1
|
Ke C, Chen C, Yang M, Chen H, Ke Y, Li L. Inhibition of infantile hemangioma growth and promotion of apoptosis via VEGF/PI3K/Akt axis by 755-nm long-pulse alexandrite laser. Biomed J 2024; 47:100675. [PMID: 37944864 PMCID: PMC11340587 DOI: 10.1016/j.bj.2023.100675] [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: 08/20/2023] [Revised: 09/28/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Infantile hemangioma (IH) is a common vascular tumor in female infants, which can lead to aesthetic issues and facial scarring. This study aimed to investigate the inhibitory effects and underlying mechanisms of 755 nm long-pulsed alexandrite laser on IH. METHODS Hemangioma endothelial cells (HemECs) were exposed to 755 nm long-pulsed alexandrite laser to evaluate its impact on cell proliferation and apoptosis. A patient-derived xenograft model was established to assess the inhibitory effects of laser treatment on IH in vivo. RESULTS In vitro, 755 nm long-pulsed alexandrite laser effectively suppressed the proliferation of HemECs and induced cell apoptosis. Laser treatment significantly inhibited the volume and weight of tumors, accompanied by significant downregulation of vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor receptor 2 (VEGFR2), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt) expression levels in both hemangioma cells and tumors. Additionally, laser treatment resulted in the conversion of VEGFA165a to VEGFA165b. TUNEL staining demonstrated increased apoptosis in tumor cells after laser treatment, along with upregulation of cleaved caspase-3 and Bax, and downregulation of Bcl-2. CONCLUSION In addition to the principle of selective photothermal decomposition, modulation of the VEGF/PI3K/Akt axis may serve as a potential mechanism for IH treatment using a long pulse-width 755 nm laser. This sheds valuable light on the molecular mechanisms underlying IH pathogenesis and potential therapeutic targets while providing a theoretical basis for the safe and efficient management of proliferative IH using laser therapy.
Collapse
Affiliation(s)
- Chen Ke
- Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Changhan Chen
- Department of Cosmetology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China; Wenzhou Key Laboratory of Laser Cosmetology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
| | - Ming Yang
- Department of Cosmetology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
| | - Hao Chen
- Department of Cosmetology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
| | - Youhui Ke
- Department of Cosmetology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China; Wenzhou Key Laboratory of Laser Cosmetology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China.
| | - Liqun Li
- Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Pawlik VE, Sonntag SR, Grisanti S, Tura A, Kakkassery V, Ranjbar M. Impact of Nintedanib and Anti-Angiogenic Agents on Uveal Melanoma Cell Behavior. Invest Ophthalmol Vis Sci 2024; 65:30. [PMID: 38381412 PMCID: PMC10893901 DOI: 10.1167/iovs.65.2.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/21/2024] [Indexed: 02/22/2024] Open
Abstract
Purpose The purpose of this study was to investigate the direct impact of the combined angiokinase inhibitor nintedanib as well as the anti-angiogenic agents ranibizumab, bevacizumab, and aflibercept on the primary uveal melanoma (UM) cell line Mel270 and liver metastasis UM cell line OMM2.5. Methods The metabolic activity, viability, and oxidative stress levels were analyzed by the Thiazolyl Blue Tetrazolium Bromide (MTT), LIVE/DEAD, and reactive oxygen species (ROS) assays. Expression of intracellular VEGF-A165 and VEGF receptor-2 was detected by immunofluorescent staining. The secretion of VEGF-A165 into the cell culture supernatants was evaluated by VEGF-A165 ELISA. Results Nintedanib, at a concentration of 1 µg/mL, resulted in a median reduction of metabolic activity (for Mel270 of approximately 38% and for OMM2.5 of 46% compared to the untreated control) without exerting toxicity in either cell line, whereas the other 3 substances did not result in any changes (which also means that none of the 4 substances led to an increased cell death). Moreover, nintedanib (1 µg/mL) induced oxidative stress in the Mel270 by approximately 1.2 to 1.5-fold compared to the untreated control, but not the OMM2.5 cells. Conclusions Nintedanib could suppress the growth of UM cells in a concentration-dependent manner. The metastatic UM cell line OMM2.5 was not sensitive to the pro-oxidant activity of nintedanib. This study was the first to investigate nintedanib in the context of UM. We propose further investigation of this substance to elucidate its effects on this tumor entity with the hope of identifying advantageous therapeutic options for future adjuvant tumor therapies.
Collapse
Affiliation(s)
- Vera E. Pawlik
- Department of Ophthalmology, University of Lübeck, Lübeck, Germany
| | | | | | - Aysegül Tura
- Department of Ophthalmology, University of Lübeck, Lübeck, Germany
| | | | - Mahdy Ranjbar
- Department of Ophthalmology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
3
|
Mou Z, Spencer J, McGrath JS, Harries LW. Comprehensive analysis of alternative splicing across multiple transcriptomic cohorts reveals prognostic signatures in prostate cancer. Hum Genomics 2023; 17:97. [PMID: 37924098 PMCID: PMC10623736 DOI: 10.1186/s40246-023-00545-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: 09/06/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Alternative splicing (AS) plays a crucial role in transcriptomic diversity and is a hallmark of cancer that profoundly influences the development and progression of prostate cancer (PCa), a prevalent and potentially life-limiting cancer among men. Accumulating evidence has highlighted the association between AS dysregulation and the onset and progression of PCa. However, a comprehensive and integrative analysis of AS profiles at the event level, utilising data from multiple high-throughput cohorts and evaluating the prognosis of PCa progression, remains lacking and calls for thorough exploration. RESULTS We identified a differentially expressed retained intron event in ZWINT across three distinct cohorts, encompassing an original array-based dataset profiled by us previously and two RNA sequencing (RNA-seq) datasets. Subsequent in-depth analyses of these RNA-seq datasets revealed 141 altered events, of which 21 demonstrated a significant association with patients' biochemical recurrence-free survival (BCRFS). We formulated an AS event-based prognostic signature, capturing six pivotal events in genes CYP4F12, NFATC4, PIGO, CYP3A5, ALS2CL, and FXYD3. This signature effectively differentiated high-risk patients diagnosed with PCa, who experienced shorter BCRFS, from their low-risk counterparts. Notably, the signature's predictive power surpassed traditional clinicopathological markers in forecasting 5-year BCRFS, demonstrating robust performance in both internal and external validation sets. Lastly, we constructed a novel nomogram that integrates patients' Gleason scores with pathological tumour stages, demonstrating improved prognostication of BCRFS. CONCLUSIONS Prediction of clinical progression remains elusive in PCa. This research uncovers novel splicing events associated with BCRFS, augmenting existing prognostic tools, thus potentially refining clinical decision-making.
Collapse
Affiliation(s)
- Zhuofan Mou
- Clinical and Biomedical Sciences, Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Barrack Road, Exeter, EX2 5DW, UK
| | - Jack Spencer
- Translational Research Exchange at Exeter, Living Systems Institute, University of Exeter, Exeter, UK
| | - John S McGrath
- Clinical and Biomedical Sciences, Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Barrack Road, Exeter, EX2 5DW, UK
- Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Lorna W Harries
- Clinical and Biomedical Sciences, Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
4
|
Quan B, Li Z, Yang H, Li S, Yan X, Wang Y. The splicing factor YBX1 promotes the progression of osteosarcoma by upregulating VEGF 165 and downregulating VEGF 165b. Heliyon 2023; 9:e18706. [PMID: 37554848 PMCID: PMC10405001 DOI: 10.1016/j.heliyon.2023.e18706] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
VEGF165 and its isoform VEGF165b have the same length but opposite functions in cancer. Some studies have indicated the important role of VEGF165 in osteosarcoma (OS); however, VEGF165b has not been taken into consideration. This study aims to clarify the roles of the two isoforms in OS and the mechanism controlling their formation from an alternative splicing perspective. By in vivo and in vitro experiments, we assessed the expression and function of VEGF165 and VEGF165b, screened the underlying splicing factors, and verified the regulatory function of splicing factor YBX1 on the two isoforms and its role in OS. The results showed that in OS, VEGF165 was upregulated but VEGF165b was downregulated. VEGF165 promoted the proliferation, migration and invasion of OS cells and induced angiogenesis in OS tumours; however, VEGF165b showed the opposite function. Of the four screened splicing factors, YBX1 was upregulated in OS tissues. It was positively correlated with VEGF165 but negatively correlated with VEGF165b. Further study indicated that YBX1 could upregulate VEGF165 but downregulate VEGF165b. Moreover, YBX1 promoted the proliferation, migration and invasion of OS cells and induced angiogenesis in OS tumours. OS patients with higher YBX1 had a poor prognosis within five years, but this difference disappeared in a longer follow-up. In conclusion, VEGF165b was antineoplastic and downregulated in OS, in contrast to VEGF165. YBX1 was found to be an important splicing factor that increased VEGF165 but decreased VEGF165b. Targeting YBX1 could endogenously alter the levels of VEGF165 and VEGF165b simultaneously.
Collapse
Affiliation(s)
- Bingxuan Quan
- The Fifth Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- The Fifth Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng, China
| | - Zhigang Li
- The Fifth Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng, China
| | - Hongbo Yang
- The Second Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng, China
| | - Shuo Li
- The Second Department of Orthopedics, The First Hospital of Qiqihar, Qiqihar, Heilongjiang Province, China
| | - Xiuchun Yan
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Yansong Wang
- The Fifth Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
5
|
Abstract
Dysregulated RNA splicing is a molecular feature that characterizes almost all tumour types. Cancer-associated splicing alterations arise from both recurrent mutations and altered expression of trans-acting factors governing splicing catalysis and regulation. Cancer-associated splicing dysregulation can promote tumorigenesis via diverse mechanisms, contributing to increased cell proliferation, decreased apoptosis, enhanced migration and metastatic potential, resistance to chemotherapy and evasion of immune surveillance. Recent studies have identified specific cancer-associated isoforms that play critical roles in cancer cell transformation and growth and demonstrated the therapeutic benefits of correcting or otherwise antagonizing such cancer-associated mRNA isoforms. Clinical-grade small molecules that modulate or inhibit RNA splicing have similarly been developed as promising anticancer therapeutics. Here, we review splicing alterations characteristic of cancer cell transcriptomes, dysregulated splicing's contributions to tumour initiation and progression, and existing and emerging approaches for targeting splicing for cancer therapy. Finally, we discuss the outstanding questions and challenges that must be addressed to translate these findings into the clinic.
Collapse
Affiliation(s)
- Robert K Bradley
- Computational Biology Program, Public Health Sciences Division and Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Olga Anczuków
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
| |
Collapse
|
6
|
The VEGF/VEGFR Axis Revisited: Implications for Cancer Therapy. Int J Mol Sci 2022; 23:ijms232415585. [PMID: 36555234 PMCID: PMC9779738 DOI: 10.3390/ijms232415585] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor (VEGFR) axis is indispensable in the process of angiogenesis and has been implicated as a key driver of tumor vascularization. Consequently, several strategies that target VEGF and its cognate receptors, VEGFR-1 and VEGFR-2, have been designed to treat cancer. While therapies targeting full-length VEGF have resulted in an improvement in both overall survival and progression-free survival in various cancers, these benefits have been modest. In addition, the inhibition of VEGFRs is associated with undesirable off-target effects. Moreover, VEGF splice variants that modulate sprouting and non-sprouting angiogenesis have been identified in recent years. Cues within the tumor microenvironment determine the expression patterns of these variants. Noteworthy is that the mechanisms of action of these variants challenge the established norm of VEGF signaling. Furthermore, the aberrant expression of some of these variants has been observed in several cancers. Herein, developments in the understanding of the VEGF/VEGFR axis and the splice products of these molecules, as well as the environmental cues that regulate these variants are reviewed. Furthermore, strategies that incorporate the targeting of VEGF variants to enhance the effectiveness of antiangiogenic therapies in the clinical setting are discussed.
