251
|
Qiu Y, Ferguson J, Oltean S, Neal CR, Kaura A, Bevan H, Wood E, Sage LM, Lanati S, Nowak DG, Salmon AHJ, Bates D, Harper SJ. Overexpression of VEGF165b in podocytes reduces glomerular permeability. J Am Soc Nephrol 2010; 21:1498-509. [PMID: 20688932 PMCID: PMC3013528 DOI: 10.1681/asn.2009060617] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Accepted: 05/08/2010] [Indexed: 11/03/2022] Open
Abstract
The observation that therapeutic agents targeting vascular endothelial growth factor-A (VEGF-A) associate with renal toxicity suggests that VEGF plays a role in the maintenance of the glomerular filtration barrier. Alternative mRNA splicing produces the VEGF(xxx)b family, which consists of antiangiogenic peptides that reduce permeability and inhibit tumor growth; the contribution of these peptides to normal glomerular function is unknown. Here, we established and characterized heterozygous and homozygous transgenic mice that overexpress VEGF(165)b specifically in podocytes. We confirmed excess production of glomerular VEGF(165)b by reverse transcriptase-PCR, immunohistochemistry, and ELISA in both heterozygous and homozygous animals. Macroscopically, the mice seemed normal up to 18 months of age, unlike the phenotype of transgenic podocyte-specific VEGF(164)-overexpressing mice. Animals overexpressing VEGF(165)b, however, had a significantly reduced normalized glomerular ultrafiltration fraction with accompanying changes in ultrastructure of the glomerular filtration barrier on the vascular side of the glomerular basement membrane. These data highlight the contrasting properties of VEGF splice variants and their impact on glomerular function and phenotype.
Collapse
Affiliation(s)
- Yan Qiu
- Microvascular Research Laboratories, Department Physiology and Pharmacology, Bristol Heart Institute, Preclinical Veterinary School, Southwell Street, Bristol, BS2 8EJ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
252
|
Baltes-Breitwisch MM, Artac RA, Bott RC, McFee RM, Kerl JG, Clopton DT, Cupp AS. Neutralization of vascular endothelial growth factor antiangiogenic isoforms or administration of proangiogenic isoforms stimulates vascular development in the rat testis. Reproduction 2010; 140:319-29. [PMID: 20457593 PMCID: PMC3122080 DOI: 10.1530/rep-09-0456] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Vascular endothelial growth factor A (VEGFA) plays a role in both angiogenesis and seminiferous cord formation, and alternative splicing of the Vegfa gene produces both proangiogenic isoforms and antiangiogenic isoforms (B-isoforms). The objectives of this study were to evaluate the expression of pro- and antiangiogenic isoforms during testis development and to determine the role of VEGFA isoforms in testis morphogenesis. Quantitative RT-PCR determined that Vegfa_165b mRNA was most abundant between embryonic days 13.5 and 16 (E13.5 and 16; P<0.05). Compared with ovarian mRNA levels, Vegfa_120 was more abundant at E13-14 (P<0.05), Vegfa_164 was less abundant at E13 (P<0.05), and Vegfa_165b tended to be less abundant at E13 (P<0.09) in testes. Immunohistochemical staining localized antiangiogenic isoforms to subsets of germ cells at E14-16, and western blot analysis revealed similar protein levels for VEGFA_165B, VEGFA_189B, and VEGFA_206B at this time point. Treatment of E13 organ culture testes with VEGFA_120, VEGFA_164, and an antibody to antiangiogenic isoforms (anti-VEGFAxxxB) resulted in less organized and defined seminiferous cords compared with paired controls. In addition, 50 ng/ml VEGFA_120 and VEGFA_164 treatments increased vascular density in cultured testes by 60 and 48% respectively, and treatment with VEGFAxxxB antibody increased vascular density by 76% in testes (0.5 ng/ml) and 81% in ovaries (5 ng/ml) compared with controls (P<0.05). In conclusion, both pro- and antiangiogenic VEGFA isoforms are involved in the development of vasculature and seminiferous cords in rat testes, and differential expression of these isoforms may be important for normal gonadal development.
Collapse
|
253
|
Magnussen AL, Rennel ES, Hua J, Bevan HS, Long NB, Lehrling C, Gammons M, Floege J, Harper SJ, Agostini HT, Bates DO, Churchill AJ. VEGF-A165b is cytoprotective and antiangiogenic in the retina. Invest Ophthalmol Vis Sci 2010; 51:4273-81. [PMID: 20237249 PMCID: PMC2910648 DOI: 10.1167/iovs.09-4296] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 11/19/2009] [Accepted: 01/16/2010] [Indexed: 01/22/2023] Open
Abstract
PURPOSE A number of key ocular diseases, including diabetic retinopathy and age-related macular degeneration, are characterized by localized areas of epithelial or endothelial damage, which can ultimately result in the growth of fragile new blood vessels, vitreous hemorrhage, and retinal detachment. VEGF-A(165), the principal neovascular agent in ocular angiogenic conditions, is formed by proximal splice site selection in its terminal exon 8. Alternative splicing of this exon results in an antiangiogenic isoform, VEGF-A(165)b, which is downregulated in diabetic retinopathy. Here the authors investigate the antiangiogenic activity of VEGF(165)b and its effect on retinal epithelial and endothelial cell survival. METHODS VEGF-A(165)b was injected intraocularly in a mouse model of retinal neovascularization (oxygen-induced retinopathy [OIR]). Cytotoxicity and cell migration assays were used to determine the effect of VEGF-A(165)b. RESULTS VEGF-A(165)b dose dependently inhibited angiogenesis (IC(50), 12.6 pg/eye) and retinal endothelial migration induced by 1 nM VEGF-A(165) across monolayers in culture (IC(50), 1 nM). However, it also acts as a survival factor for endothelial cells and retinal epithelial cells through VEGFR2 and can stimulate downstream signaling. Furthermore, VEGF-A(165)b injection, while inhibiting neovascular proliferation in the eye, reduced the ischemic insult in OIR (IC(50), 2.6 pg/eye). Unlike bevacizumab, pegaptanib did not interact directly with VEGF-A(165)b. CONCLUSIONS The survival effects of VEGF-A(165)b signaling can protect the retina from ischemic damage. These results suggest that VEGF-A(165)b may be a useful therapeutic agent in ischemia-induced angiogenesis and a cytoprotective agent for retinal pigment epithelial cells.
