine. RESULTS In March 2016, at the time of data analysis, 3 patients (14.4%) were still alive with stable disease, whereas 48 died due to disease progression. Kaplan-Meier estimated median survival from the diagnosis of GBM was 28 months (95% confidence interval [CI], 22.1-33.9 mo). Median survival measured from the beginning of fotemustine and bevacizumab therapy were 11.3 (95% CI, 8.4-13.6 mo) and 6 months (95% CI, 3.8-8.1 mo), respectively. The 6- and 12-month progression free survival rates from the beginning of bevacizumab treatment were 18% and 13%, respectively. CONCLUSIONS On the basis of our data, in patients with recurrent GBM, the failure of a second-line chemotherapy with cytotoxic agents might not exclude the administration of bevacizumab as third-line chemotherapy.
Collapse
|
32
|
Zeng Y, Yao X, Liu X, He X, Li L, Liu X, Yan Z, Wu J, Fu BM. Anti-angiogenesis triggers exosomes release from endothelial cells to promote tumor vasculogenesis. J Extracell Vesicles 2019; 8:1629865. [PMID: 31258881 PMCID: PMC6586113 DOI: 10.1080/20013078.2019.1629865] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 02/05/2023] Open
Abstract
Although anti-angiogenic therapies (AATs) have some effects against multiple malignancies, they are limited by subsequent tumor vasculogenesis and progression. To investigate the mechanisms by which tumor vasculogenesis and progression following AATs, we transfected microRNA (miR)-9 into human umbilical vein endothelial cells (HUVECs) to mimic the tumor-associated endothelial cells in hepatocellular carcinoma and simulated the AATs in vitro and in vivo. We found that administration of the angiogenesis inhibitor vandetanib completely abolished miR-9-induced angiogenesis and promoted autophagy in HUVECs, but induced the release of vascular endothelial growth factor (VEGF)-enriched exosomes. These VEGF-enriched exosomes significantly promoted the formation of endothelial vessels and vasculogenic mimicry in hepatocellular carcinoma and its progression in mice. Anti-autophagic therapy is proposed to improve the efficacy of AATs. However, similar effects by AATs were observed with the application of anti-autophagy by 3-methyladenine. Our results revealed that tumor vasculogenesis and progression after AATs and anti-autophagic therapies were due to the cross-talk between endothelial and tumor cells via VEGF-enriched exosomes.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xinghong Yao
- Radiation Therapy Center, Sichuan Cancer Hospital and Institute, Chengdu, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xueling He
- Laboratory Animal Center, Sichuan University, Chengdu, China
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiping Yan
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiang Wu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Bingmei M Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY, USA
| |
Collapse
|
33
|
Hu Q, Liu F, Yan T, Wu M, Ye M, Shi G, Lv S, Zhu X. MicroRNA‑576‑3p inhibits the migration and proangiogenic abilities of hypoxia‑treated glioma cells through hypoxia‑inducible factor‑1α. Int J Mol Med 2019; 43:2387-2397. [PMID: 31017266 PMCID: PMC6488173 DOI: 10.3892/ijmm.2019.4157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
The most common and aggressive type of brain cancer in adults is glioblastoma multiforme (GBM), and hypoxia is a common feature of glioblastoma. As the histological features of glioma include capillary endothelial cell proliferation, they are highly prone to invading the surrounding normal brain tissue, which is often one of the reasons for the failure of treatment. However, the mechanisms involved in this process are not fully understood. MicroRNAs (miRs) are a class of non‑coding RNA that are able to inhibit the malignant progression of tumor cells through the regulation of downstream genes. In the present study, the low expression of miR‑576‑3p was detected in glioma samples and hypoxia‑treated glioma cells using a reverse transcription‑quantitative polymerase chain reaction. The present study focused on the effects of miR‑576‑3p on hypoxia‑induced glioma. The results of the functional experiments revealed that the overexpression of miR‑576‑3p significantly inhibited the migration and pro‑angiogenic abilities of the glioma cells under hypoxic conditions (P<0.05) compared with in the lentivirus‑miR‑negative control group. Furthermore, luciferase reporter gene assays were used to validate the hypothesis that miR‑576‑3p interacts with the 3'‑untranslated region of hypoxia‑inducible factor‑1α (HIF‑1α) and induces a reduction in the protein levels of matrix metalloproteinase‑2 and vascular endothelial growth factor. Rescue experiments demonstrated that the restoration of HIF‑1α expression attenuated the effect of miR‑576‑3p on the migration and proangiogenic abilities of glioma cells. In conclusion, the present study confirms that miR‑576‑3p is a novel GBM inhibitor and its inhibition of the migration and proangiogenic capacity of hypoxia‑induced glioma cells is mediated by HIF‑1α.