Collapse
|
7
|
Arrigo A, Aragona E, Bandello F. VEGF-targeting drugs for the treatment of retinal neovascularization in diabetic retinopathy. Ann Med 2022; 54:1089-1111. [PMID: 35451900 PMCID: PMC9891228 DOI: 10.1080/07853890.2022.2064541] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Diabetic retinopathy (DR) is the most common microangiopathic complication of diabetes mellitus, representing a major cause of visual impairment in developed countries. Proliferative DR (PDR) represents the last stage of this extremely complex retinal disease, characterized by the development of neovascularization induced by the abnormal production and release of vascular endothelial growth factor (VEGF). The term VEGF includes different isoforms; VEGF-A represents one of the most important pathogenic factors of DR. Anti-VEGF intravitreal therapies radically changed the outcome of DR, due to combined anti-angiogenic and anti-edematous activities. Nowadays, several anti-VEGF molecules exist, characterized by different pharmacological features and duration. With respect to PDR, although anti-VEGF treatments represented a fundamental step forward in the management of this dramatic complication, a big debate is present in the literature regarding the role of anti-VEGF as substitute of panretinal photocoagulation or if these two approaches may be used in combination. In the present review, we provided an update on VEGF isoforms and their role in DR pathogenesis, on current anti-VEGF molecules and emerging new drugs, and on the current management strategies of PDR. There is an overall agreement regarding the relative advantage provided by anti-VEGF, especially looking at the management of PDR patients requiring vitrectomy, with respect to laser. Based on the current data, laser approaches might be avoided when a perfectly planned anti-VEGF therapeutic strategy can be adopted. Conversely, laser treatment may have a role for those patients unable to guarantee enough compliance to anti-VEGF injections.Key messagesVEGF increased production, stimulated by retinal hypoperfusion and ischaemia, is a major pathogenic factor of neovascular complication onset in diabetic retinopathy and of DR stages progression.Nowadays, several anti-VEGF molecules are available in clinical practice and other molecules are currently under investigation. Each anti-VEGF molecule is characterized by different targets and may interact with multiple biochemical pathways within the eye.All the data agreed in considering anti-VEGF molecules as a first line choice for the management of diabetic retinopathy. Laser treatments may have a role in selected advanced cases and for those patients unable to guarantee enough compliance to intravitreal treatments schemes.
Collapse
Affiliation(s)
- Alessandro Arrigo
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Emanuela Aragona
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Bandello
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
8
|
Wimberger P, Gerber MJ, Pfisterer J, Erdmann K, Füssel S, Link T, du Bois A, Kommoss S, Heitz F, Sehouli J, Kimmig R, de Gregorio N, Schmalfeldt B, Park-Simon TW, Baumann K, Hilpert F, Grube M, Schröder W, Burges A, Belau A, Hanker L, Kuhlmann JD. Bevacizumab May Differentially Improve Prognosis of Advanced Ovarian Cancer Patients with Low Expression of VEGF-A165b, an Antiangiogenic VEGF-A Splice Variant. Clin Cancer Res 2022; 28:4660-4668. [PMID: 36001383 DOI: 10.1158/1078-0432.ccr-22-1326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE The identification of a robust IHC marker to predict the response to antiangiogenic bevacizumab in ovarian cancer is of high clinical interest. VEGF-A, the molecular target of bevacizumab, is expressed as multiple isoforms with pro- or antiangiogenic properties, of which VEGF-A165b is the most dominant antiangiogenic isoform. The balance of VEGF-A isoforms is closely related to the angiogenic capacity of a tumor and may define its vulnerability to antiangiogenic therapy. We investigated whether the expression of VEGF-A165b could be related to the effect of bevacizumab in advanced ovarian cancer patients. EXPERIMENTAL DESIGN Formalin-fixed paraffin-embedded tissues from 413 patients of the ICON7 multicenter phase III trial, treated with standard platinum-based chemotherapy with or without bevacizumab, were probed for VEGF-A165b expression by IHC. RESULTS In patients with low VEGF-A165b expression, the addition of bevacizumab to standard platinum-based chemotherapy significantly improved progression-free (HR: 0.727; 95% CI, 0.538-0.984; P = 0.039) and overall survival (HR: 0.662; 95% CI, 0.458-0.958; P = 0.029). Multivariate analysis showed that the addition of bevacizumab in low VEGF-A165b-expressing patients conferred significant improvements in progression-free survival (HR: 0.610; 95% CI, 0.446-0.834; P = 0.002) and overall survival (HR: 0.527; 95% CI, 0.359-0.775; P = 0.001), independently from established risk factors. CONCLUSIONS We demonstrate for the first time that bevacizumab may differentially improve the prognosis of advanced ovarian cancer patients with low expression of VEGF-A165b, an antiangiogenic VEGF-A splice variant. We envision that this novel biomarker could be implemented into routine diagnostics and may have direct clinical implications for guiding bevacizumab-related treatment decisions in advanced ovarian cancer patients.
Collapse
Affiliation(s)
- Pauline Wimberger
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,AGO Study Group, Wiesbaden, Germany
| | - Mara Julia Gerber
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jacobus Pfisterer
- AGO Study Group, Wiesbaden, Germany.,Gynecologic Oncology Center, Kiel, Germany
| | - Kati Erdmann
- National Center for Tumour Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Susanne Füssel
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Theresa Link
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte (KEM), Essen, Germany
| | - Stefan Kommoss
- Department Gynecology and Gynecologic Oncology, University of Tuebingen, Tübingen, Germany
| | - Florian Heitz
- AGO Study Group, Wiesbaden, Germany.,Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte (KEM), Essen, Germany
| | - Jalid Sehouli
- AGO Study Group, Wiesbaden, Germany.,Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Rainer Kimmig
- AGO Study Group, Wiesbaden, Germany.,University Hospital Essen, Essen, Germany
| | - Nikolaus de Gregorio
- AGO Study Group, Wiesbaden, Germany.,University Hospital Ulm, Ulm, Germany and SLK-Kliniken Heilbronn, Klinikum am Gesundbrunnen, Heilbronn, Germany
| | - Barbara Schmalfeldt
- AGO Study Group, Wiesbaden, Germany.,University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Klaus Baumann
- AGO Study Group, Wiesbaden, Germany.,University Hospital Gießen and Marburg, Marburg, Germany; Hospital Ludwigshafen, Ludwigshafen, Germany
| | - Felix Hilpert
- AGO Study Group, Wiesbaden, Germany.,University Hospital Schleswig-Holstein, Kiel, Germany; Krankenhaus Jerusalem, Mammazentrum Hamburg, Hamburg, Germany
| | - Marcel Grube
- AGO Study Group, Wiesbaden, Germany.,Department Gynecology and Gynecologic Oncology, University of Tuebingen, Tübingen, Germany
| | - Willibald Schröder
- AGO Study Group, Wiesbaden, Germany.,Klinikum Bremen-Mitte, Bremen, Germany; GYNAEKOLOGICUM Bremen, Bremen, Germany
| | - Alexander Burges
- AGO Study Group, Wiesbaden, Germany.,University Hospital LMU Munich, Munich, Germany
| | - Antje Belau
- AGO Study Group, Wiesbaden, Germany.,University Hospital Greifswald, Greifswald, Germany; Frauenarztpraxis Dr. Belau, Greifswald, Germany
| | - Lars Hanker
- AGO Study Group, Wiesbaden, Germany.,Department of Gynecology and Obstetrics University Hospital Frankfurt, Frankfurt, Germany; Department of Gynecology and Obstetrics University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumour Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
9
|
Al Kawas H, Saaid I, Jank P, Westhoff CC, Denkert C, Pross T, Weiler KBS, Karsten MM. How VEGF-A and its splice variants affect breast cancer development - clinical implications. Cell Oncol (Dordr) 2022; 45:227-239. [PMID: 35303290 PMCID: PMC9050780 DOI: 10.1007/s13402-022-00665-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Altered expression levels and structural variations in the vascular endothelial growth factor (VEGF) have been found to play important roles in cancer development and to be associated with the overall survival and therapy response of cancer patients. Particularly VEGF-A and its splice variants have been found to affect physiological and pathological angiogenic processes, including tumor angiogenesis, correlating with tumor progression, mostly caused by overexpression. This review focuses on the expression and impact of VEGF-A splice variants under physiologic conditions and in tumors and, in particular, the distribution and role of isoform VEGF165b in breast cancer. CONCLUSIONS AND PERSPECTIVES Many publications already highlighted the importance of VEGF-A and its splice variants in tumor therapy, especially in breast cancer, which are summarized in this review. Furthermore, we were able to demonstrate that cytoplasmatic VEGFA/165b expression is higher in invasive breast cancer tumor cells than in normal tissues or stroma. These examples show that the detection of VEGF splice variants can be performed also on the protein level in formalin fixed tissues. Although no quantitative conclusions can be drawn, these results may be the starting point for further studies at a quantitative level, which can be a major step towards the design of targeted antibody-based (breast) cancer therapies.
Collapse
Affiliation(s)
- Hivin Al Kawas
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Inas Saaid
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Paul Jank
- Institute of Pathology, Philipps-Universität Marburg, 35043, Marburg, Germany
| | | | - Carsten Denkert
- Institute of Pathology, Philipps-Universität Marburg, 35043, Marburg, Germany
| | - Therese Pross
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Maria Margarete Karsten
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
10
|
Calderon-Aparicio A, Wang BD. Prostate cancer: Alternatively spliced mRNA transcripts in tumor progression and their uses as therapeutic targets. Int J Biochem Cell Biol 2021; 141:106096. [PMID: 34653618 PMCID: PMC8639776 DOI: 10.1016/j.biocel.2021.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 10/20/2022]
Abstract
Prostate cancer is the most frequently diagnosed cancer and second leading cause of cancer deaths among American men. Current therapies show early antitumor responses, but ultimately lead to treatment resistance, relapse and poorer survival in patients. Alternative RNA splicing, a cell mechanism increasing the proteome diversity by producing multiple transcripts from a single gene, has been associated with prostate cancer development/progression. Reports showed that many aberrant mRNA splice variants are upregulated in prostate cancer, promoting malignancy through enhanced proliferation, metastasis, tumor growth, anti-apoptosis, and/or treatment resistance. Here, we discuss the oncogenic properties of aberrant splicing mechanisms underlying prostate cancer pathogenesis, as well as the uses of the splicing variants as potential diagnostics and treatment targets. Finally, we discuss the pharmacologic and molecular approaches for targeting aberrant splicing mechanisms as effective therapies to correct the splicing errors and overcome the drug resistance, ultimately improving the clinical outcome of prostate cancer patients.
Collapse
Affiliation(s)
- Ali Calderon-Aparicio
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA.
| |
Collapse
|
11
|
Lin Z, Wu Z, Luo W. A Novel Treatment for Ewing's Sarcoma: Chimeric Antigen Receptor-T Cell Therapy. Front Immunol 2021; 12:707211. [PMID: 34566963 PMCID: PMC8461297 DOI: 10.3389/fimmu.2021.707211] [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: 05/09/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022] Open
Abstract
Ewing's sarcoma (EWS) is a malignant and aggressive tumor type that predominantly occurs in children and adolescents. Traditional treatments such as surgery, radiotherapy and chemotherapy, while successful in the early disease stages, are ineffective in patients with metastases and relapses who often have poor prognosis. Therefore, new treatments for EWS are needed to improve patient's outcomes. Chimeric antigen receptor (CAR)-T cells therapy, a novel adoptive immunotherapy, has been developing over the past few decades, and is increasingly popular in researches and treatments of various cancers. CAR-T cell therapy has been approved by the Food and Drug Administration (FDA) for the treatment of leukemia and lymphoma. Recently, this therapeutic approach has been employed for solid tumors including EWS. In this review, we summarize the safety, specificity and clinical transformation of the treatment targets of EWS, and point out the directions for further research.