Collapse
Affiliation(s)
- Anette L. Magnussen
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Emma S. Rennel
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Jing Hua
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Heather S. Bevan
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Nicholas Beazley Long
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | | | - Melissa Gammons
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Juergen Floege
- Division of Nephrology and Clinical Immunology, University Hospital, RWTH University of Aachen, Achen, Germany; and
| | - Steven J. Harper
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | | | - David O. Bates
- From the Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Amanda J. Churchill
- Unit of Ophthalmology, University of Bristol, Bristol Eye Hospital, Bristol, United Kingdom
| |
Collapse
|
254
|
Merdzhanova G, Gout S, Keramidas M, Edmond V, Coll JL, Brambilla C, Brambilla E, Gazzeri S, Eymin B. The transcription factor E2F1 and the SR protein SC35 control the ratio of pro-angiogenic versus antiangiogenic isoforms of vascular endothelial growth factor-A to inhibit neovascularization in vivo. Oncogene 2010; 29:5392-403. [PMID: 20639906 DOI: 10.1038/onc.2010.281] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The transcription factor E2F1 has a crucial role in the control of cell growth and has been shown to regulate neoangiogenesis in a p53-dependent manner through inhibition of activity of the VEGF-A (vascular endothelial growth factor) promoter. Besides being regulated by transcription, VEGF-A is also highly regulated by pre-mRNA alternative splicing, resulting in the expression of several VEGF isoforms with either pro-(VEGF(xxx)) or anti-(VEGF(xxx)b) angiogenic properties. Recently, we identified the SR (Ser-Rich/Arg) protein SC35, a splicing factor, as a new transcriptional target of E2F1. Here, we show that E2F1 downregulates the activity of the VEGF-A promoter in tumour cells independently of p53, leading to a strong decrease in VEGF(xxx) mRNA levels. We further show that, strikingly, E2F1 alters the ratio of pro-VEGF(xxx) versus anti-VEGF(xxx)b angiogenic isoforms, favouring the antiangiogenic isoforms, by a mechanism involving the induction of SC35 expression. Finally, using lung tumour xenografts in nude mice, we provide evidence that E2F1 and SC35 proteins increase the VEGF(165)b/VEGF ratio and decrease tumour neovascularization in vivo. Overall, these findings highlight E2F1 and SC35 as two regulators of the VEGF(xxx)/VEGF(xxx)b angiogenic switch in human cancer cells, a role that could be crucial during tumour progression, as well as in tumour response to antiangiogenic therapies.
Collapse
Affiliation(s)
- G Merdzhanova
- INSERM, U823, Equipe 2 Bases Moléculaires de la Progression des Cancers du Poumon, Grenoble, France
| | | | | | | | | | | | | | | | | |
Collapse
|
255
|
Varet J, Douglas SK, Gilmartin L, Medford ARL, Bates DO, Harper SJ, Millar AB. VEGF in the lung: a role for novel isoforms. Am J Physiol Lung Cell Mol Physiol 2010; 298:L768-L774. [PMID: 20228180 PMCID: PMC2886605 DOI: 10.1152/ajplung.00353.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 03/12/2010] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial cell growth factor (VEGF) is a potent mitogen and permogen that increases in the plasma and decreases in the alveolar space in respiratory diseases such as acute respiratory distress syndrome (ARDS). This observation has led to controversy over the role of this potent molecule in lung physiology and disease. We hypothesized that some of the VEGF previously detected in normal lung may be of the anti-angiogenic family (VEGF(xxx)b) with significant potential effects on VEGF bioactivity. VEGF(xxx)b protein expression was assessed by indirect immunohistochemistry in normal and ARDS tissue. Expression of VEGF(xxx)b was also detected by immunoblotting in normal lung tissue, primary human alveolar type II (ATII) cells, and bronchoalveolar lavage (BAL) fluid in normal subjects and by ELISA in normal, "at risk," and ARDS subjects. The effect of VEGF(165) and VEGF(165)b on both human primary endothelial cells and alveolar epithelial cell proliferation was assessed by [(3)H]thymidine uptake. We found that VEGF(165)b was widely expressed in normal healthy lung tissue but is reduced in ARDS lung. VEGF(121)b and VEGF(165)b were present in whole lung, BAL, and ATII lysate. The proliferative effect of VEGF(165) on both human primary endothelial cells and human alveolar epithelial cells was significantly inhibited by VEGF(165)b (P < 0.01). These data demonstrate that the novel VEGF(xxx)b family members are expressed in normal lung and are reduced in ARDS. A specific functional effect on primary human endothelial and alveolar epithelial cells has also been shown. These data suggest that the VEGF(xxx)b family may have a role in repair after lung injury.