Collapse
Affiliation(s)
- Qing Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Feng Liu
- Department of Neurosurgery, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi 330006
| | - Tengfeng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Miaojing Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Minhua Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Guangyao Shi
- Queen Mary School, Medical College, Nanchang University School of Medicine, Nanchang, Jiangxi 330031, P.R. China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| |
Collapse
|
34
|
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: 5.8] [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
|
35
|
Senchukova MA, Makarova EV, Kalinin EA, Tkachev VV. Modern ideas about the origin, features of morphology, prognostic and predictive significance of tumor vessels. RUSSIAN JOURNAL OF BIOTHERAPY 2019; 18:6-15. [DOI: 10.17650/1726-9784-2019-18-1-6-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The review presents modern ideas about the origin of tumor vessels and the features of their morphology. The various approaches to the classification of tumor vessel types and to the assessment of their clinical and prognostic significance are described. Also, the main problems associated with the use of angiogenesis blockers in the treatment of malignancies and their possible solutions are reflected in the review.
Collapse
Affiliation(s)
- M. A. Senchukova
- Orenburg State Medical University of the Ministry of Health of the Russian Federation; Orenburg Regional Clinical Oncology Dispensary
| | - E. V. Makarova
- Orenburg State Medical University of the Ministry of Health of the Russian Federation; Orenburg Regional Clinical Oncology Dispensary
| | | | | |
Collapse
|
36
|
Cenciarini M, Valentino M, Belia S, Sforna L, Rosa P, Ronchetti S, D'Adamo MC, Pessia M. Dexamethasone in Glioblastoma Multiforme Therapy: Mechanisms and Controversies. Front Mol Neurosci 2019; 12:65. [PMID: 30983966 PMCID: PMC6449729 DOI: 10.3389/fnmol.2019.00065] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/26/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant of the glial tumors. The world-wide estimates of new cases and deaths annually are remarkable, making GBM a crucial public health issue. Despite the combination of radical surgery, radio and chemotherapy prognosis is extremely poor (median survival is approximately 1 year). Thus, current therapeutic interventions are highly unsatisfactory. For many years, GBM-induced brain oedema and inflammation have been widely treated with dexamethasone (DEX), a synthetic glucocorticoid (GC). A number of studies have reported that DEX also inhibits GBM cell proliferation and migration. Nevertheless, recent controversial results provided by different laboratories have challenged the widely accepted dogma concerning DEX therapy for GBM. Here, we have reviewed the main clinical features and genetic and epigenetic abnormalities underlying GBM. Finally, we analyzed current notions and concerns related to DEX effects on cerebral oedema, cancer cell proliferation and migration and clinical outcome.
Collapse
Affiliation(s)
- Marta Cenciarini
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Simona Ronchetti
- Section of Pharmacology, Department of Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Mauro Pessia
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy.,Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| |
Collapse
|
37
|
Abstract
Leucurogin is an ECD disintegrin-like protein, cloned from Bothrops leucurus venom gland. This new protein, encompassing the disintegrin region of a PIII metalloproteinase, is produced by recombinant technology and its biological and functional activity was partially characterized in this study. Biological activity was characterized in vitro using human fibroblasts. Functional activity of leucurogin was analysed in vitro and in vivo with murine B16F10 Nex-2 and human melanoma BLM cells. The results show that leucurogin inhibits cellular processes dependent on collagen type I. In a competition assay with collagen, leucurogin inhibits, in a dose-dependent manner, the adhesion of fibroblast to collagen. At 10 μM leucurogin reduces adhesion (40%) and migration (70%) of hFb and inhibits migration (32%) and proliferation (65%) of BLM cells. At 2.5 μM leucurogin inhibits 80% cell proliferation of B16F10 Nex-2 melanoma cells. At 4.8 μM leucurogin inhibits, in vitro, the vascular structures formation by endothelial cells by 66%. Leucurogin, injected intraperitoneally, i.p. (5 μg/animal, two-month old C57/Bl6 male mice) on alternate days for 15 days, inhibits lung metastasis of B16F10 Nex-2 cells by 70-75%. In the treatment of human melanoma, grafted intradermally in the nude mice flank, leucurogin (7.5 μg/kg in alternate days during 17 days) inhibits tumor growth by more than 40%. Leucurogin can be considered a promising agent for melanoma treatment.