Collapse
Affiliation(s)
| | | | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Star E, Stevens M, Gooding C, Smith CWJ, Li L, Ayine ML, Harper SJ, Bates DO, Oltean S. A drug-repositioning screen using splicing-sensitive fluorescent reporters identifies novel modulators of VEGF-A splicing with anti-angiogenic properties. Oncogenesis 2021; 10:36. [PMID: 33941763 PMCID: PMC8093282 DOI: 10.1038/s41389-021-00323-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 03/25/2021] [Accepted: 04/09/2021] [Indexed: 12/30/2022] Open
Abstract
Alternative splicing of the vascular endothelial growth factor A (VEGF-A) terminal exon generates two protein families with differing functions. Pro-angiogenic VEGF-Axxxa isoforms are produced via selection of the proximal 3' splice site of the terminal exon. Use of an alternative distal splice site generates the anti-angiogenic VEGF-Axxxb proteins. A bichromatic splicing-sensitive reporter was designed to mimic VEGF-A alternative splicing and was used as a molecular tool to further investigate this alternative splicing event. Part of VEGF-A's terminal exon and preceding intron were inserted into a minigene construct followed by the coding sequences for two fluorescent proteins. A different fluorescent protein is expressed depending on which 3' splice site of the exon is used during splicing (dsRED denotes VEGF-Axxxa and EGFP denotes VEGF-Axxxb). The fluorescent output can be used to follow splicing decisions in vitro and in vivo. Following successful reporter validation in different cell lines and altering splicing using known modulators, a screen was performed using the LOPAC library of small molecules. Alterations to reporter splicing were measured using a fluorescent plate reader to detect dsRED and EGFP expression. Compounds of interest were further validated using flow cytometry and assessed for effects on endogenous VEGF-A alternative splicing at the mRNA and protein level. Ex vivo and in vitro angiogenesis assays were used to demonstrate the anti-angiogenic effect of the compounds. Furthermore, anti-angiogenic activity was investigated in a Matrigel in vivo model. To conclude, we have identified a set of compounds that have anti-angiogenic activity through modulation of VEGF-A terminal exon splicing.
Collapse
Affiliation(s)
- Eleanor Star
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Megan Stevens
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Clare Gooding
- grid.5335.00000000121885934Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW UK
| | - Christopher W. J. Smith
- grid.5335.00000000121885934Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW UK
| | - Ling Li
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Monica Lamici Ayine
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Steve J. Harper
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - David O. Bates
- grid.415598.40000 0004 0641 4263Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen’s Medical Centre, West Block, D floor, Nottingham, NG7 2UH UK
| | - Sebastian Oltean
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| |
Collapse
|
13
|
Li K, Wang Z. Splicing factor SRSF2-centric gene regulation. Int J Biol Sci 2021; 17:1708-1715. [PMID: 33994855 PMCID: PMC8120470 DOI: 10.7150/ijbs.58888] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/04/2021] [Indexed: 01/14/2023] Open
Abstract
Serine/arginine-rich splicing factor 2 (SRSF2) is a splicing factor that is widely expressed in a variety of mammalian cell types. Increasing evidence has confirmed that SRSF2 plays vital roles in a number of biological and pathological processes. Therefore, it is important to understand how its expression is regulated, and how it regulates the expression of its target genes. Recently, we found that SRSF2 expression could be upregulated by herpes simplex virus-1 (HSV-1) infection, and that altered SRSF2 expression, in turn, epigenetically regulates the transcription of HSV-1 genes. Further studies on T cell exhaustion demonstrated that upregulated SRSF2 in exhausted T cells elevated the levels of multiple immune checkpoint molecules by associating with the acyl-transferases, P300 and CBP, and by altering histone modification near the transcription start sites of these genes, thereby influencing signal transducer and activator of transcription 3 binding to these gene promoters. These findings suggest that SRSF2 acts as an important sensor and effector during disease progression. Here, we discuss the molecules that regulate SRSF2 gene expression and their associated mechanisms, and the mechanisms via which SRSF2 regulates the expression of target genes, thus providing novel insights into the central role of SRSF2 in gene regulation.
Collapse
Affiliation(s)
- Kun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Ziqiang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China.,Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| |
Collapse
|
14
|
Tekin V, Aweda T, Kozgus Guldu O, Biber Muftuler FZ, Bartels J, Lapi SE, Unak P. A novel anti-angiogenic radio/photo sensitizer for prostate cancer imaging and therapy: 89Zr-Pt@TiO 2-SPHINX, synthesis and in vitro evaluation. Nucl Med Biol 2021; 94-95:20-31. [PMID: 33482596 DOI: 10.1016/j.nucmedbio.2020.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 01/27/2023]
Abstract
Prostate cancer is the most common malignancy and leading cause of cancer deaths in men. Thus, the development of novel strategies for performing combined prostate cancer imaging and therapy methods is crucial and could have a significant impact on patient care. This current study aimed to design a multimodality nanoconjugate to be used for both PET and optical imaging and as a therapeutic radio/photo sensitizer and anti-angiogenesis agent. Initial characterization of this novel nanoconjugate was performed via HPLC, FTIR, TEM and DLS analyses. Pt@TiO2-SPHINX was further evaluated using fluorometric and radiochromatographic methods. Cytotoxicity, cell uptake and internalization were also investigated as well as therapy with photodynamic/radio therapy combinations. Both nanoparticles and nanoconjugates were robustly synthesized according to literature methods. Radiochemistry and cell culture assays showed high 89Zr radiolabeling efficiency with sufficient stability for studies at later time points. Pt@TiO2-SPHINX was shown to target prostate cancer cells (PC3 and LNCaP), and was non-toxic to normal prostate cells (RWPE-1). This finding was supported by the WST-8 assay and AFM images. The uptake of the compound in prostate cancer cells is significantly higher than prostate normal cells and according to ELISA results, Pt@TiO2-SPHINX can increase anti-angiogenic VEGFA165b. Additionally, Pt@TiO2-SPHINX dramatically decreased the cell viability of prostate cancer cells when photodynamic and radio therapy were performed at the same time. In vitro results are promising for future studies of Pt@TiO2-SPHINX as a PET imaging agent and anti-angiogenic radio sensitizer.
Collapse
Affiliation(s)
- Volkan Tekin
- Institute of Nuclear Science, Ege University, Izmir, Turkey.
| | - Tolulope Aweda
- Department of Radiology, University of Alabama at Birmingham, AL, United States of America
| | | | | | - Jennifer Bartels
- Department of Radiology, University of Alabama at Birmingham, AL, United States of America
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, AL, United States of America
| | - Perihan Unak
- Institute of Nuclear Science, Ege University, Izmir, Turkey
| |
Collapse
|
15
|
Bessa C, Matos P, Jordan P, Gonçalves V. Alternative Splicing: Expanding the Landscape of Cancer Biomarkers and Therapeutics. Int J Mol Sci 2020; 21:ijms21239032. [PMID: 33261131 PMCID: PMC7729450 DOI: 10.3390/ijms21239032] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Alternative splicing (AS) is a critical post-transcriptional regulatory mechanism used by more than 95% of transcribed human genes and responsible for structural transcript variation and proteome diversity. In the past decade, genome-wide transcriptome sequencing has revealed that AS is tightly regulated in a tissue- and developmental stage-specific manner, and also frequently dysregulated in multiple human cancer types. It is currently recognized that splicing defects, including genetic alterations in the spliced gene, altered expression of both core components or regulators of the precursor messenger RNA (pre-mRNA) splicing machinery, or both, are major drivers of tumorigenesis. Hence, in this review we provide an overview of our current understanding of splicing alterations in cancer, and emphasize the need to further explore the cancer-specific splicing programs in order to obtain new insights in oncology. Furthermore, we also discuss the recent advances in the identification of dysregulated splicing signatures on a genome-wide scale and their potential use as biomarkers. Finally, we highlight the therapeutic opportunities arising from dysregulated splicing and summarize the current approaches to therapeutically target AS in cancer.
Collapse
Affiliation(s)
- Cláudia Bessa
- Department of Human Genetics, National Health Institute Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal; (C.B.); (P.M.)
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Paulo Matos
- Department of Human Genetics, National Health Institute Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal; (C.B.); (P.M.)
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Peter Jordan
- Department of Human Genetics, National Health Institute Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal; (C.B.); (P.M.)
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Correspondence: (P.J.); (V.G.); Tel.: +351-217-519-380 (P.J.)
| | - Vânia Gonçalves
- Department of Human Genetics, National Health Institute Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal; (C.B.); (P.M.)
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Correspondence: (P.J.); (V.G.); Tel.: +351-217-519-380 (P.J.)
| |
Collapse
|
16
|
Dardente H, English WR, Valluru MK, Kanthou C, Simpson D. Debunking the Myth of the Endogenous Antiangiogenic Vegfaxxxb Transcripts. Trends Endocrinol Metab 2020; 31:398-409. [PMID: 32396842 DOI: 10.1016/j.tem.2020.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/28/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
In this opinion article we critically assess evidence for the existence of a family of antiangiogenic vascular endothelial growth factor (Vegfaxxxb) transcripts, arising from the use of a phylogenetically conserved alternative distal splice site within exon 8 of the VEGFA gene. We explain that prior evidence for Vegfaxxxb transcripts in tissues rests heavily upon flawed RT-PCR methodologies, with the extensive use of 5'-tailing in primer design being the main issue. Furthermore, our analysis of large RNA-seq data sets (human and ovine) fails to identify a single Vegfaxxxb transcript. Therefore, we challenge the very existence of Vegfaxxxb transcripts, which further questions the physiological relevance of studies based on the use of 'anti-VEGFAxxxb' antibodies. Our analysis has implications for the proposed therapeutic use of isoform-specific anti-VEGFA strategies for treating cancer and retinopathies.
Collapse
Affiliation(s)
- Hugues Dardente
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France.
| | - William R English
- Department of Oncology and Metabolism, Tumour Microcirculation Group, University of Sheffield, School of Medicine, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Manoj K Valluru
- Department of Oncology and Metabolism, Tumour Microcirculation Group, University of Sheffield, School of Medicine, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Chryso Kanthou
- Department of Oncology and Metabolism, Tumour Microcirculation Group, University of Sheffield, School of Medicine, Beech Hill Road, Sheffield, S10 2RX, UK
| | - David Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT7 1NN, UK
| |
Collapse
|
17
|
Karsten MM, Beck MH, Rademacher A, Knabl J, Blohmer JU, Jückstock J, Radosa JC, Jank P, Rack B, Janni W. VEGF-A165b levels are reduced in breast cancer patients at primary diagnosis but increase after completion of cancer treatment. Sci Rep 2020; 10:3635. [PMID: 32108136 PMCID: PMC7046696 DOI: 10.1038/s41598-020-59823-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/30/2020] [Indexed: 01/26/2023] Open
Abstract
The antiangiogenic splice variant VEGF-A165b is downregulated in a variety of cancer entities, but little is known so far about circulating plasma levels. The present analysis addresses this question and examines circulating VEGF-A/VEGF-A165b levels in a collective of female high-risk breast cancer patients over the course of treatment. Within the SUCCES-A trial 205 patients were recruited after having received primary breast surgery. Using ELISA VEGF-A/VEGF-A165b concentrations were determined and correlated to clinical characteristics (1) before adjuvant chemotherapy, (2) four weeks and (3) two years after therapy and compared to healthy controls (n = 107). VEGF165b levels were significantly elevated after completion of chemotherapy. Within the breast cancer cohort, VEGF-A165b levels increased two years after completion of chemotherapy. VEGF-A plasma concentrations were significantly elevated in the breast cancer cohort at all examined time points and decreased after treatment. VEGF-A levels two years after chemotherapy correlated with increased cancer related mortality, no such correlation could be found between VEGF-A165b and the examined clinical characteristics. Compared to controls, VEGF-A/VEGF-A165b ratios were decreased in patients before and after chemotherapy. Our data suggests that circulating VEGF-A165b is significantly reduced in women with primary breast cancer at time of diagnosis; furthermore, levels change during adjuvant treatment.