Collapse
Affiliation(s)
- Julia Varet
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Samantha K. Douglas
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
| | - Laura Gilmartin
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Andrew R. L. Medford
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
| | - David O. Bates
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Steven J. Harper
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Ann B. Millar
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
| |
Collapse
|
256
|
Hua J, Spee C, Kase S, Rennel ES, Magnussen AL, Qiu Y, Varey A, Dhayade S, Churchill AJ, Harper SJ, Bates DO, Hinton DR. Recombinant human VEGF165b inhibits experimental choroidal neovascularization. Invest Ophthalmol Vis Sci 2010; 51:4282-8. [PMID: 20237252 DOI: 10.1167/iovs.09-4360] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Vascular endothelial growth factor (VEGF-A) is the principal stimulator of angiogenesis in wet age-related macular degeneration (AMD). However, VEGF-A is generated by alternate splicing into two families, the proangiogenic VEGF-A(xxx) family and the antiangiogenic VEGF-A(xxx)b family. It is the proangiogenic family that is responsible for the blood vessel growth seen in AMD. METHODS To determine the role of antiangiogenic isoforms of VEGF-A as inhibitors of choroidal neovascularization, the authors used a model of laser-induced choroidal neovascularization in the mouse eye and investigated VEGF-A(165)b effects on endothelial cells and VEGFRs in vitro. RESULTS VEGF-A(165)b inhibited VEGF-A(165)-mediated endothelial cell migration with a dose effect similar to that of ranibizumab and bevacizumab and 200-fold more potent than that of pegaptanib. VEGF-A(165)b bound both VEGFR1 and VEGFR2 with affinity similar to that of VEGF-A(165). After laser injury, mice were injected either intraocularly or subcutaneously with recombinant human VEGF-A(165)b. Intraocular injection of rhVEGF-A(165)b gave a pronounced dose-dependent inhibition of fluorescein leakage, with an IC(50) of 16 pg/eye, neovascularization (IC(50), 0.8 pg/eye), and lesion as assessed by histologic staining (IC(50), 8 pg/eye). Subcutaneous administration of 100 microg twice a week also inhibited fluorescein leakage and neovascularization and reduced lesion size. CONCLUSIONS These results show that VEGF-A(165)b is a potent antiangiogenic agent in a mouse model of age-related macular degeneration and suggest that increasing the ratio of antiangiogenic-to-proangiogenic isoforms may be therapeutically effective in this condition.
Collapse
Affiliation(s)
- Jing Hua
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
257
|
Ciura J, Jagodziński PP. Butyrate increases the formation of anti-angiogenic vascular endothelial growth factor variants in human lung microvascular endothelial cells. Mol Biol Rep 2010; 37:3729-34. [PMID: 20213511 DOI: 10.1007/s11033-010-0026-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 02/24/2010] [Indexed: 11/30/2022]
Abstract
The primary transcript of vascular endothelial growth factor (VEGF) can be alternatively spliced and translated to pro-angiogenic and anti-angiogenic VEGF variants. We investigated the effect of sodium butyrate (NaB) on pro-angiogenic and anti-angiogenic VEGF variants production in immortalized human lung microvascular endothelial cells (HLMEC). These cells were cultured in the absence or in the presence of NaB, followed by total RNA and protein isolation. The transcript and protein levels of pro-angiogenic and anti-angiogenic VEGF variants were evaluated by reverse transcription, real-time quantitative PCR and western blot analysis. We found that NaB significantly increased the anti-angiogenic transcript and protein levels of the VEGF 121b, VEGF165b and VEGF189b variants in HLMEC cells. We did not find the pro-angiogenic VEGF189a transcript variant either in control or NaB treated cells. By contrast, the pro-angiogenic VEGF121a and VEGF165a transcript variants were present in HLMEC cells, but their levels were slightly modulated in the cells treated with NaB compared to controls. Since anti-angiogenic VEGF variants inhibit angiogenesis and tumour progression, and NaB is considered an anticancer drug, our findings may have clinical significance.
Collapse
Affiliation(s)
- J Ciura
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 6 Swiecickiego St., 60-781, Poznan, Poland
| | | |
Collapse
|
258
|
Qiu Y, Hoareau-Aveilla C, Oltean S, Harper SJ, Bates DO. The anti-angiogenic isoforms of VEGF in health and disease. Biochem Soc Trans 2009; 37:1207-13. [PMID: 19909248 PMCID: PMC2882696 DOI: 10.1042/bst0371207] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Anti-angiogenic VEGF (vascular endothelial growth factor) isoforms, generated from differential splicing of exon 8, are widely expressed in normal human tissues but down-regulated in cancers and other pathologies associated with abnormal angiogenesis (cancer, diabetic retinopathy, retinal vein occlusion, the Denys-Drash syndrome and pre-eclampsia). Administration of recombinant VEGF(165)b inhibits ocular angiogenesis in mouse models of retinopathy and age-related macular degeneration, and colorectal carcinoma and metastatic melanoma. Splicing factors and their regulatory molecules alter splice site selection, such that cells can switch from the anti-angiogenic VEGF(xxx)b isoforms to the pro-angiogenic VEGF(xxx) isoforms, including SRp55 (serine/arginine protein 55), ASF/SF2 (alternative splicing factor/splicing factor 2) and SRPK (serine arginine domain protein kinase), and inhibitors of these molecules can inhibit angiogenesis in the eye, and splice site selection in cancer cells, opening up the possibility of using splicing factor inhibitors as novel anti-angiogenic therapeutics. Endogenous anti-angiogenic VEGF(xxx)b isoforms are cytoprotective for endothelial, epithelial and neuronal cells in vitro and in vivo, suggesting both an improved safety profile and an explanation for unpredicted anti-VEGF side effects. In summary, C-terminal distal splicing is a key component of VEGF biology, overlooked by the vast majority of publications in the field, and these findings require a radical revision of our understanding of VEGF biology in normal human physiology.
Collapse
Affiliation(s)
- Yan Qiu
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, U.K
| | - Coralie Hoareau-Aveilla
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, U.K
| | - Sebastian Oltean
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, U.K
| | - Steven J. Harper
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, U.K
| | - David O. Bates
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, U.K
| |
Collapse
|
259
|
Wang R, Crystal RG, Hackett NR. Identification of an exonic splicing silencer in exon 6A of the human VEGF gene. BMC Mol Biol 2009; 10:103. [PMID: 19922608 PMCID: PMC2784459 DOI: 10.1186/1471-2199-10-103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Accepted: 11/17/2009] [Indexed: 11/15/2022] Open
Abstract
Background The different isoforms of vascular endothelial growth factor (VEGF) play diverse roles in vascular growth, structure and function. Alternative splicing of the VEGF gene results in the expression of three abundant isoforms: VEGF121, VEGF165 and VEGF189. The mRNA for VEGF189 contains the alternatively spliced exon 6A whereas the mRNA for VEGF165 lacks this exon. The objective of this study was to identify the cis elements that control utilization of exon 6A. A reporter minigene was constructed (pGFP-E6A) containing the coding sequence for GFP whose translation was dependent on faithful splicing for removal of the VEGF exon 6A. To identify cis-acting splicing elements, sequential deletions were made across exon 6A in the pGFP-E6A plasmid. Results A candidate cis-acting exonic splicing silencer (ESS) comprising nucleotides 22-30 of exon 6A sequence was identified corresponding to the a silencer consensus sequence of AAGGGG. The function of this sequence as an ESS was confirmed in vivo both in the context of the reporter minigene as a plasmid and in the context of a longer minigene with VEGF exon 6A in its native context in an adenoviral gene transfer vector. Further mutagenesis studies resulted in the identification of the second G residue of the putative ESS as the most critical for function. Conclusion This work establishes the identity of cis sequences that regulate alternative VEGF splicing and dictate the relative expression levels of VEGF isoforms.