Collapse
|
38
|
Song J, Huang S, Wang K, Li W, Pao L, Chen F, Zhao X. Long Non-coding RNA MEG3 Attenuates the Angiotensin II-Induced Injury of Human Umbilical Vein Endothelial Cells by Interacting With p53. Front Genet 2019; 10:78. [PMID: 30838022 PMCID: PMC6389612 DOI: 10.3389/fgene.2019.00078] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 01/28/2019] [Indexed: 01/01/2023] Open
Abstract
Angiotensin II (Ang II)-induced damage to endothelial cells (ECs) plays a crucial role in the pathogenesis of cardiovascular disease. This study aimed to investigate the role of maternally expressed gene 3 (Meg3) in endothelial cell injury. A lncRNA human gene expression microarray analysis was used to identify differentially expressed lncRNAs in human umbilical vein endothelial cell (HUVECs). Cell viability, apoptosis, and migration were then assessed Ang II-treated HUVECs. qRT-PCR and western blotting were performed to detect the expression level of p53 after Meg3 knockdown and overexpression. We observed that Ang II treatment decreased the Meg3 level in HUVECs. Next, both knockdown of Meg3 and Ang II decreased cell viability, increased apoptotic cell rate and impair migration function in HUVECs. Furthermore, overexpression of Meg3 inhibited cell apoptosis, and increased cell migration by enhancing p53 transcription on its target genes, including CRP, ICAM-1, VEGF, and HIF-1α. Our findings indicate that Meg3 might be associated with cardiovascular disease development.
Collapse
Affiliation(s)
- Jingwen Song
- Department of Cardiovascularology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Songqun Huang
- Department of Cardiovascularology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kaizhong Wang
- Department of Cardiovascularology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wei Li
- Institute of Tumor, Second Military Medical University, Shanghai, China
| | - Lizhi Pao
- Department of Cardiovascularology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Feng Chen
- Department of Cardiovascularology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xianxian Zhao
- Department of Cardiovascularology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
39
|
CXCR2-Expressing Tumor Cells Drive Vascular Mimicry in Antiangiogenic Therapy-Resistant Glioblastoma. Neoplasia 2019; 20:1070-1082. [PMID: 30236892 PMCID: PMC6151844 DOI: 10.1016/j.neo.2018.08.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND: Glioblastoma (GBM) was shown to relapse faster and displayed therapeutic resistance to antiangiogenic therapies (AATs) through an alternative tumor cell-driven mechanism of neovascularization called vascular mimicry (VM). We identified highly upregulated interleukin 8 (IL-8)-CXCR2 axis in tumor cells in high-grade human glioma and AAT-treated orthotopic GBM tumors. METHODS: Human GBM tissue sections and tissue array were used to ascertain the clinical relevance of CXCR2-positive tumor cells in the formation of VM. We utilized U251 and U87 human tumor cells to understand VM in an orthotopic GBM model and AAT-mediated enhancement in VM was modeled using vatalanib (anti-VEGFR2) and avastin (anti-VEGF). Later, VM was inhibited by SB225002 (CXCR2 inhibitor) in a preclinical study. RESULTS: Overexpression of IL8 and CXCR2 in human datasets and histological analysis was identified as a bonafide candidate to validate VM through in vitro and animal model studies. AAT-treated tumors displayed a higher number of CXCR2-positive GBM-stem cells with endothelial-like phenotypes. Stable knockdown of CXCR2 expression in tumor cells led to decreased tumor growth as well as incomplete VM structures in the animal models. Similar data were obtained following SB225002 treatment. CONCLUSIONS: The present study suggests that tumor cell autonomous IL-8-CXCR2 pathway is instrumental in AAT-mediated resistance and VM formation in GBM. Therefore, CXCR2 can be targeted through SB225002 and can be combined with standard therapies to improve the therapeutic outcomes in clinical trials.