Collapse
Affiliation(s)
- Maria Margarete Karsten
- Department of Gynecology and Breast Care Center, University Hospital, Charité Universitätsmedizin Berlin, Berlin, Germany.
| | - Maximilian Heinz Beck
- Department of Gynecology and Breast Care Center, University Hospital, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Angela Rademacher
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany.,Department of Orthopedics, Schön Clinic, Munich, Harlaching, Germany
| | - Julia Knabl
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology and Breast Care Center, University Hospital, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Jückstock
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Julia Caroline Radosa
- Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, Germany
| | - Paul Jank
- Department of Pathology, Philipps-University Marburg, Marburg, Germany
| | - Brigitte Rack
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany.,Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
18
|
Ji Y, Mishra RK, Davuluri RV. In silico analysis of alternative splicing on drug-target gene interactions. Sci Rep 2020; 10:134. [PMID: 31924844 PMCID: PMC6954184 DOI: 10.1038/s41598-019-56894-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/18/2019] [Indexed: 12/24/2022] Open
Abstract
Identifying and evaluating the right target are the most important factors in early drug discovery phase. Most studies focus on one protein ignoring the multiple splice-variant or protein-isoforms, which might contribute to unexpected therapeutic activity or adverse side effects. Here, we present computational analysis of cancer drug-target interactions affected by alternative splicing. By integrating information from publicly available databases, we curated 883 FDA approved or investigational stage small molecule cancer drugs that target 1,434 different genes, with an average of 5.22 protein isoforms per gene. Of these, 618 genes have ≥5 annotated protein-isoforms. By analyzing the interactions with binding pocket information, we found that 76% of drugs either miss a potential target isoform or target other isoforms with varied expression in multiple normal tissues. We present sequence and structure level alignments at isoform-level and make this information publicly available for all the curated drugs. Structure-level analysis showed ligand binding pocket architectures differences in size, shape and electrostatic parameters between isoforms. Our results emphasize how potentially important isoform-level interactions could be missed by solely focusing on the canonical isoform, and suggest that on- and off-target effects at isoform-level should be investigated to enhance the productivity of drug-discovery research.
Collapse
Affiliation(s)
- Yanrong Ji
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rama K Mishra
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA.,Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ramana V Davuluri
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
19
|
An Intricate Connection between Alternative Splicing and Phenotypic Plasticity in Development and Cancer. Cells 2019; 9:cells9010034. [PMID: 31877720 PMCID: PMC7016785 DOI: 10.3390/cells9010034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
During tumor progression, hypoxia, nutrient deprivation or changes in the extracellular environment (i.e., induced by anti-cancer drugs) elicit adaptive responses in cancer cells. Cellular plasticity increases the chance that tumor cells may survive in a challenging microenvironment, acquire new mechanisms of resistance to conventional drugs, and spread to distant sites. Re-activation of stem pathways appears as a significant cause of cellular plasticity because it promotes the acquisition of stem-like properties through a profound phenotypic reprogramming of cancer cells. In addition, it is a major contributor to tumor heterogeneity, depending on the coexistence of phenotypically distinct subpopulations in the same tumor bulk. Several cellular mechanisms may drive this fundamental change, in particular, high-throughput sequencing technologies revealed a key role for alternative splicing (AS). Effectively, AS is one of the most important pre-mRNA processes that increases the diversity of transcriptome and proteome in a tissue- and development-dependent manner. Moreover, defective AS has been associated with several human diseases. However, its role in cancer cell plasticity and tumor heterogeneity remains unclear. Therefore, unravelling the intricate relationship between AS and the maintenance of a stem-like phenotype may explain molecular mechanisms underlying cancer cell plasticity and improve cancer diagnosis and treatment.
Collapse
|
20
|
Pentheroudakis G, Mavroeidis L, Papadopoulou K, Koliou GA, Bamia C, Chatzopoulos K, Samantas E, Mauri D, Efstratiou I, Pectasides D, Makatsoris T, Bafaloukos D, Papakostas P, Papatsibas G, Bombolaki I, Chrisafi S, Kourea HP, Petraki K, Kafiri G, Fountzilas G, Kotoula V. Angiogenic and Antiangiogenic VEGFA Splice Variants in Colorectal Cancer: Prospective Retrospective Cohort Study in Patients Treated With Irinotecan-Based Chemotherapy and Bevacizumab. Clin Colorectal Cancer 2019; 18:e370-e384. [DOI: 10.1016/j.clcc.2019.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/07/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022]
|
21
|
Contribution of the VEGF system to the follicular persistence associated with bovine cystic ovaries. Theriogenology 2019; 138:52-65. [DOI: 10.1016/j.theriogenology.2019.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/05/2019] [Accepted: 07/02/2019] [Indexed: 01/09/2023]
|
22
|
Olender J, Lee NH. Role of Alternative Splicing in Prostate Cancer Aggressiveness and Drug Resistance in African Americans. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:119-139. [PMID: 31576545 PMCID: PMC6777849 DOI: 10.1007/978-3-030-22254-3_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alternative splicing, the process of removing introns and joining exons of pre-mRNA, is critical for growth, development, tissue homeostasis, and species diversity. Dysregulation of alternative splicing can initiate and drive disease. Aberrant alternative splicing has been shown to promote the "hallmarks of cancer" in both hematological and solid cancers. Of interest, recent work has focused on the role of alternative splicing in prostate cancer and prostate cancer health disparities. We will provide a review of prostate cancer health disparities involving the African American population, alternative RNA splicing, and alternative splicing in prostate cancer. Lastly, we will summarize our work on differential alternative splicing in prostate cancer disparities and its implications for disparate health outcomes and therapeutic targets.
Collapse
Affiliation(s)
- Jacqueline Olender
- Department of Pharmacology and Physiology, GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Norman H Lee
- Department of Pharmacology and Physiology, GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
23
|
Wu JB, Tang YL, Liang XH. Targeting VEGF pathway to normalize the vasculature: an emerging insight in cancer therapy. Onco Targets Ther 2018; 11:6901-6909. [PMID: 30410348 PMCID: PMC6200071 DOI: 10.2147/ott.s172042] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vascular normalization is a new concept of targeting angiogenesis to restore vessel structure and function and to increase blood perfusion and delivery of drugs. It has been confirmed that vascular normalization can decrease relapse and benefit other cancer therapy, including chemotherapy, radiotherapy, and immune cell therapy. The key point of this therapy is to inhibit pro-angiogenic factors and make it be balanced with anti-angiogenic factors, resulting in a mature and normal vessel characteristic. Vascular endothelial growth factor (VEGF) is a key player in the process of tumor angiogenesis, and inhibiting VEGF is a primary approach to tumor vessel normalization. Herein, we review newly uncovered mechanisms governing angiogenesis and vascular normalization of cancer and place emphasis on targeting VEGF pathway to normalize the vasculature. Also, important methods to depress VEGF pathway and make tumor vascular are discussed.
Collapse
Affiliation(s)
- Jing-Biao Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, People's Republic of China, ;
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, People's Republic of China, ;
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, People's Republic of China, ;
| |
Collapse
|
24
|
Zhang H, Jia E, Xia W, Lu C, Zhu W. VEGF165b mutant with a prolonged half-life and enhanced anti-tumor potency in a mouse model. J Biotechnol 2018; 284:84-90. [DOI: 10.1016/j.jbiotec.2018.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 08/04/2018] [Accepted: 08/06/2018] [Indexed: 01/26/2023]
|
25
|
Boudria A, Abou Faycal C, Jia T, Gout S, Keramidas M, Didier C, Lemaître N, Manet S, Coll JL, Toffart AC, Moro-Sibilot D, Albiges-Rizo C, Josserand V, Faurobert E, Brambilla C, Brambilla E, Gazzeri S, Eymin B. VEGF 165b, a splice variant of VEGF-A, promotes lung tumor progression and escape from anti-angiogenic therapies through a β1 integrin/VEGFR autocrine loop. Oncogene 2018; 38:1050-1066. [PMID: 30194450 DOI: 10.1038/s41388-018-0486-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 06/25/2018] [Accepted: 07/29/2018] [Indexed: 12/21/2022]
Abstract
Vascular endothelial growth factor-A (VEGF-A) is highly subjected to alternative pre-mRNA splicing that generates several splice variants. The VEGFxxx and VEGFxxxb families encode splice variants of VEGF-A that differ only at the level of six amino acids in their C-terminal part. The expression level of VEGFxxx splice variants and their function as pro-angiogenic factors during tumor neo-angiogenesis have been well-described. The role of VEGFxxxb isoforms is less well known, but they have been shown to inhibit VEGFxxx-mediated angiogenesis, while being partial or weak activators of VEGFR receptors in endothelial cells. On the opposite, their role on tumor cells expressing VEGFRs at their surface remains largely unknown. In this study, we find elevated levels of VEGF165b, the main VEGFxxxb isoform, in 36% of non-small cell lung carcinoma (NSCLC), mainly lung adenocarcinoma (46%), and show that a high VEGF165b/VEGF165 ratio correlates with the presence of lymph node metastases. At the molecular level, we demonstrate that VEGF165b stimulates proliferation and invasiveness of two lung tumor cell lines through a VEGFR/β1 integrin loop. We further provide evidence that the isoform-specific knockdown of VEGF165b reduces tumor growth, demonstrating a tumor-promoting autocrine role for VEGF165b in lung cancer cells. Importantly, we show that bevacizumab, an anti-angiogenic compound used for the treatment of lung adenocarcinoma patients, increases the expression of VEGF165b and activates the invasive VEGFR/β1 integrin loop. Overall, these data highlight an unexpected role of the VEGF165b splice variant in the progression of lung tumors and their response to anti-angiogenic therapies.
Collapse
Affiliation(s)
- Asma Boudria
- INSERM U1209, UMR CNRS 5309, Team RNA splicing, Cell Signaling and Response to Therapies, Grenoble, 38042, France.,Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France
| | - Cherine Abou Faycal
- INSERM U1209, UMR CNRS 5309, Team RNA splicing, Cell Signaling and Response to Therapies, Grenoble, 38042, France.,Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France
| | - Tao Jia
- INSERM U1209, UMR CNRS 5309, Team RNA splicing, Cell Signaling and Response to Therapies, Grenoble, 38042, France.,Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France
| | - Stephanie Gout
- INSERM U1209, UMR CNRS 5309, Team RNA splicing, Cell Signaling and Response to Therapies, Grenoble, 38042, France.,Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France
| | - Michelle Keramidas
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM, U1209, UMR CNRS 5309, Team Cancer Targets and Experimental Therapeutics, Grenoble, 38042, France
| | - Chloé Didier
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM, U1209, UMR CNRS 5309, Team Cancer Targets and Experimental Therapeutics, Grenoble, 38042, France
| | - Nicolas Lemaître
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Tumor Molecular Pathology and Biomarkers, Grenoble, 38042, France
| | - Sandra Manet
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Cell Adhesion Dynamics and Differentiation, Grenoble, 38042, France
| | - Jean-Luc Coll
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM, U1209, UMR CNRS 5309, Team Cancer Targets and Experimental Therapeutics, Grenoble, 38042, France
| | - Anne-Claire Toffart
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Tumor Molecular Pathology and Biomarkers, Grenoble, 38042, France
| | - Denis Moro-Sibilot
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Tumor Molecular Pathology and Biomarkers, Grenoble, 38042, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Cell Adhesion Dynamics and Differentiation, Grenoble, 38042, France
| | - Véronique Josserand
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM, U1209, UMR CNRS 5309, Team Cancer Targets and Experimental Therapeutics, Grenoble, 38042, France
| | - Eva Faurobert
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Cell Adhesion Dynamics and Differentiation, Grenoble, 38042, France
| | - Christian Brambilla
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Tumor Molecular Pathology and Biomarkers, Grenoble, 38042, France
| | - Elisabeth Brambilla
- Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.,INSERM U1209, UMR CNRS 5309, Team Tumor Molecular Pathology and Biomarkers, Grenoble, 38042, France
| | - Sylvie Gazzeri
- INSERM U1209, UMR CNRS 5309, Team RNA splicing, Cell Signaling and Response to Therapies, Grenoble, 38042, France.,Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France
| | - Beatrice Eymin
- INSERM U1209, UMR CNRS 5309, Team RNA splicing, Cell Signaling and Response to Therapies, Grenoble, 38042, France. .,Université Grenoble Alpes, Institut Pour l'Avancée des Biosciences, Grenoble, 38041, France.