Collapse
Affiliation(s)
- Rui Wang
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA.
| | | | | |
Collapse
|
260
|
Nowak DG, Amin EM, Rennel ES, Hoareau-Aveilla C, Gammons M, Damodoran G, Hagiwara M, Harper SJ, Woolard J, Ladomery MR, Bates DO. Regulation of vascular endothelial growth factor (VEGF) splicing from pro-angiogenic to anti-angiogenic isoforms: a novel therapeutic strategy for angiogenesis. J Biol Chem 2009; 285:5532-40. [PMID: 19906640 PMCID: PMC2820781 DOI: 10.1074/jbc.m109.074930] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is produced either as a pro-angiogenic or anti-angiogenic protein depending upon splice site choice in the terminal, eighth exon. Proximal splice site selection (PSS) in exon 8 generates pro-angiogenic isoforms such as VEGF165, and distal splice site selection (DSS) results in anti-angiogenic isoforms such as VEGF165b. Cellular decisions on splice site selection depend upon the activity of RNA-binding splice factors, such as ASF/SF2, which have previously been shown to regulate VEGF splice site choice. To determine the mechanism by which the pro-angiogenic splice site choice is mediated, we investigated the effect of inhibition of ASF/SF2 phosphorylation by SR protein kinases (SRPK1/2) on splice site choice in epithelial cells and in in vivo angiogenesis models. Epithelial cells treated with insulin-like growth factor-1 (IGF-1) increased PSS and produced more VEGF165 and less VEGF165b. This down-regulation of DSS and increased PSS was blocked by protein kinase C inhibition and SRPK1/2 inhibition. IGF-1 treatment resulted in nuclear localization of ASF/SF2, which was blocked by SPRK1/2 inhibition. Pull-down assay and RNA immunoprecipitation using VEGF mRNA sequences identified an 11-nucleotide sequence required for ASF/SF2 binding. Injection of an SRPK1/2 inhibitor reduced angiogenesis in a mouse model of retinal neovascularization, suggesting that regulation of alternative splicing could be a potential therapeutic strategy in angiogenic pathologies.
Collapse
Affiliation(s)
- Dawid G Nowak
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol BS2 8EJ, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
261
|
Artac RA, McFee RM, Longfellow Smith RA, Baltes-Breitwisch MM, Clopton DT, Cupp AS. Neutralization of vascular endothelial growth factor antiangiogenic isoforms is more effective than treatment with proangiogenic isoforms in stimulating vascular development and follicle progression in the perinatal rat ovary. Biol Reprod 2009; 81:978-88. [PMID: 19605786 PMCID: PMC2770023 DOI: 10.1095/biolreprod.109.078097] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 05/05/2009] [Accepted: 06/16/2009] [Indexed: 11/01/2022] Open
Abstract
Inhibition of vascular endothelial growth factor A (VEGFA) signal transduction arrests vascular and follicle development. Because antiangiogenic VEGFA isoforms are proposed to block proangiogenic VEGFA isoforms from binding to their receptors, we hypothesized that proangiogenic isoforms promote and antiangiogenic isoforms inhibit these processes. The antiangiogenic isoforms Vegfa_165b and Vegfa_189b were amplified and sequenced from rat ovaries. The Vegfa_165b sequence was 90% homologous to human VEGFA_165B. Quantitative RT-PCR determined that Vegfa_165b mRNA was more abundant around Embryonic Day 18, but Vegfa_189b lacked a distinct pattern of abundance. Antiangiogenic VEGFA isoforms were localized to pregranulosa and granulosa cells of all follicle stages and to theca cells of advanced-stage follicles. To determine the effects of VEGFA isoforms in developing ovaries, Postnatal Day 3/4 rat ovaries were cultured with VEGFA_164 or an antibody to antiangiogenic isoforms (anti-VEGFAxxxB). Treatment with 50 ng/ml of VEGFA_164 resulted in a 93% increase in vascular density (P < 0.01), and treated ovaries were composed of fewer primordial follicles (stage 0) and more developing follicles (stages 1-4) than controls (P < 0.04). Ovaries treated with 5 ng/ml of VEGFAxxxB antibody had a 93% increase in vascular density (P < 0.02), with fewer primordial and early primary follicles (stage 1) and more primary, transitional, and secondary follicles (stages 2, 3, and 4, respectively) compared with controls (P < 0.005). We conclude that neutralization of antiangiogenic VEGFA isoforms may be a more effective mechanism of enhancing vascular and follicular development in perinatal rat ovaries than treatment with the proangiogenic isoform VEGFA_164.
Collapse
Affiliation(s)
- Robin A. Artac
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Renee M. McFee
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska
| | | | | | - Debra T. Clopton
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Andrea S. Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
262
|
Abstract
The vascular endothelial growth factor (VEGF) family of proteins regulates blood flow, growth, and function in both normal physiology and disease processes. VEGF-A is alternatively spliced to form multiple isoforms, in two subfamilies, that have specific, novel functions. Alternative splicing of exons 5-7 of the VEGF gene generates forms with differing bioavailability and activities, whereas alternative splice-site selection in exon 8 generates proangiogenic, termed VEGF(xxx), or antiangiogenic proteins, termed VEGF(xxx)b. Despite its name, emerging roles for VEGF isoforms on cell types other than endothelium have now been identified. Although VEGF-A has conventionally been considered to be a family of proangiogenic, propermeability vasodilators, the identification of effects on nonendothelial cells, and the discovery of the antiangiogenic subfamily of splice isoforms, has added further complexity to their regulation of microvascular function. The distally spliced antiangiogenic isoforms are expressed in normal human tissue, but downregulated in angiogenic diseases, such as cancer and proliferative retinopathy, and in developmental pathologies, such as Denys Drash syndrome and preeclampsia. Here, we examine the molecular diversity of VEGF-A as a regulator of its biological activity and compare the role of the pro- and antiangiogenic VEGF-A splice isoforms in both normal and pathophysiological processes.