Collapse
|
40
|
Mahdi A, Darvishi B, Majidzadeh-A K, Salehi M, Farahmand L. Challenges facing antiangiogenesis therapy: The significant role of hypoxia-inducible factor and MET in development of resistance to anti-vascular endothelial growth factor-targeted therapies. J Cell Physiol 2018; 234:5655-5663. [PMID: 30515806 DOI: 10.1002/jcp.27414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023]
Abstract
It is now fully recognized that along with multiple physiological functions, angiogenesis is also involved in the fundamental process and pathobiology of several disorders including cancer. Recent studies have fully established the role of angiogenesis in cancer progression as well as invasion and metastasis. Consequently, many therapeutic agents such as monoclonal antibodies targeting angiogenesis pathway have been introduced in clinic with the hope for improving the outcomes of cancer therapy. Bevacizumab (Avastin®) was the first anti-vascular endothelial growth factor (VEGF) targeting monoclonal antibody developed with this purpose and soon received its accelerated US Food and Drug Administration (FDA) approval for treatment of patients with metastatic breast cancer in 2008. However, the failure to meet expecting results in different follow-up studies, forced FDA to remove bevacizumab approval for metastatic breast cancer. Investigations have now revealed that while suppressing VEGF pathway initially decreases tumor progression rate and vasculature density, activation of several interrelated pathways and signaling molecules following VEGF blockade compensate the insufficiency of VEGF and initially blocked angiogenesis, explaining in part the failure observed with bevacizumab single therapy. In present review, we introduce some of the main pathways and signaling molecules involved in angiogenesis and then propose how their interconnection may result in development of resistance to bevacizumab.
Collapse
Affiliation(s)
- Ali Mahdi
- Department of Recombinant Proteins, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Behrad Darvishi
- Department of Recombinant Proteins, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Department of Recombinant Proteins, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.,Tasnim Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Malihe Salehi
- Department of Recombinant Proteins, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Department of Recombinant Proteins, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
41
|
Thijssen VLJL, Paulis YWJ, Nowak‐Sliwinska P, Deumelandt KL, Hosaka K, Soetekouw PMMB, Cimpean AM, Raica M, Pauwels P, van den Oord JJ, Tjan‐Heijnen VCG, Hendrix MJ, Heldin C, Cao Y, Griffioen AW. Targeting PDGF-mediated recruitment of pericytes blocks vascular mimicry and tumor growth. J Pathol 2018; 246:447-458. [PMID: 30101525 PMCID: PMC6587443 DOI: 10.1002/path.5152] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/12/2018] [Accepted: 08/07/2018] [Indexed: 12/28/2022]
Abstract
Aggressive tumor cells can adopt an endothelial cell-like phenotype and contribute to the formation of a tumor vasculature, independent of tumor angiogenesis. This adoptive mechanism is referred to as vascular mimicry and it is associated with poor survival in cancer patients. To what extent tumor cells capable of vascular mimicry phenocopy the angiogenic cascade is still poorly explored. Here, we identify pericytes as important players in vascular mimicry. We found that pericytes are recruited by vascular mimicry-positive tumor cells in order to facilitate sprouting and to provide structural support of the vascular-like networks. The pericyte recruitment is mediated through platelet-derived growth factor (PDGF)-B. Consequently, preventing PDGF-B signaling by blocking the PDGF receptors with either the small tyrosine kinase inhibitor imatinib or blocking antibodies inhibits vascular mimicry and tumor growth. Collectively, the current study identifies an important role for pericytes in the formation of vascular-like structures by tumor cells. Moreover, the mechanism that controls the pericyte recruitment provides therapeutic opportunities for patients with aggressive vascular mimicry-positive cancer types. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Victor LJL Thijssen