| |
Collapse
|
26
|
Urbanski L, Leclair N, Anczuków O. Alternative-splicing defects in cancer: Splicing regulators and their downstream targets, guiding the way to novel cancer therapeutics. WILEY INTERDISCIPLINARY REVIEWS. RNA 2018; 9:e1476. [PMID: 29693319 PMCID: PMC6002934 DOI: 10.1002/wrna.1476] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/14/2022]
Abstract
Defects in alternative splicing are frequently found in human tumors and result either from mutations in splicing-regulatory elements of specific cancer genes or from changes in the regulatory splicing machinery. RNA splicing regulators have emerged as a new class of oncoproteins and tumor suppressors, and contribute to disease progression by modulating RNA isoforms involved in the hallmark cancer pathways. Thus, dysregulation of alternative RNA splicing is fundamental to cancer and provides a potentially rich source of novel therapeutic targets. Here, we review the alterations in splicing regulatory factors detected in human tumors, as well as the resulting alternatively spliced isoforms that impact cancer hallmarks, and discuss how they contribute to disease pathogenesis. RNA splicing is a highly regulated process and, as such, the regulators are themselves tightly regulated. Differential transcriptional and posttranscriptional regulation of splicing factors modulates their levels and activities in tumor cells. Furthermore, the composition of the tumor microenvironment can also influence which isoforms are expressed in a given cell type and impact drug responses. Finally, we summarize current efforts in targeting alternative splicing, including global splicing inhibition using small molecules blocking the spliceosome or splicing-factor-modifying enzymes, as well as splice-switching RNA-based therapeutics to modulate cancer-specific splicing isoforms. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Processing > Splicing Regulation/Alternative Splicing.
Collapse
|
27
|
Błochowiak K, Sokalski J, Golusińska E, Trzybulska D, Witmanowski H, Bodnar M, Marszałek A. Salivary levels and immunohistochemical expression of selected angiogenic factors in benign and malignant parotid gland tumours. Clin Oral Investig 2018; 23:995-1006. [DOI: 10.1007/s00784-018-2524-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 06/11/2018] [Indexed: 01/06/2023]
|
28
|
Peach CJ, Mignone VW, Arruda MA, Alcobia DC, Hill SJ, Kilpatrick LE, Woolard J. Molecular Pharmacology of VEGF-A Isoforms: Binding and Signalling at VEGFR2. Int J Mol Sci 2018; 19:E1264. [PMID: 29690653 PMCID: PMC5979509 DOI: 10.3390/ijms19041264] [Citation(s) in RCA: 287] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/14/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A) is a key mediator of angiogenesis, signalling via the class IV tyrosine kinase receptor family of VEGF Receptors (VEGFRs). Although VEGF-A ligands bind to both VEGFR1 and VEGFR2, they primarily signal via VEGFR2 leading to endothelial cell proliferation, survival, migration and vascular permeability. Distinct VEGF-A isoforms result from alternative splicing of the Vegfa gene at exon 8, resulting in VEGFxxxa or VEGFxxxb isoforms. Alternative splicing events at exons 5⁻7, in addition to recently identified posttranslational read-through events, produce VEGF-A isoforms that differ in their bioavailability and interaction with the co-receptor Neuropilin-1. This review explores the molecular pharmacology of VEGF-A isoforms at VEGFR2 in respect to ligand binding and downstream signalling. To understand how VEGF-A isoforms have distinct signalling despite similar affinities for VEGFR2, this review re-evaluates the typical classification of these isoforms relative to the prototypical, “pro-angiogenic” VEGF165a. We also examine the molecular mechanisms underpinning the regulation of VEGF-A isoform signalling and the importance of interactions with other membrane and extracellular matrix proteins. As approved therapeutics targeting the VEGF-A/VEGFR signalling axis largely lack long-term efficacy, understanding these isoform-specific mechanisms could aid future drug discovery efforts targeting VEGF receptor pharmacology.
Collapse
Affiliation(s)
- Chloe J Peach
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Viviane W Mignone
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
- CAPES-University of Nottingham Programme in Drug Discovery, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Maria Augusta Arruda
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
- CAPES-University of Nottingham Programme in Drug Discovery, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Diana C Alcobia
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Laura E Kilpatrick
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, UK.
| |
Collapse
|
29
|
Hatcher JM, Wu G, Zeng C, Zhu J, Meng F, Patel S, Wang W, Ficarro SB, Leggett AL, Powell CE, Marto JA, Zhang K, Ngo JCK, Fu XD, Zhang T, Gray NS. SRPKIN-1: A Covalent SRPK1/2 Inhibitor that Potently Converts VEGF from Pro-angiogenic to Anti-angiogenic Isoform. Cell Chem Biol 2018; 25:460-470.e6. [PMID: 29478907 PMCID: PMC5973797 DOI: 10.1016/j.chembiol.2018.01.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/03/2017] [Accepted: 01/26/2018] [Indexed: 01/07/2023]
Abstract
The SRPK family of kinases regulates pre-mRNA splicing by phosphorylating serine/arginine (SR)-rich splicing factors, signals splicing control in response to extracellular stimuli, and contributes to tumorigenesis, suggesting that these splicing kinases are potential therapeutic targets. Here, we report the development of the first irreversible SRPK inhibitor, SRPKIN-1, which is also the first kinase inhibitor that forms a covalent bond with a tyrosine phenol group in the ATP-binding pocket. Kinome-wide profiling demonstrates its selectivity for SRPK1/2, and SRPKIN-1 attenuates SR protein phosphorylation at submicromolar concentrations. Vascular endothelial growth factor (VEGF) is a known target for SRPK-regulated splicing and, relative to the first-generation SRPK inhibitor SRPIN340 or small interfering RNA-mediated SRPK knockdown, SRPKIN-1 is more potent in converting the pro-angiogenic VEGF-A165a to the anti-angiogenic VEGF-A165b isoform and in blocking laser-induced neovascularization in a murine retinal model. These findings encourage further development of SRPK inhibitors for treatment of age-related macular degeneration.
Collapse
Affiliation(s)
- John M. Hatcher
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Guowei Wu
- Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chuyue Zeng
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region NA, China
| | - Jie Zhu
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA,Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Fan Meng
- Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sherrina Patel
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenqiu Wang
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Scott B. Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Oncologic Pathology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alan L. Leggett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Chelsea E. Powell
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Jarrod A. Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Oncologic Pathology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kang Zhang
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jacky Chi Ki Ngo
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region NA, China
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA,Correspondence: (X.-D.F.), (T.Z.), (N.S.G.)
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA,Correspondence: (X.-D.F.), (T.Z.), (N.S.G.)
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA,Correspondence: (X.-D.F.), (T.Z.), (N.S.G.)
| |
Collapse
|
30
|
Touyz RM, Herrmann SMS, Herrmann J. Vascular toxicities with VEGF inhibitor therapies-focus on hypertension and arterial thrombotic events. ACTA ACUST UNITED AC 2018; 12:409-425. [PMID: 29703600 PMCID: PMC6168784 DOI: 10.1016/j.jash.2018.03.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/02/2018] [Accepted: 03/14/2018] [Indexed: 12/21/2022]
Abstract
The vascular endothelial growth factor (VEGF) signaling pathway (VSP) fulfills a cardinal role in endothelial cells and its inhibition has profound cardiovascular impact. This is true not only for the normal vasculature but also for the tumor vasculature when VSP inhibitors are used as anti-angiogenic therapies. Generalized endothelial dysfunction predisposes to vasoconstriction, atherosclerosis, platelet activation, and thrombosis (arterial more than venous). All of these have been reported with VSP inhibitors and collectively give rise to vascular toxicities, the most concerning of which are arterial thromboembolic events (ATE). VSP inhibitors include antibodies, acting extracelluarly on VEGF, such as bevacizumab and tyrosine kinases inhibitors, acting intracellularly on the kinase domain of VEGF receptors, such as sunintib and sorafenib. The addition of bevacizumab and VSP tyrosine kinase inhibitor therapy to the cancer treatment regimen is associated with a 1.5-2.5-fold and 2.3-4.6-fold increase risk of ATEs, respectively. Risk factors for ATEs while on VSP inhibitor therapy include age older than 65 years, previous thromboembolic events, history of atherosclerotic disease, and duration of VSP inhibitor therapy. In clinical practice, hypertension remains the most commonly noted vascular manifestation of VSP inhibition. Optimal blood pressure goals and preferred therapeutic strategies toward reaching these goals are not defined at present. This review summarizes current data on this topic and proposes a more intensive management approach to patients undergoing VSP inhibitor therapy including Systolic Blood PRessure Intervention Trial (SPRINT) blood pressure goals, pleiotropic vasoprotective agents such as angiotensin converting enzyme inhibitors, amlodipine, and carvedilol, high-dose statin therapy, and aspirin.
Collapse
Affiliation(s)
- Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Sandra M S Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
31
|
Wang Z, Liu T. Placental growth factor signaling regulates isoform splicing of vascular endothelial growth factor A in the control of lung cancer cell metastasis. Mol Cell Biochem 2017; 439:163-169. [PMID: 28861767 DOI: 10.1007/s11010-017-3145-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/02/2017] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF) family members play critical and complex roles in the regulation of cancer vascularization and metastasis. The exact molecular control of lung cancer metastasis by VEGF family members is not completely understood. Here, we showed that specimens from non-small cell lung cancer (NSCLC) contained significantly higher levels of placental growth factor (PlGF) than paired non-cancer tissue (p < 0.05, N = 25). Moreover, higher levels of PlGF were detected in NSCLC specimens from the patients who had distal metastases than those who had not. High-PlGF levels appeared to be associated with poor patient survival. In vitro, PlGF dose-dependently increased the ratio of pro-angiogenic VEGF isoform (VEGF165) versus anti-angiogenic VEGF isoform (VEGF165b), seemingly through induction of expression of splicing regulatory factor SRp40, resulting in the enhancement of the cancer cell metastatic potential. Higher levels of SRp40 were detected in NSCLC specimens, compared to paired non-cancer tissue (p < 0.05, N = 25). Finally, a strong correlation was detected between the levels of PlGF and SRp40 in NSCLC specimens (r = 0.83, p < 0.0001, N = 25). Together, these data suggest that PlGF may increase NSCLC metastasis through SRp40-mediated mRNA splicing of VEGF.