Collapse
Affiliation(s)
- Jeanette Woolard
- Department of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Sciences, University of Bristol, Bristol, UK.
| | | | | | | |
Collapse
|
263
|
Structure-function analysis of VEGF receptor activation and the role of coreceptors in angiogenic signaling. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2009; 1804:567-80. [PMID: 19761875 DOI: 10.1016/j.bbapap.2009.09.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 08/22/2009] [Accepted: 09/04/2009] [Indexed: 12/11/2022]
Abstract
Vascular endothelial growth factors (VEGFs) constitute a family of six polypeptides, VEGF-A, -B, -C, -D, -E and PlGF, that regulate blood and lymphatic vessel development. VEGFs specifically bind to three type V receptor tyrosine kinases (RTKs), VEGFR-1, -2 and -3, and to coreceptors such as neuropilins and heparan sulfate proteoglycans (HSPG). VEGFRs are activated upon ligand-induced dimerization mediated by the extracellular domain (ECD). A study using receptor constructs carrying artificial dimerization-promoting transmembrane domains (TMDs) showed that receptor dimerization is necessary, but not sufficient, for receptor activation and demonstrates that distinct orientation of receptor monomers is required to instigate transmembrane signaling. Angiogenic signaling by VEGF receptors also depends on cooperation with specific coreceptors such as neuropilins and HSPG. A number of VEGF isoforms differ in binding to coreceptors, and ligand-specific signal output is apparently the result of the specific coreceptor complex assembled by a particular VEGF isoform. Here we discuss the structural features of VEGF family ligands and their receptors in relation to their distinct signal output and angiogenic potential.
Collapse
|
264
|
VEGF(121)b, a new member of the VEGF(xxx)b family of VEGF-A splice isoforms, inhibits neovascularisation and tumour growth in vivo. Br J Cancer 2009; 101:1183-93. [PMID: 19707198 PMCID: PMC2768092 DOI: 10.1038/sj.bjc.6605249] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: The key mediator of new vessel formation in cancer and other diseases is VEGF-A. VEGF-A exists as alternatively spliced isoforms - the pro-angiogenic VEGFxxx family generated by exon 8 proximal splicing, and a sister family, termed VEGFxxxb, exemplified by VEGF165b, generated by distal splicing of exon 8. However, it is unknown whether this anti-angiogenic property of VEGF165b is a general property of the VEGFxxxb family of isoforms. Methods: The mRNA and protein expression of VEGF121b was studied in human tissue. The effect of VEGF121b was analysed by saturation binding to VEGF receptors, endothelial migration, apoptosis, xenograft tumour growth, pre-retinal neovascularisation and imaging of biodistribution in tumour-bearing mice with radioactive VEGF121b. Results: The existence of VEGF121b was confirmed in normal human tissues. VEGF121b binds both VEGF receptors with similar affinity as other VEGF isoforms, but inhibits endothelial cell migration and is cytoprotective to endothelial cells through VEGFR-2 activation. Administration of VEGF121b normalised retinal vasculature by reducing both angiogenesis and ischaemia. VEGF121b reduced the growth of xenografted human colon tumours in association with reduced microvascular density, and an intravenous bolus of VEGF121b is taken up into colon tumour xenografts. Conclusion: Here we identify a second member of the family, VEGF121b, with similar properties to those of VEGF165b, and underline the importance of the six amino acids of exon 8b in the anti-angiogenic activity of the VEGFxxxb isoforms.
Collapse
|
265
|
Dasgupta S, Wasson LM, Rauniyar N, Prokai L, Borejdo J, Vishwanatha JK. Novel gene C17orf37 in 17q12 amplicon promotes migration and invasion of prostate cancer cells. Oncogene 2009; 28:2860-72. [PMID: 19503095 PMCID: PMC2726905 DOI: 10.1038/onc.2009.145] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 04/17/2009] [Accepted: 05/06/2009] [Indexed: 01/30/2023]
Abstract
C17orf37/MGC14832, a novel gene located on human chromosome 17q12 in the ERBB2 amplicon, is abundantly expressed in breast cancer. C17orf37 expression has been reported to positively correlate with grade and stage of cancer progression; however the functional significance of C17orf37 overexpression in cancer biology is not known. Here, we show that C17orf37 is highly expressed in prostate cancer cell lines and tumors, compared to minimal expression in normal prostate cells and tissues. Cellular localization studies by confocal and total internal reflection fluorescence microscopy revealed predominant expression of C17orf37 in the cytosol with intense staining in the membrane of prostate cancer cells. RNA-interference-mediated downregulation of C17orf37 resulted in decreased migration and invasion of DU-145 prostate cancer cells, and suppressed the DNA-binding activity of nuclear factor-kappaB (NF-kappaB) transcription factor resulting in reduced expression of downstream target genes matrix metalloproteinase 9, urokinase plasminogen activator and vascular endothelial growth factor. Phosphorylation of PKB/Akt was also reduced upon C17orf37 downregulation, suggesting C17orf37 acts as a signaling molecule that increases invasive potential of prostate cancer cells by NF-kappaB-mediated downstream target genes. Our data strongly suggest C17orf37 overexpression in prostate cancer functionally enhances migration and invasion of tumor cells, and is an important target for cancer therapy.