- Angiogenesis Laboratory, Department of Medical OncologyVU University Medical CenterAmsterdamThe Netherlands
- Department of Radiation OncologyVU University Medical CenterAmsterdamThe Netherlands
| | - Yvette WJ Paulis
- Angiogenesis Laboratory, Department of Medical OncologyVU University Medical CenterAmsterdamThe Netherlands
- Division of Medical Oncology, GROW – School for Oncology and Developmental BiologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Patrycja Nowak‐Sliwinska
- Angiogenesis Laboratory, Department of Medical OncologyVU University Medical CenterAmsterdamThe Netherlands
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
| | - Katrin L Deumelandt
- Angiogenesis Laboratory, Department of Medical OncologyVU University Medical CenterAmsterdamThe Netherlands
| | - Kayoko Hosaka
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstituteStockholmSweden
| | - Patricia MMB Soetekouw
- Division of Medical Oncology, GROW – School for Oncology and Developmental BiologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Anca M Cimpean
- Department of Microscopic Morphology, Histology, Angiogenesis Research CenterVictor Babes University of Medicine and PharmacyTimisoaraRomania
| | - Marius Raica
- Department of Microscopic Morphology, Histology, Angiogenesis Research CenterVictor Babes University of Medicine and PharmacyTimisoaraRomania
| | - Patrick Pauwels
- Department of PathologyAntwerp University HospitalEdegemBelgium
| | - Joost J van den Oord
- Laboratory of Translational Cell and Tissue ResearchUniversity of LeuvenLeuvenBelgium
| | - Vivianne CG Tjan‐Heijnen
- Division of Medical Oncology, GROW – School for Oncology and Developmental BiologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Mary J Hendrix
- Department of Biology, Shepherd UniversityShepherdstown UniversityWVUSA
| | - Carl‐Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life LaboratoryUppsala UniversityUppsalaSweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstituteStockholmSweden
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical OncologyVU University Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
42
|
InVivo Molecular Ultrasound Assessment of Glioblastoma Neovasculature with Endoglin-Targeted Microbubbles. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8425495. [PMID: 30498402 PMCID: PMC6220748 DOI: 10.1155/2018/8425495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/16/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022]
Abstract
Objectives Glioblastoma, as one of the most malignant cancer in the world, usually shows substantially increased angiogenesis. Endoglin (CD105), which is an alternative proangiogenic growth factor, has been remarkably upregulated on the proliferating glioblastoma neovasculature. However, little is known on the noninvasive assessment of the expression levels of CD105 during glioblastoma progression. Herein, we investigated the potential of the molecular ultrasound imaging for the noninvasive assessment of the expression levels of the biomarker CD105 during the glioblastoma progression. Materials and Methods The CD105-targeted perfluorocarbon-containing lipid-shelled microbubbles (MBs) were prepared. A parallel flow chamber was employed, in which the CD105-targeted and non-targeted MBs were tested across the CD105 ± expression cell lines. In vivo molecular US imaging was conducted based on a subcutaneous xenograft tumor model (n=9). Finally, the statistical analysis was conducted to quantitatively correlate the attachment numbers of MBs in the parallel flow chamber test with the CD105 expression levels of the cells in the flow cytometry test and the in vivo molecular ultrasound signals with the ex vivo expression levels of CD105 in the immunohistochemical test. Results and Discussion The attachment numbers of the CD105-targeted MBs significantly correlated with the CD105 expression levels of the cells in the parallel flow chamber test. There was a good correlation between the in vivo molecular ultrasound signals with the CD105-targeted MBs and the ex vivo expression levels of CD105 in the immunohistochemical test. The results indicate that the molecular US imaging is much potential to assess the progression of the glioblastoma neovasculature noninvasively.