Collapse
Affiliation(s)
- Zanfeng Wang
- Department of Respiratory Medicine, First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Shenyang, 110001, China.
| | - Tingwei Liu
- Department of Respiratory Medicine, First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Shenyang, 110001, China
| |
Collapse
|
32
|
Dynamics and implications of circulating anti-angiogenic VEGF-A 165b isoform in patients with ST-elevation myocardial infarction. Sci Rep 2017; 7:9962. [PMID: 28855597 PMCID: PMC5577291 DOI: 10.1038/s41598-017-10505-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis is crucial to restore microvascular perfusion in the jeopardized myocardium in the weeks following reperfused ST-segment elevation myocardial infarction (STEMI). (VEGF)-A165b, an anti-angiogenic factor, has been identified as a regulator of vascularization; however, it has not been previously implicated in acute myocardial infarction. We sought to investigate the dynamics of circulating VEGF-A165b and its association with cardiac magnetic resonance-derived infarct size and left ventricular ejection fraction (LVEF). 50 STEMI patients and 23 controls were included. Compared with control individuals, serum VEGF-A165b was elevated in STEMI patients prior to primary percutaneous coronary intervention (PCI). Following PCI, serum VEGF-A165b increased further, reaching a maximum level at 24 h and decreased one month after reperfusion. VEGF-A165b levels at 24 h were associated with a large infarct size and inversely related to LVEF. VEGF-A165b expression was increased in myocardial infarct areas from patients with previous history of AMI. An ex vivo assay using serum from STEMI patients showed that neutralization of VEGF-A165b increased tubulogenesis. Overall, the study suggests that VEGF-A165b might play a deleterious role after AMI as an inhibitor of angiogenesis in the myocardium. Accordingly, neutralization of VEGF-A165b could represent a novel pro-angiogenic therapy for reperfusion of myocardium in STEMI.
Collapse
|
33
|
Pruszko M, Milano E, Forcato M, Donzelli S, Ganci F, Di Agostino S, De Panfilis S, Fazi F, Bates DO, Bicciato S, Zylicz M, Zylicz A, Blandino G, Fontemaggi G. The mutant p53-ID4 complex controls VEGFA isoforms by recruiting lncRNA MALAT1. EMBO Rep 2017; 18:1331-1351. [PMID: 28652379 PMCID: PMC5538427 DOI: 10.15252/embr.201643370] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022] Open
Abstract
The abundant, nuclear-retained, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been associated with a poorly differentiated and aggressive phenotype of mammary carcinomas. This long non-coding RNA (lncRNA) localizes to nuclear speckles, where it interacts with a subset of splicing factors and modulates their activity. In this study, we demonstrate that oncogenic splicing factor SRSF1 bridges MALAT1 to mutant p53 and ID4 proteins in breast cancer cells. Mutant p53 and ID4 delocalize MALAT1 from nuclear speckles and favor its association with chromatin. This enables aberrant recruitment of MALAT1 on VEGFA pre-mRNA and modulation of VEGFA isoforms expression. Interestingly, VEGFA-dependent expression signatures associate with ID4 expression specifically in basal-like breast cancers carrying TP53 mutations. Our results highlight a key role for MALAT1 in control of VEGFA isoforms expression in breast cancer cells expressing gain-of-function mutant p53 and ID4 proteins.
Collapse
Affiliation(s)
- Magdalena Pruszko
- Department of Molecular Biology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, PAS, Warsaw, Poland
| | - Elisa Milano
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Mattia Forcato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Federica Ganci
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Silvia Di Agostino
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Simone De Panfilis
- Centre for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - David O Bates
- Division of Cancer and Stem Cells, Cancer Biology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Maciej Zylicz
- Department of Molecular Biology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Alicja Zylicz
- Department of Molecular Biology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Giulia Fontemaggi
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| |
Collapse
|
34
|
Ye X, Abou-Rayyah Y, Bischoff J, Ritchie A, Sebire NJ, Watts P, Churchill AJ, Bates DO. Altered ratios of pro- and anti-angiogenic VEGF-A variants and pericyte expression of DLL4 disrupt vascular maturation in infantile haemangioma. J Pathol 2017; 239:139-51. [PMID: 26957058 PMCID: PMC4869683 DOI: 10.1002/path.4715] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 02/27/2016] [Accepted: 03/03/2016] [Indexed: 12/26/2022]
Abstract
Infantile haemangioma (IH), the most common neoplasm in infants, is a slowly resolving vascular tumour. Vascular endothelial growth factor A (VEGF‐A), which consists of both the pro‐ and anti‐angiogenic variants, contributes to the pathogenesis of IH. However, the roles of different VEGF‐A variants in IH progression and its spontaneous involution is unknown. Using patient‐derived cells and surgical specimens, we showed that the relative level of VEGF‐A165b was increased in the involuting phase of IH and the relative change in VEGF‐A isoforms may be dependent on endothelial differentiation of IH stem cells. VEGFR signalling regulated IH cell functions and VEGF‐A165b inhibited cell proliferation and the angiogenic potential of IH endothelial cells in vitro and in vivo. The inhibition of angiogenesis by VEGF‐A165b was associated with the extent of VEGF receptor 2 (VEGFR2) activation and degradation and Delta‐like ligand 4 (DLL4) expression. These results indicate that VEGF‐A variants can be regulated by cell differentiation and are involved in IH progression. We also demonstrated that DLL4 expression was not exclusive to the endothelium in IH but was also present in pericytes, where the expression of VEGFR2 is absent, suggesting that pericyte‐derived DLL4 may prevent sprouting during involution, independently of VEGFR2. Angiogenesis in IH therefore appears to be controlled by DLL4 within the endothelium in a VEGF‐A isoform‐dependent manner, and in perivascular cells in a VEGF‐independent manner. The contribution of VEGF‐A isoforms to disease progression also indicates that IH may be associated with altered splicing. © 2016 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)
- Xi Ye
- Ophthalmology Unit, School of Clinical Sciences, University of Bristol, UK.,Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, UK
| | | | - Joyce Bischoff
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, MA, USA
| | - Alison Ritchie
- Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, UK
| | - Neil J Sebire
- Histopathology, Great Ormond Street Hospital, London, UK
| | | | - Amanda J Churchill
- Ophthalmology Unit, School of Clinical Sciences, University of Bristol, UK
| | - David O Bates
- Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, UK
| |
Collapse
|
35
|
Francis JH, Kim J, Lin A, Folberg R, Iyer S, Abramson DH. Growth of Uveal Melanoma following Intravitreal Bevacizumab. Ocul Oncol Pathol 2016; 3:117-121. [PMID: 28868282 DOI: 10.1159/000450859] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/13/2016] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Typically treatment of large melanomas (by Collaborative Ocular Melanoma Study criteria) is restricted to enucleation, due to size constraints for plaque brachytherapy. Because primary and metastatic uveal melanoma cells are inhibited by bevacizumab (an anti-vascular endothelial growth factor), this prospective study evaluated the impact of intravitreal bevacizumab on large uveal melanomas that were destined for enucleation. Size reduction by bevacizumab would potentially salvage these eyes by making them eligible for treatment with plaque brachytherapy. PROCEDURES Two patients with large uveal melanoma were each treated with one intravitreous injection of bevacizumab (1.25 mg/0.05 mL). RESULTS Both tumors displayed paradoxical growth 1 week following the injection, with confirmed growth 1 week later (increase from baseline of 1.1 mm in one eye and 3.1 mm in the other eye). Both eyes were enucleated and monosomy 3 and vasculogenic mimicry patterns were identified in both tumors. At 9 years follow-up, both patients were alive and metastasis free. CONCLUSION These patients demonstrate that neoadjuvant intravitreous bevacizumab does not decrease the size of large uveal melanomas and may, in fact, result in their paradoxical growth. This observation supports a cautious approach in the use of intravitreous bevacizumab for uveal melanoma, particularly in the neoadjuvant setting.
Collapse
Affiliation(s)
- Jasmine H Francis
- Ophthalmic Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, NY.,Department of Ophthalmology, Weill Cornell Medical College, New York, NY
| | - Jonathan Kim
- Department of Ophthalmology, University of Southern California, Los Angeles, CA, USA
| | - Amy Lin
- University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| | - Robert Folberg
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Saipriya Iyer
- Ophthalmic Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - David H Abramson
- Ophthalmic Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, NY.,Department of Ophthalmology, Weill Cornell Medical College, New York, NY
| |
Collapse
|
36
|
NAGASAKI MASAHIRO, KONDO SEIJI, MUKUDAI YOSHIKI, KAMATANI TAKAAKI, AKIZUKI AYAKO, YASO ATSUSHI, SHIMANE TOSHIKAZU, SHIROTA TATSUO. Clinicopathological implications of vascular endothelial growth factor 165b expression in oral squamous cell carcinoma stroma. Oncol Rep 2016; 36:573-81. [DOI: 10.3892/or.2016.4826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/05/2016] [Indexed: 11/05/2022] Open
|
37
|
Zhao M, Xie WK, Bai YJ, Huang LZ, Wang B, Liang JH, Yin H, Li XX, Shi X. Expression of Total Vascular Endothelial Growth Factor and the Anti-angiogenic VEGF 165 b Isoform in the Vitreous of Patients with Retinopathy of Prematurity. Chin Med J (Engl) 2016; 128:2505-9. [PMID: 26365970 PMCID: PMC4725572 DOI: 10.4103/0366-6999.164937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background: This study was to examine the expression of total vascular endothelial growth factor (VEGF) and the anti-angiogenic VEGF165b isoform in the vitreous body of retinopathy of prematurity (ROP) patients, and to further study the role of the VEGF splicing in the development of ROP. Methods: This was a prospective clinical laboratory investigation study. All patients enrolled received standard ophthalmic examination with stage 4 ROP that required vitrectomy to collect the vitreous samples. The control samples were from congenital cataract patients. The expression of total VEGF and the anti-angiogenic VEGF165b were measured by enzyme-linked immunosorbent assay. Results were analyzed statistically using nonparametric tests. Results: The total VEGF level was markedly elevated in ROP samples while VEGF165b was markedly decreased compared to control group. The relative protein expression level of VEGF165b isoform was significantly decreased in ROP patients which were correlated with the ischemia-induced neovascularization. Conclusions: There was a switch of VEGF splicing from anti-angiogenic to pro-angiogenic family in ROP patients. A specific inhibitor that more selectively targets VEGF165and controls the VEGF splicing between pro- and anti-angiogenic families might be a more effective therapy for ROP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xuan Shi
- Department of Ophthalmology, Peking University People's Hospital, Beijing 100044; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044; Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
| |
Collapse
|
38
|
Zhao YY, Li YG, Yan BQ, Liu ZZ, Qin GT, Sun YJ. Expression of vascular endothelial growth factor 165b in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2016; 24:355-361. [DOI: 10.11569/wcjd.v24.i3.355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To detect the expression of vascular endothelial growth factor 165b (VEGF165b) in hepatocellular carcinoma (HCC), and to investigate the relationship between VEGF165b and HCC.
METHODS: Expression of VEGF165b protein in 28 HCC specimens and 30 normal liver tissue specimens was detected by immunohistochemistry. The expression of VEGF165 and VEGF165b mRNAs was detected by RT-PCR. The expression of VEGF165, VEGF165b, FAK and P-Akt proteins in HCC and normal liver tissues was detected by Western blot.
RESULTS: The positive rate of VEGF165b protein expression in normal liver tissues was significantly higher than that in HCC tissues [96.67% (29/30) vs 21.4% (6/28), P < 0.05]. VEGF165b mRNA and protein expression in HCC tissues was significantly lower than that in normal liver tissues (P < 0.01). The expression of VEGF165 mRNA and protein in HCC tissues was significantly higher than that in normal liver tissues (P < 0.01). The expression of FAK and P-Akt proteins in HCC tissues was significantly higher than that in normal liver tissues (P < 0.01).
CONCLUSION: The expression of VEGF165b in HCC tissues is significantly lower than that in normal liver tissues, and the expression of VEGF165, FAK and P-Akt in HCC tissues is significantly higher than that in normal liver tissues. These findings suggest that VEGF165b may be related to the occurrence and development of HCC possibly by inhibiting the expression of VEGF165, FAK and P-Akt and their effects on angiogenesis and tumor growth.