Collapse
Affiliation(s)
- S Dasgupta
- Department of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas
| | - L M Wasson
- Staff Pathologist, Harris Methodist Fort Worth Hospital, Fort Worth, TX, USA
| | - N Rauniyar
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - L Prokai
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - J Borejdo
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - J K Vishwanatha
- Department of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
- Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
266
|
Rajan P, Elliott DJ, Robson CN, Leung HY. Alternative splicing and biological heterogeneity in prostate cancer. Nat Rev Urol 2009; 6:454-60. [PMID: 19657379 DOI: 10.1038/nrurol.2009.125] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The biological diversity of prostate cancer confounds standardization of therapy. Advances in molecular profiling suggest that differences in the genetic composition of tumors significantly contribute to the complexity of the disease. Alternative pre-mRNA splicing is a key genetic process underlying biological diversity. During alternative splicing, coding and noncoding regions of a single gene are rearranged to generate several messenger RNA transcripts yielding distinct protein isoforms with differing biological functions. Misregulation of the splicing machinery and mutations in key regulatory elements affect splicing of cancer-relevant genes. In prostate cancer, aberrant and alternative splicing generates proteins that influence cell phenotypes and survival of patients. Splicing events may be exploited for clinical benefit, and technological advances are beginning to uncover novel biomarkers and therapeutic targets. Since splicing mediates information transfer from the genome to the proteome, it adds an important dimension to '-omics'-based molecular signatures used to individualize care of patients.
Collapse
|
267
|
Sakly N, Mirshahi P, Ducros E, Soria J, Ghedira I, Mirshahi M. Angiogenic activity in sera of patients with systemic lupus erythematosus. Lupus 2009; 18:705-712. [PMID: 19502266 DOI: 10.1177/0961203309103087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Angiogenesis plays a critical role in the pathogenesis of several connective tissue diseases. There is, however, relatively little information available on the role of angiogenesis in systemic lupus erythematosus (SLE). The aim of this study was to investigate the angiogenic activity in sera of patients with SLE and to determine the association between angiogenic activity and clinical complications. Sera from 66 Tunisian females with SLE and from 32 healthy blood donors were studied for their angiogenic activity using the in-vitro tube formation test on Matrigel. Samples were divided into five groups according to their angiogenic activity, which was scored from 0 (no angiogenesis) to 4 (high angiogenic activity). Samples from each group were then tested randomly to assess serum concentration of vascular endothelial growth factor (VEGF). No correlation was found between angiogenic activity scores and serum VEGF levels. Considering angiogenesis assessment in-vitro, sera of patients with SLE showed a much higher angiogenic activity than healthy controls since a high angiogenic score (score 4) is present in 43.9% of patients and in 6.3% of controls (P < 0.0002). This high angiogenic activity is not correlated with disease activity; however, SLE patients with anti-dsDNA antibodies and those with nephritis showed higher angiogenic activity compared with patients without these complications since score 4 is found in 50.9% and 67.9% versus 9.1% (P = 0.017) and 26.3% (P < 0.001), respectively. In conclusion, our study showed that high serum angiogenic activity in SLE was not correlated with the VEGF levels. We suggest the use of the 'in-vitro' tube formation test as a better tool to study the angiogenic potential of sera. We found that in patients with SLE, serum angiogenic activity is increased compared with healthy controls. This high angiogenic activity is associated with renal complications and with the presence of anti-dsDNA antibodies. These findings suggest an involvement of angiogenesis disturbance in the pathogenesis of SLE.
Collapse
Affiliation(s)
- N Sakly
- Faculty of Pharmacy, Research Unit 03/UR/07-2, Monastir 5000, Tunisia; Faculty of Medicine, UMRS 872, CRC, Paris VI 15 rue de l'Ecole de Médecine, 75006 Paris, France
| | | | | | | | | | | |
Collapse
|
268
|
Grépin R, Pagès G. Le Vascular Endothelial Growth Factor (VEGF) : un modèle de régulation d'expression génique et un marqueur d'agressivité tumorale. Une cible thérapeutique évidente ? ACTA ACUST UNITED AC 2009; 203:181-92. [DOI: 10.1051/jbio/2009022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
269
|
Abstract
Anti-angiogenic therapies currently revolve around targeting vascular endothelial growth factor-A (VEGF-A) or its receptors. These therapies are effective to some degree, but have low response rates and poor side-effect profiles. Part of these problems is likely to be due to their lack of specificity between pro- and anti-angiogenic isoforms, and their nonspecific effects on proactive, pleiotropic survival and maintenance roles of VEGF-A in endothelial and other cell types. An alternative approach, and one which has recently been shown to be effective in animal models of neovascularization in the eye, is to target the mechanisms by which the cell generates pro-angiogenic splice forms of VEGF-A, its receptors and, co-incidentally, by targeting the upstream processes, other oncogenes that have antagonistic splice isoforms. The concept here is to target the splicing mechanisms that control splice site choice in the VEGF-A mRNA. Recent evidence on the pharmacological possibilities of such splice factors is described.
Collapse
Affiliation(s)
- Emma S Rennel
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK Tel:.+44 117 982 8367 Fax: +44 117 982 8151
| | - Steven J Harper
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - David O Bates
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK Tel:. +44 117 982 8367 Fax: +44 117 982 8151
| |
Collapse
|
270
|
Ruiz de Almodovar C, Lambrechts D, Mazzone M, Carmeliet P. Role and therapeutic potential of VEGF in the nervous system. Physiol Rev 2009; 89:607-48. [PMID: 19342615 DOI: 10.1152/physrev.00031.2008] [Citation(s) in RCA: 348] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The development of the nervous and vascular systems constitutes primary events in the evolution of the animal kingdom; the former provides electrical stimuli and coordination, while the latter supplies oxygen and nutrients. Both systems have more in common than originally anticipated. Perhaps the most striking observation is that angiogenic factors, when deregulated, contribute to various neurological disorders, such as neurodegeneration, and might be useful for the treatment of some of these pathologies. The prototypic example of this cross-talk between nerves and vessels is the vascular endothelial growth factor or VEGF. Although originally described as a key angiogenic factor, it is now well established that VEGF also plays a crucial role in the nervous system. We describe the molecular properties of VEGF and its receptors and review the current knowledge of its different functions and therapeutic potential in the nervous system during development, health, disease and in medicine.