Collapse
|
43
|
Erices JI, Torres Á, Niechi I, Bernales I, Quezada C. Current natural therapies in the treatment against glioblastoma. Phytother Res 2018; 32:2191-2201. [PMID: 30109743 DOI: 10.1002/ptr.6170] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/08/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor, which causes the highest number of deaths worldwide. It is a highly vascularized tumor, infiltrative, and its tumorigenic capacity is exacerbated. All these hallmarks are therapeutic targets in GBM treatment, including surgical removal followed by radiotherapy and chemotherapy. Current therapies have not been sufficient for the effective patient's management, so the classic therapies have had to expand and incorporate new alternative treatments, including natural compounds. This review summarizes natural products and their physiological effects in in vitro and in vivo models of GBM, specifically by modulating signaling pathways involved in angiogenesis, cell migration/invasion, cell viability, apoptosis, and chemoresistance. The most important aspects of natural products and their derivatives were described in relation to its antitumoral effects. As a final result, it can be obtained that within the compounds with more evidence that supports or suggests its clinical use are the cannabinoids, terpenes, and curcumin, because many have been shown to have a significant effect in decreasing the progress of GBM through known mechanisms, such as chemo-sensitization or decrease migration and cell invasion. Natural compounds emerge as promising therapies to attack the progress of GBM.
Collapse
Affiliation(s)
- José Ignacio Erices
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Ángelo Torres
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Isabel Bernales
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Quezada
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, Chile
| |
Collapse
|
44
|
Dalley CB, Wroblewska B, Wolfe BB, Wroblewski JT. The Role of Metabotropic Glutamate Receptor 1 Dependent Signaling in Glioma Viability. J Pharmacol Exp Ther 2018; 367:59-70. [PMID: 30054311 DOI: 10.1124/jpet.118.250159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 12/31/2022] Open
Abstract
Glioma refers to malignant central nervous system tumors that have histologic characteristics in common with glial cells. The most prevalent type, glioblastoma multiforme, is associated with a poor prognosis and few treatment options. On the basis of reports of aberrant expression of mGluR1 mRNA in glioma, evidence that melanoma growth is directly influenced by glutamate metabotropic receptor 1 (mGluR1), and characterization of β-arrestin-dependent prosurvival signaling by this receptor, this study investigated the hypothesis that glioma cell lines aberrantly express mGluR1 and depend on mGluR1-mediated signaling to maintain viability and proliferation. Three glioma cell lines (Hs683, A172, and U87) were tested to confirm mGluR1 mRNA expression and the dependence of glioma cell viability on glutamate. Pharmacologic and genetic evidence is presented that suggests mGluR1 signaling specifically supports glioma proliferation and viability. For example, selective noncompetitive antagonists of mGluR1, CPCCOEt and JNJ16259685, decreased the viability of these cells in a dose-dependent manner, and glutamate metabotropic receptor 1 gene silencing significantly reduced glioma cell proliferation. Also, results of an anchorage-independent growth assay suggested that noncompetitive antagonism of mGluR1 may decrease the tumorigenic potential of Hs683 glioma cells. Finally, data are provided that support the hypothesis that a β-arrestin-dependent signaling cascade may be involved in glutamate-stimulated viability in glioma cells and that ligand bias may exist at mGluR1 expressed in these cells. Taken together, the results strongly suggest that mGluR1 may act as a proto-oncogene in glioma and be a viable drug target in glioma treatment.
Collapse
Affiliation(s)
- Carrie Bowman Dalley
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| | - Barbara Wroblewska
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| | - Barry B Wolfe
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| | - Jarda T Wroblewski
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
45
|
Zheng LC, Wang XQ, Lu K, Deng XL, Zhang CW, Luo H, Xu XD, Chen XM, Yan L, Wang YQ, Shi SL. Ephrin-B2/Fc promotes proliferation and migration, and suppresses apoptosis in human umbilical vein endothelial cells. Oncotarget 2018; 8:41348-41363. [PMID: 28489586 PMCID: PMC5522204 DOI: 10.18632/oncotarget.17298] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/03/2017] [Indexed: 01/12/2023] Open
Abstract
Tumor growth and metastasis are angiogenesis dependent. Angiogenic growth involves endothelial cell proliferation, migration, and invasion. Ephrin-B2 is a ligand for Eph receptor tyrosine kinases and is an important mediator in vascular endothelial growth factor-mediated angiogenesis. However, research offer controversial information regarding effects of ephrin-B2 on vascular endothelial cells. In this paper, proteome analyses showed that ephrin-B2/Fc significantly activates multiple signaling pathways related to cell proliferation, survival, and migration and suppresses apoptosis and cell death. Cytological experiments further confirm that ephrin-B2/Fc stimulates endothelial cell proliferation, triggers dose-dependent migration, and suppresses cell apoptosis. Results demonstrate that soluble dose-dependent ephrinB2 can promote proliferation and migration and inhibit apoptosis of human umbilical vein endothelial cells. These results also suggest that ephrinB2 prevents ischemic disease and can potentially be a new therapeutic target for treating angiogenesis-related diseases and tumors.