Collapse
|
39
|
Amadio M, Govoni S, Pascale A. Targeting VEGF in eye neovascularization: What's new? Pharmacol Res 2016; 103:253-69. [DOI: 10.1016/j.phrs.2015.11.027] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/19/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
|
40
|
Abstract
Much of the knowledge we have gained into the development of pathological ocular angiogenesis has come from the development of in vivo models that enable functional assessment of key components of signaling pathways in disease progression. Indeed, rodent models have facilitated identification of several therapeutics that target pathological angiogenesis. Two of the most widely used rodent models of oxygen induced retinopathy (OIR), Smith's mouse model and Penn's rat model reproducibly induce neovascularization reminiscent of the disease retinopathy of prematurity (ROP). In this chapter we discuss development of ROP in humans and compare features with that of the rat and mouse models, focusing both on the benefits and caveats of using such models. Furthermore, we discuss in detail the methodology of both procedures and discuss the importance of various features of the model.
Collapse
Affiliation(s)
- Melissa V Gammons
- MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, C Floor, Pope Building, University Park, Nottingham, NG2 7UH, UK.
| |
Collapse
|
41
|
Sargent KM, Clopton DT, Lu N, Pohlmeier WE, Cupp AS. VEGFA splicing: divergent isoforms regulate spermatogonial stem cell maintenance. Cell Tissue Res 2015; 363:31-45. [PMID: 26553653 DOI: 10.1007/s00441-015-2297-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/24/2015] [Indexed: 12/22/2022]
Abstract
Despite being well-known for regulating angiogenesis in both normal and tumorigenic environments, vascular endothelial growth factor A (VEGFA) has been recently implicated in male fertility, namely in the maintenance of spermatogonial stem cells (SSC). The VEGFA gene can be spliced into multiple distinct isoforms that are either angiogenic or antiangiogenic in nature. Although studies have demonstrated the alternative splicing of VEGFA, including the divergent roles of the two isoform family types, many investigations do not differentiate between them. Data concerning VEGFA in the mammalian testis are limited, but the various angiogenic isoforms appear to promote seminiferous cord formation and to form a gradient across which cells may migrate. Treatment with either antiangiogenic isoforms of VEGFA or with inhibitors to angiogenic signaling impair these processes. Serendipitously, expression of KDR, the primary receptor for both types of VEGFA isoforms, was observed on male germ cells. These findings led to further investigation of the way that VEGFA elicits avascular functions within testes. Following treatment of donor perinatal male mice with either antiangiogenic VEGFA165b or angiogenic VEGFA164 isoforms, seminiferous tubules were less colonized following transplantation with cells from VEGFA165b-treated donors. Thus, VEGFA165b and possibly other antiangiogenic isoforms of VEGFA reduce SSC number either by promoting premature differentiation, inducing cell death, or by preventing SSC formation. Thus, angiogenic isoforms of VEGFA are hypothesized to promote SSC self-renewal, and the divergent isoforms are thought to balance one another to maintain SSC homeostasis in vivo.
Collapse
Affiliation(s)
- Kevin M Sargent
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - Debra T Clopton
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - Ningxia Lu
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - William E Pohlmeier
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA.
| |
Collapse
|
42
|
Wang L, Shan M, Liu Y, Yang F, Qi H, Zhou L, Qiu L, Li Y. miR-205 suppresses the proliferative and migratory capacity of human osteosarcoma Mg-63 cells by targeting VEGFA. Onco Targets Ther 2015; 8:2635-42. [PMID: 26396534 PMCID: PMC4577256 DOI: 10.2147/ott.s80088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary bone malignancy in children and young adults. MiR-205 has been reported to be negatively correlated with the proliferation and metastasis of many types of cancer, while its effects on the malignant phenotype of OS are unclear. METHODS Using TaqMan RT polymerase chain reaction analysis, we firstly explored the expression of miR-205 in a panel of OS cell lines. As the expression of miR-205 was significantly decreased in these cell lines, we sought to compensate for its loss by transfection of exogenous miR-205 mimic into MG-63 cells. To further understand the role of miR-205 in OS, we investigated the effects of miR-205 on the proliferation, migration, and invasion of MG-63 cells, and further explored the mechanisms that might be involved. RESULTS We found that miR-205 was consistently suppressed in OS cells when compared with the normal human osteoblast (NHOst) cell line. Restored expression of miR-205 in the OS (MG-63) cell line significantly inhibited cell proliferation, migration, and invasion. Moreover, bioinformatic prediction suggested that vascular endothelial growth factor A (VEGFA) was the target oncogene for miR-205 in OS cells. Further quantitative RT polymerase chain reaction and Western blot assays identified that overexpression of miR-205 suppressed expression of VEGFA mRNA and protein. Restored expression of VEGFA in MG-63 cells previously treated with miR-205 mimic could partially abolish miR-205-mediated suppression of proliferation and invasion of these cells. CONCLUSION Collectively, these data suggest that miR-205 might function as a tumor suppressor in OS by, at least partially, targeting VEGFA.
Collapse
Affiliation(s)
- Li Wang
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Minhong Shan
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Yang Liu
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Fengyi Yang
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Hongxia Qi
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Lijuan Zhou
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Lirong Qiu
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Yanshuang Li
- Department of Operating Room 1, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
43
|
Burgoyne JR, Rudyk O, Cho HJ, Prysyazhna O, Hathaway N, Weeks A, Evans R, Ng T, Schröder K, Brandes RP, Shah AM, Eaton P. Deficient angiogenesis in redox-dead Cys17Ser PKARIα knock-in mice. Nat Commun 2015; 6:7920. [PMID: 26258640 DOI: 10.1038/ncomms8920] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 06/24/2015] [Indexed: 02/05/2023] Open
Abstract
Angiogenesis is essential for tissue development, wound healing and tissue perfusion, with its dysregulation linked to tumorigenesis, rheumatoid arthritis and heart disease. Here we show that pro-angiogenic stimuli couple to NADPH oxidase-dependent generation of oxidants that catalyse an activating intermolecular-disulphide between regulatory-RIα subunits of protein kinase A (PKA), which stimulates PKA-dependent ERK signalling. This is crucial to blood vessel growth as 'redox-dead' Cys17Ser RIα knock-in mice fully resistant to PKA disulphide-activation have deficient angiogenesis in models of hind limb ischaemia and tumour-implant growth. Disulphide-activation of PKA represents a new therapeutic target in diseases with aberrant angiogenesis.
Collapse
Affiliation(s)
- Joseph R Burgoyne
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Olena Rudyk
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Hyun-Ju Cho
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Oleksandra Prysyazhna
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Natasha Hathaway
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Amanda Weeks
- King's College London, Division of Imaging Sciences, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Rachel Evans
- King's College London, Division of Cancer Studies, 2nd Floor, New Hunt's House, Guy's Medical School Campus, London SE1 1UL, UK
| | - Tony Ng
- King's College London, Division of Cancer Studies, 2nd Floor, New Hunt's House, Guy's Medical School Campus, London SE1 1UL, UK
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Ajay M Shah
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The James Black Centre, Denmark Hill Campus, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Philip Eaton
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| |
Collapse
|
44
|
Zhao YJ, Han HZ, Liang Y, Shi CZ, Zhu QC, Yang J. Alternative splicing of VEGFA, APP and NUMB genes in colorectal cancer. World J Gastroenterol 2015; 21:6550-60. [PMID: 26074693 PMCID: PMC4458765 DOI: 10.3748/wjg.v21.i21.6550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 02/10/2015] [Accepted: 03/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate alternative splicing in vascular endothelial growth factor A (VEGFA), amyloid beta precursor protein (APP), and Numb homolog (NUMB) in colorectal cancer (CRC). METHODS Real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and PCR-restriction fragment length polymorphism analyses were performed to detect the expression of VEGFA, APP, and NUMB mRNA in 20 CRC tissues and matched adjacent normal tissues, as well as their alternative splicing variants. RESULTS qRT-PCR analysis revealed that the expression of APP, NUMB, and VEGFA165b mRNA were significantly downregulated, while VEGFA mRNA was upregulated, in CRC tissues (all P < 0.05). PCR-restriction fragment length polymorphism analysis revealed that the expression of VEGFA165a/b in CRC tissues was significantly higher than in adjacent normal tissues (P < 0.05). Compared with adjacent normal tissues, the expression of NUMB-PRR(S) in CRC tissues was significantly decreased (P < 0.05), and the expression of NUMB-PRR(L) was increased (P < 0.05). CONCLUSION Alternative splicing of VEGFA, APP, and NUMB may regulate the development of CRC, and represent new targets for its diagnosis, prognosis, and treatment.
Collapse
|
45
|
Bunni J, Shelley-Fraser G, Stevenson K, Oltean S, Salmon A, Harper SJ, Carter JG, Bates DO. Circulating levels of anti-angiogenic VEGF-A isoform (VEGF-Axxxb) in colorectal cancer patients predicts tumour VEGF-A ratios. Am J Cancer Res 2015; 5:2083-2089. [PMID: 26269767 PMCID: PMC4529627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/10/2015] [Indexed: 06/04/2023] Open
Abstract
PURPOSE Bevacizumab as an adjunct to chemotherapy improves survival for some patients with metastatic colorectal cancer. Immunohistochemical staining of samples from the registration ECOG E3200 trial of bevacizumab with FOLFOX demonstrated that only patients with carcinomas expressing low levels of VEGF-A165b, an anti-angiogenic splice variant of the Vascular Endothelial Growth Factor family of proteins, benefited from bevacizumab treatment. To identify a more useful biomarker of response we tested the hypothesis that circulating VEGF-A165b levels correlate with immunohistochemical staining. EXPERIMENTAL DESIGN 17 patients with biopsy proven colorectal adenocarcinoma had pre-operative blood samples drawn. They underwent resection and had post-resection blood drawn. The plasma was analysed for levels of VEGF-Axxxb using enzyme-linked immunosorbent assay (ELISA) and the tumour blocks stained for VEGF-Axxxb and pan-VEGF-A. The normalised ratio of VEGF-Axxxb expression to that of panVEGF-A expression scored by IHC was calculated and correlated with plasma VEGF-A165b levels. RESULTS Plasma levels of VEGF-Axxxb significantly correlated with the VEGF-Axxxb:panVEGF-A ratio (r=0.594, P<0.02) in colorectal cancers. Median plasma VEGF-Axxxb levels were 151 pg/ml. The mean (1.5±0.17) and median, IQR (1.8, 1-2) IHC scores of the patients with greater than median plasma VEGF-Axxxb were significantly greater than those with less than median plasma VEGF-Axxxb levels (mean ± SEM=0.85±10.12, median, IQR=1, 0.54-1). CONCLUSION These results suggest that plasma VEGF-Axxxb levels could be an effective biomarker of response to Bevacizumab. These results indicate that a prospective trial is warranted to explore the use of plasma VEGF-Axxxb levels to stratify patients for colorectal cancer treatment by bevacizumab.