Collapse
|
271
|
Varey AH, Dunn DB. Comment on: Akhavani MA, Sivakumar B, Paleolog EM and Kang, N: ‘Angiogenesis and plastic surgery’ J Plast Reconstr Aesthet Surg 2008;61:1425–37. J Plast Reconstr Aesthet Surg 2009; 62:710. [DOI: 10.1016/j.bjps.2008.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 12/12/2008] [Indexed: 11/15/2022]
|
272
|
Bills V, Varet J, Millar A, Harper S, Soothill P, Bates D. Failure to up-regulate VEGF165b in maternal plasma is a first trimester predictive marker for pre-eclampsia. Clin Sci (Lond) 2009; 116:265-72. [PMID: 18826376 PMCID: PMC2635543 DOI: 10.1042/cs20080270] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 09/17/2008] [Accepted: 10/01/2008] [Indexed: 12/03/2022]
Abstract
Pre-eclampsia is a pregnancy-related condition characterized by hypertension, proteinuria and endothelial dysfunction. VEGF(165)b, formed by alternative splicing of VEGF (vascular endothelial growth factor) pre-mRNA, inhibits VEGF(165)-mediated vasodilation and angiogenesis, but has not been quantified in pregnancy. ELISAs were used to measure means+/-S.E.M. plasma VEGF(165)b, sEng (soluble endoglin) and sFlt-1 (soluble fms-like tyrosine kinase-1). At 12 weeks of gestation, the plasma VEGF(165)b concentration was significantly up-regulated in plasma from women who maintained normal blood pressure throughout their pregnancy (normotensive group, 4.90+/-1.6 ng/ml; P<0.01, as determined using a Mann-Whitney U test) compared with non-pregnant women (0.40+/-0.22 ng/ml). In contrast, in patients who later developed pre-eclampsia, VEGF(165)b levels were lower than in the normotensive group (0.467+/-0.209 ng/ml), but were no greater than non-pregnant women. At term, plasma VEGF(165)b concentrations were greater than normal in both pre-eclamptic (3.75+/-2.24 ng/ml) and normotensive (10.58 ng/ml+/-3.74 ng/ml; P>0.1 compared with pre-eclampsia) pregnancies. Patients with a lower than median plasma VEGF(165)b at 12 weeks had elevated sFlt-1 and sEng pre-delivery. Concentrations of sFlt-1 (1.20+/-0.07 and 1.27+/-0.18 ng/ml) and sEng (4.4+/-0.18 and 4.1+/-0.5 ng/ml) were similar at 12 weeks of gestation in the normotensive and pre-eclamptic groups respectively. Plasma VEGF(165)b levels were elevated in pregnancy, but this increase is delayed in women that subsequently develop pre-eclampsia. In conclusion, low VEGF(165)b may therefore be a clinically useful first trimester plasma marker for increased risk of pre-eclampsia.
Collapse
Key Words
- angiogenesis
- plasma marker
- pre-eclampsia
- splice variant
- vascular endothelial growth factor165b (vegf165b)
- vascular permeability
- auc, area under the curve
- bp, blood pressure
- cv, coefficient of variation
- eia, enzyme immunoassay
- ceia, competitive eia
- flt-1, fms-like tyrosine kinase-1
- roc, receiver operating characteristic
- seng, soluble endoglin
- sflt-1, soluble flt-1
- vegf, vascular endothelial growth factor
- vegfr, vegf receptor
Collapse
Affiliation(s)
- Victoria L. Bills
- *Microvascular Research Laboratories, Department of
Physiology, Preclinical Veterinary School, University of Bristol, Bristol BS2
8EJ, U.K
- †Department of Obstetrics and Gynaecology, St Michael's
Hospital, University of Bristol, Bristol BS2 8EG, U.K
| | - Julia Varet
- ‡Clinical Sciences at North Bristol, Medical School Unit,
Southmead Hospital, Bristol BS10 5NB, U.K
| | - Ann Millar
- ‡Clinical Sciences at North Bristol, Medical School Unit,
Southmead Hospital, Bristol BS10 5NB, U.K
| | - Steven J. Harper
- *Microvascular Research Laboratories, Department of
Physiology, Preclinical Veterinary School, University of Bristol, Bristol BS2
8EJ, U.K
| | - Peter W. Soothill
- †Department of Obstetrics and Gynaecology, St Michael's
Hospital, University of Bristol, Bristol BS2 8EG, U.K
| | - David O. Bates
- *Microvascular Research Laboratories, Department of
Physiology, Preclinical Veterinary School, University of Bristol, Bristol BS2
8EJ, U.K
| |
Collapse
|
273
|
Qutub AA, Popel AS. Elongation, proliferation & migration differentiate endothelial cell phenotypes and determine capillary sprouting. BMC SYSTEMS BIOLOGY 2009; 3:13. [PMID: 19171061 PMCID: PMC2672076 DOI: 10.1186/1752-0509-3-13] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 01/26/2009] [Indexed: 12/22/2022]
Abstract
BACKGROUND Angiogenesis, the growth of capillaries from preexisting blood vessels, has been extensively studied experimentally over the past thirty years. Molecular insights from these studies have lead to therapies for cancer, macular degeneration and ischemia. In parallel, mathematical models of angiogenesis have helped characterize a broader view of capillary network formation and have suggested new directions for experimental pursuit. We developed a computational model that bridges the gap between these two perspectives, and addresses a remaining question in angiogenic sprouting: how do the processes of endothelial cell elongation, migration and proliferation contribute to vessel formation? RESULTS We present a multiscale systems model that closely simulates the mechanisms underlying sprouting at the onset of angiogenesis. Designed by agent-based programming, the model uses logical rules to guide the behavior of individual endothelial cells and segments of cells. The activation, proliferation, and movement of these cells lead to capillary growth in three dimensions. By this means, a novel capillary network emerges out of combinatorially complex interactions of single cells. Rules and parameter ranges are based on literature data on endothelial cell behavior in vitro. The model is designed generally, and will subsequently be applied to represent species-specific, tissue-specific in vitro and in vivo conditions. Initial results predict tip cell activation, stalk cell development and sprout formation as a function of local vascular endothelial growth factor concentrations and the Delta-like 4 Notch ligand, as it might occur in a three-dimensional in vitro setting. Results demonstrate the differential effects of ligand concentrations, cell movement and proliferation on sprouting and directional persistence. CONCLUSION This systems biology model offers a paradigm closely related to biological phenomena and highlights previously unexplored interactions of cell elongation, migration and proliferation as a function of ligand concentration, giving insight into key cellular mechanisms driving angiogenesis.