Collapse
Affiliation(s)
- Li-Chun Zheng
- Medical College of Xiamen University, Jinshan Community Health Service Center, Xiamen Traditional Chinese Medical Hospital, Xiamen 361000, P.R. China.,Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xiao-Qing Wang
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Kun Lu
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| | - Xiao-Ling Deng
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| | - Cheng-Wei Zhang
- Department of Cardiology, Affiliated Dongnan Hospital of Xiamen University, Zhangzhou 363000, P.R. China
| | - Hong Luo
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xu-Dong Xu
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xiao-Man Chen
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Lu Yan
- Department of Basic Medicine, Medical College of Xiamen University, Xiamen 361102, P.R. China
| | - Yi-Qing Wang
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Song-Lin Shi
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| |
Collapse
|
46
|
Quiros-Gonzalez I, Tomaszewski MR, Aitken SJ, Ansel-Bollepalli L, McDuffus LA, Gill M, Hacker L, Brunker J, Bohndiek SE. Optoacoustics delineates murine breast cancer models displaying angiogenesis and vascular mimicry. Br J Cancer 2018; 118:1098-1106. [PMID: 29576623 PMCID: PMC5931091 DOI: 10.1038/s41416-018-0033-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Optoacoustic tomography (OT) of breast tumour oxygenation is a promising new technique, currently in clinical trials, which may help to determine disease stage and therapeutic response. However, the ability of OT to distinguish breast tumours displaying different vascular characteristics has yet to be established. The aim of the study is to prove OT as a sensitive technique for differentiating breast tumour models with manifestly different vasculatures. METHODS Multispectral OT (MSOT) was performed in oestrogen-dependent (MCF-7) and oestrogen-independent (MDA-MB-231) orthotopic breast cancer xenografts. Total haemoglobin (THb) and oxygen saturation (SO2MSOT) were calculated. Pathological and biochemical evaluation of the tumour vascular phenotype was performed for validation. RESULTS MCF-7 tumours show SO2MSOT similar to healthy tissue in both rim and core, despite significantly lower THb in the core. MDA-MB-231 tumours show markedly lower SO2MSOT with a significant rim-core disparity. Ex vivo analysis revealed that MCF-7 tumours contain fewer blood vessels (CD31+) that are more mature (CD31+/aSMA+) than MDA-MB-231. MCF-7 presented higher levels of stromal VEGF and iNOS, with increased NO serum levels. The vasculogenic process observed in MCF-7 was consistent with angiogenesis, while MDA-MB-231 appeared to rely more on vascular mimicry. CONCLUSIONS OT is sensitive to differences in the vascular phenotypes of our breast cancer models.