Collapse
Affiliation(s)
- John Bunni
- Department of Physiology and Pharmacology, Microvascular Research Laboratories, University of Bristol Bristol, United Kingdom
| | | | - Kirsty Stevenson
- Department of Pathology, Bristol Royal Infirmary Bristol, United Kingdom
| | - Sebastian Oltean
- Department of Physiology and Pharmacology, Microvascular Research Laboratories, University of Bristol Bristol, United Kingdom
| | - Andy Salmon
- Department of Physiology and Pharmacology, Microvascular Research Laboratories, University of Bristol Bristol, United Kingdom
| | - Steven J Harper
- Department of Physiology and Pharmacology, Microvascular Research Laboratories, University of Bristol Bristol, United Kingdom
| | - James G Carter
- Department of Physiology and Pharmacology, Microvascular Research Laboratories, University of Bristol Bristol, United Kingdom
| | - David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Bristol, United Kingdom
| |
Collapse
|
46
|
Guyot M, Pagès G. VEGF Splicing and the Role of VEGF Splice Variants: From Physiological-Pathological Conditions to Specific Pre-mRNA Splicing. Methods Mol Biol 2015; 1332:3-23. [PMID: 26285742 DOI: 10.1007/978-1-4939-2917-7_1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During this past decade, the vascular endothelial growth factor (VEGF) pathway has been extensively studied. VEGF is a paradigm of molecular regulation since its expression is controlled at all possible steps including transcription, mRNA stability, translation, and pre-mRNA splicing. The latter form of molecular regulation is probably the least studied. This field has been neglected; yet different forms of VEGF with different sizes and different physiological properties issued from alternative splicing have been described a long time ago. Recently a new level of complexity was added to the field of splicing of VEGF pre-mRNA. Whereas thousands of publications have described VEGF as a pro-angiogenic factor, an alternative splicing event generates specific anti-angiogenic forms of VEGF that only differ from the others by a modification in the last six amino acids of the protein. According to the scientists who discovered these isoforms, which are indistinguishable from the pro-angiogenic ones with pan VEGF antibodies, some of the literature on VEGF is at least inexact if not completely false. Moreover, the presence of anti-angiogenic forms of VEGF may explain the disappointing efficacy of anti-VEGF therapies on the overall survival of patients with different forms of cancers and with wet age-related macular degeneration. This review focuses on the existence of the different alternative splice variants of VEGF and the molecular mechanisms associated with their expression and function.
Collapse
Affiliation(s)
- Mélanie Guyot
- Institute for Research on Cancer and Aging of Nice (IRCAN), University of Nice Sophia Antipolis, Centre Antoine Lacassagne 33 Avenue de Valombrose, UMR CNRS 7284/INSERM U 1081, Nice, 06189, France
| | | |
Collapse
|
47
|
Oltean S, Qiu Y, Ferguson JK, Stevens M, Neal C, Russell A, Kaura A, Arkill KP, Harris K, Symonds C, Lacey K, Wijeyaratne L, Gammons M, Wylie E, Hulse RP, Alsop C, Cope G, Damodaran G, Betteridge KB, Ramnath R, Satchell SC, Foster RR, Ballmer-Hofer K, Donaldson LF, Barratt J, Baelde HJ, Harper SJ, Bates DO, Salmon AHJ. Vascular Endothelial Growth Factor-A165b Is Protective and Restores Endothelial Glycocalyx in Diabetic Nephropathy. J Am Soc Nephrol 2014; 26:1889-904. [PMID: 25542969 DOI: 10.1681/asn.2014040350] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 10/15/2014] [Indexed: 01/11/2023] Open
Abstract
Diabetic nephropathy is the leading cause of ESRD in high-income countries and a growing problem across the world. Vascular endothelial growth factor-A (VEGF-A) is thought to be a critical mediator of vascular dysfunction in diabetic nephropathy, yet VEGF-A knockout and overexpression of angiogenic VEGF-A isoforms each worsen diabetic nephropathy. We examined the vasculoprotective effects of the VEGF-A isoform VEGF-A165b in diabetic nephropathy. Renal expression of VEGF-A165b mRNA was upregulated in diabetic individuals with well preserved kidney function, but not in those with progressive disease. Reproducing this VEGF-A165b upregulation in mouse podocytes in vivo prevented functional and histologic abnormalities in diabetic nephropathy. Biweekly systemic injections of recombinant human VEGF-A165b reduced features of diabetic nephropathy when initiated during early or advanced nephropathy in a model of type 1 diabetes and when initiated during early nephropathy in a model of type 2 diabetes. VEGF-A165b normalized glomerular permeability through phosphorylation of VEGF receptor 2 in glomerular endothelial cells, and reversed diabetes-induced damage to the glomerular endothelial glycocalyx. VEGF-A165b also improved the permeability function of isolated diabetic human glomeruli. These results show that VEGF-A165b acts via the endothelium to protect blood vessels and ameliorate diabetic nephropathy.
Collapse
Affiliation(s)
| | - Yan Qiu
- School of Physiology and Pharmacology and
| | | | | | - Chris Neal
- School of Physiology and Pharmacology and
| | | | - Amit Kaura
- School of Physiology and Pharmacology and
| | | | | | | | | | | | | | - Emma Wylie
- School of Physiology and Pharmacology and Academic Renal Unit, School of Clinical Science, University of Bristol, Bristol, United Kingdom
| | | | | | - George Cope
- Academic Renal Unit, School of Clinical Science, University of Bristol, Bristol, United Kingdom
| | | | | | - Raina Ramnath
- Academic Renal Unit, School of Clinical Science, University of Bristol, Bristol, United Kingdom
| | - Simon C Satchell
- Academic Renal Unit, School of Clinical Science, University of Bristol, Bristol, United Kingdom
| | - Rebecca R Foster
- Academic Renal Unit, School of Clinical Science, University of Bristol, Bristol, United Kingdom
| | - Kurt Ballmer-Hofer
- Biomolecular Research, Molecular Cell Biology, Paul Scherrer Institut, Villigen, Switzerland
| | - Lucy F Donaldson
- School of Physiology and Pharmacology and School of Life Sciences and
| | - Jonathan Barratt
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom; and
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - David O Bates
- Cancer Biology, Division of Oncology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Andrew H J Salmon
- School of Physiology and Pharmacology and Academic Renal Unit, School of Clinical Science, University of Bristol, Bristol, United Kingdom;
| |
Collapse
|
48
|
Orief YI, Karkor TAE, Aly Saleh H, El Hadidy AS, Badr N. Comparative evaluation of vascular endothelial growth factor-A expression in pre-ovulatory follicular fluid in normogonadotrophic and endometriotic patients undergoing assisted reproductive techniques. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2014. [DOI: 10.1016/j.mefs.2013.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
49
|
Mavrou A, Brakspear K, Hamdollah-Zadeh M, Damodaran G, Babaei-Jadidi R, Oxley J, Gillatt DA, Ladomery MR, Harper SJ, Bates DO, Oltean S. Serine-arginine protein kinase 1 (SRPK1) inhibition as a potential novel targeted therapeutic strategy in prostate cancer. Oncogene 2014; 34:4311-9. [PMID: 25381816 PMCID: PMC4351909 DOI: 10.1038/onc.2014.360] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/31/2014] [Accepted: 09/16/2014] [Indexed: 12/11/2022]
Abstract
Angiogenesis is required for tumour growth and is induced principally by VEGF-A. VEGF-A pre-mRNA is alternatively spliced at the terminal exon to produce two families of isoforms, pro- and anti-angiogenic, only the former of which is upregulated in prostate cancer. In renal epithelial cells and colon cancer cells, the choice of VEGF splice isoforms is controlled by the splicing factor SRSF1, phosphorylated by SRPK1. Immunohistochemistry staining of human samples revealed a significant increase in SRPK1 expression both in prostate intra-epithelial neoplasia lesions as well as malignant adenocarcinoma compared to benign prostate tissue. We therefore tested the hypothesis that the selective upregulation of pro-angiogenic VEGF in prostate cancer may be under the control of SRPK1 activity. A switch in the expression of VEGF165 towards the anti-angiogenic splice isoform, VEGF165b, was seen in PC-3 cells with SRPK1 knock-down (KD). PC-3 SRPK1-KD cells resulted in tumours that grew more slowly in xenografts, with decreased microvessel density. No effect was seen as a result of SRPK1-KD on growth, proliferation, migration and invasion capabilities of PC-3 cells in vitro. Small molecule inhibitors of SRPK1 switched splicing towards the anti-angiogenic isoform VEGF165b in PC3 cells and decreased tumour growth when administered intraperitoneally in an orthotopic mouse model of prostate cancer. Our study suggests that modulation of SRPK1 and subsequent inhibition of tumour angiogenesis by regulation of VEGF splicing can alter prostate tumour growth and supports further studies into the use of SRPK1 inhibition as a potential anti-angiogenic therapy in prostate cancer.
Collapse
Affiliation(s)
- A Mavrou
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - K Brakspear
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - M Hamdollah-Zadeh
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - G Damodaran
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - R Babaei-Jadidi
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - J Oxley
- Department of Cellular Pathology, North Bristol NHS Trust, Bristol, UK
| | - D A Gillatt
- Department of Urological Sciences, North Bristol NHS Trust, Bristol, UK
| | - M R Ladomery
- Centre for Research in Bioscience, Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK
| | - S J Harper
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - D O Bates
- 1] School of Physiology and Pharmacology, University of Bristol, Bristol, UK [2] Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - S Oltean
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| |
Collapse
|
50
|
Carter JG, Gammons MVR, Damodaran G, Churchill AJ, Harper SJ, Bates DO. The carboxyl terminus of VEGF-A is a potential target for anti-angiogenic therapy. Angiogenesis 2014; 18:23-30. [PMID: 25274272 PMCID: PMC4280485 DOI: 10.1007/s10456-014-9444-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 09/10/2014] [Indexed: 11/25/2022]
Abstract
Anti-VEGF-A therapy has become a mainstay of treatment for ocular neovascularisation and in cancer; however, their effectiveness is not universal, in some cases only benefiting a minority of patients. Anti-VEGF-A therapies bind and block both pro-angiogenic VEGF-Axxx and the partial agonist VEGF-Axxxb isoforms, but their anti-angiogenic benefit only comes about from targeting the pro-angiogenic isoforms. Therefore, antibodies that exclusively target the pro-angiogenic isoforms may be more effective. To determine whether C-terminal-targeted antibodies could inhibit angiogenesis, we generated a polyclonal antibody to the last nine amino acids of VEGF-A165 and tested it in vitro and in vivo. The exon8a polyclonal antibody (Exon8apab) did not bind VEGF-A165b even at greater than 100-fold excess concentration, and dose dependently inhibited VEGF-A165 induced endothelial migration in vitro at concentrations similar to the VEGF-A antibody fragment ranibizumab. Exon8apab can inhibit tumour growth of LS174t cells implanted in vivo and blood vessel growth in the eye in models of age-related macular degeneration, with equal efficacy to non-selective anti-VEGF-A antibodies. It also showed that it was the VEGF-Axxx levels specifically that were upregulated in plasma from patients with proliferative diabetic retinopathy. These results suggest that VEGF-A165-specific antibodies can be therapeutically useful.
Collapse
Affiliation(s)
- James G. Carter
- Microvascular Research Laboratories, School of Physiology and Pharmacology, University of Bristol, Preclinical Veterinary Sciences Building, Southwell Street, Bristol, BS2 8EJ UK
- Cancer Biology, Queens Medical Centre, University of Nottingham, D Floor West Block, Nottingham, NG7 2UH UK
| | - Melissa V. R. Gammons
- Microvascular Research Laboratories, School of Physiology and Pharmacology, University of Bristol, Preclinical Veterinary Sciences Building, Southwell Street, Bristol, BS2 8EJ UK
| | - Gopinath Damodaran
- Microvascular Research Laboratories, School of Physiology and Pharmacology, University of Bristol, Preclinical Veterinary Sciences Building, Southwell Street, Bristol, BS2 8EJ UK
| | - Amanda J. Churchill
- Microvascular Research Laboratories, School of Physiology and Pharmacology, University of Bristol, Preclinical Veterinary Sciences Building, Southwell Street, Bristol, BS2 8EJ UK
| | - Steven J. Harper
- Microvascular Research Laboratories, School of Physiology and Pharmacology, University of Bristol, Preclinical Veterinary Sciences Building, Southwell Street, Bristol, BS2 8EJ UK
| | - David O. Bates
- Microvascular Research Laboratories, School of Physiology and Pharmacology, University of Bristol, Preclinical Veterinary Sciences Building, Southwell Street, Bristol, BS2 8EJ UK
- Cancer Biology, Queens Medical Centre, University of Nottingham, D Floor West Block, Nottingham, NG7 2UH UK
| |
Collapse
|