Collapse
Affiliation(s)
- Isabel Quiros-Gonzalez
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Michal R Tomaszewski
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Sarah J Aitken
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Laura Ansel-Bollepalli
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Leigh-Ann McDuffus
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Michael Gill
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Lina Hacker
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Joanna Brunker
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
47
|
Tang W, Chen Y, Wang X, Chen Y, Zhang J, Lin Z. Expression of CXC-motif-chemokine 12 and the receptor C-X-C receptor 4 in glioma and theeffect on peritumoral brain edema. Oncol Lett 2018; 15:2501-2507. [PMID: 29434965 DOI: 10.3892/ol.2017.7547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
The present study aimed to evaluate the association between CXC-motif-chemokine 12 (CXCL12)/C-X-C receptor 4 (CXCR4) expression and peritumoral brain edema (PTBE) in glioma patients. Immunohistochemical techniques were used to detect the expression of CXCR4 and CXCL12 in 58 glioma tissues. Magnetic resonance imaging was used to evaluate the extent and type of brain edema in preoperative glioma patients. The association between edema and CXCL12/CXCR4 expression was examined by χ2 analysis. The prognostic significance of CXCL12 or CXCR4 was determined by log-rank tests and Cox's proportional hazards model. Expression of CXCL12 and CXCR4 was observed in vascular endothelial cells and tumor cells. The degree (P=0.033) and morphology (P=0.033) of PTBE were significantly associated with the level of CXCL12 expression in vascular endothelial cells. The degree (P=0.001) and morphology (P=0.001) of PTBE were associated with the level of CXCR4 expression in tumor cells. CXCR4-positive vascular endothelial cells were significantly associated only with the degree of edema (P=0.030). Therefore, the present study indicated that levels of CXCL12 expression in vascular endothelial cells and CXCR4 expression in tumor cells are associated with PTBE.
Collapse
Affiliation(s)
- Wenlong Tang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yupeng Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Xingfu Wang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yao Chen
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Jiandong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Zhixiong Lin
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| |
Collapse
|
48
|
Abstract
Resistance to chemotherapy and cancer relapse are major clinical challenges attributed to a sub population of cancer stem cells (CSCs). The concept of CSCs has been the subject of intense research by the oncology community since evidence for their existence was first published over twenty years ago. Emerging data indicates that they are also able to evade novel therapies such as targeted agents, immunotherapies and anti-angiogenics. The inability to appropriately identify and isolate CSCs is a major hindrance to the field and novel technologies are now being utilized. Agents that target CSC-associated cell surface receptors and signaling pathways have generated promising pre-clinical results and are now entering clinical trial. Here we discuss and evaluate current therapeutic strategies to target CSCs.
Collapse
Affiliation(s)
- Stephanie Annett
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland
| | - Tracy Robson
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland.
| |
Collapse
|
49
|
Arnone GD, Bhimani AD, Aguilar T, Mehta AI. Localized targeted antiangiogenic drug delivery for glioblastoma. J Neurooncol 2018; 137:223-231. [DOI: 10.1007/s11060-018-2747-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
|
50
|
Xue W, Du X, Wu H, Liu H, Xie T, Tong H, Chen X, Guo Y, Zhang W. Aberrant glioblastoma neovascularization patterns and their correlation with DCE-MRI-derived parameters following temozolomide and bevacizumab treatment. Sci Rep 2017; 7:13894. [PMID: 29066764 PMCID: PMC5654943 DOI: 10.1038/s41598-017-14341-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 10/10/2017] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a highly angiogenic malignancy, and its abundant, aberrant neovascularization is closely related to the proliferation and invasion of tumor cells. However, anti-angiogenesis combined with standard radio-/chemo-therapy produces little improvement in treatment outcomes. Determining the reason for treatment failure is pivotal for GBM treatment. Here, histopathological analysis and dynamic contrast-enhanced MRI (DCE-MRI) were used to explore the effects of temozolomide (TMZ) and bevacizumab (BEV) on GBM neovascularization patterns in an orthotopic U87MG mouse model at 1, 3 and 6 days after treatment. We found that the amount of vascular mimicry (VM) significantly increased 6 days after BEV treatment. TMZ inhibited neovascularization at an early stage, but the microvessel density (MVD) and transfer coefficient (Ktrans) derived from DCE-MRI increased 6 days after treatment. TMZ and BEV combination therapy slightly prolonged the inhibitory effect on tumor microvessels. Sprouting angiogenesis was positively correlated with Ktrans in all treatment groups. The increase in VM after BEV administration and the increase in MVD and Ktrans after TMZ administration may be responsible for treatment resistance. Ktrans holds great potential as an imaging biomarker for indicating the variation in sprouting angiogenesis during drug treatment for GBM.
Collapse
Affiliation(s)
- Wei Xue
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xuesong Du
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Hao Wu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Tian Xie
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yu Guo
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, 400042, China.
| |
Collapse
|