Collapse
Affiliation(s)
- Amina A Qutub
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
274
|
Potiron VA, Roche J, Drabkin HA. Semaphorins and their receptors in lung cancer. Cancer Lett 2009; 273:1-14. [PMID: 18625544 PMCID: PMC2630718 DOI: 10.1016/j.canlet.2008.05.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 02/28/2008] [Accepted: 05/23/2008] [Indexed: 12/30/2022]
Abstract
Semaphorins are a large family of secreted, transmembrane and GPI-linked proteins initially characterized in the development of the nervous system and axonal guidance. Semaphorins are expressed in many tissues where they regulate normal development, organ morphogenesis, immunity and angiogenesis. They affect the cytoskeleton, actin filament organization, microtubules and cell adhesion. Semaphorin signaling is transduced by plexins, which in the case of most class-3 semaphorins requires high-affinity neuropilin receptors. The neuropilins also function as receptors for VEGF and other growth factors, and their expression is often abnormal in tumors. In cancer, semaphorins have both tumor suppressor and tumor promoting functions. We review here the current status of semaphorins and their receptors in tumor development with a focus on lung cancer.
Collapse
Affiliation(s)
- Vincent A Potiron
- Division of Hematology/Oncology, Medical University of South Carolina, P.O. Box 250623, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | | | | |
Collapse
|
275
|
Abstract
Continuous monitoring of neovascular growth in vivo is required for the development and evaluation of drugs acting as suppressors or stimulators of angiogenesis. The cornea assay consists of the placement of an angiogenesis stimulus (tumor tissue, cell suspension, growth factor) into a micropocket produced in the cornea thickness to evoke vascular outgrowth from the peripherally located limbal vasculature. Neovascular development and progression can be modified by the presence of locally released or applied inhibitory factors or by systemically given antiangiogenic drugs. This assay has the advantage over other in vivo assays of measuring new blood vessels only since the cornea is initially avascular. The experimental details of the avascular cornea assay and its advantages and disadvantages in different species are discussed.
Collapse
Affiliation(s)
- Marina Ziche
- Department of Molecular Biology, University of Siena, Siena, Italy
| | | |
Collapse
|
276
|
Abstract
Successful therapeutic angiogenesis requires an understanding of how the milieu of growth factors available combine to form a mature vascular bed. This requires a model in which multiple physiological and cell biological parameters can be identified. The adenoviral-mediated mesenteric angiogenesis assay as described here is ideal for that purpose. Adenoviruses expressing growth factors (vascular endothelial growth factor [VEGF] and angiopoietin 1 [Ang-1]) were injected into the mesenteric fat pad of adult male Wistar rats. The clear, thin, and relatively avascular mesenteric panel was used to measure increased vessel perfusion by intravital microscopy. In addition, high-powered microvessel analysis was carried out by immunostaining of features essential for the study of angiogenesis (endothelium, pericyte, smooth muscle cell area, and proliferation), allowing functional data to be obtained in conjunction with high-power microvessel ultrastructural analysis. A combination of individual growth factors resulted in a distinct vascular phenotype from either factor alone, with all treatments increasing the functional vessel area. VEGF produced shorter, narrow, highly branched, and sprouting vessels with normal pericyte coverage. Ang-1 induced broader, longer neovessels with no apparent increase in branching or sprouting. However, Ang-1-induced blood vessels displayed a significantly higher pericyte ensheathment. Combined treatment resulted in higher perfusion, larger and less-branched vessels, with normal pericyte coverage, suggesting them to be more mature. This model can be used to show that Ang-1 and VEGF use different physiological mechanisms to enhance vascularisation of relatively avascular tissue.
Collapse
Affiliation(s)
- Andrew V Benest
- Division of Vascular Oncology and Metastasis, DKFZ, Heidelberg, Germany
| | | |
Collapse
|
277
|
Bevan HS, van den Akker NM, Qiu Y, Polman JA, Foster RR, Yem J, Nishikawa A, Satchell SC, Harper SJ, Gittenberger-de Groot AC, Bates DO. The alternatively spliced anti-angiogenic family of VEGF isoforms VEGFxxxb in human kidney development. NEPHRON. PHYSIOLOGY 2008; 110:p57-67. [PMID: 19039247 PMCID: PMC2635558 DOI: 10.1159/000177614] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 09/01/2008] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIM Vascular endothelial growth factor (VEGF), required for renal development, is generated by alternative splicing of 8 exons to produce two families, pro-angiogenic VEGF(xxx), formed by proximal splicing in exon 8 (exon 8a), and anti-angiogenic VEGF(xxx)b, generated by distal splicing in exon 8 (exon 8b). VEGF(165)b, the first described exon 8b-containing isoform, antagonises VEGF(165) and is anti-angiogenic in vivo. METHODS Using VEGF(xxx)b-specific antibodies, we investigated its expression quantitatively and qualitatively in developing kidney, and measured the effect of VEGF(165)b on renal endothelial and epithelial cells. RESULTS VEGF(xxx)b formed 45% of total VEGF protein in adult renal cortex, and VEGF(165)b does not increase glomerular endothelial cell permeability, it inhibits migration, and is cytoprotective for podocytes. During renal development, VEGF(xxx)b was expressed in the condensed vesicles of the metanephros, epithelial cells of the comma-shaped bodies, invading endothelial cells and epithelial cells of the S-shaped body, and in the immature podocytes. Expression reduced as the glomerulus matured. CONCLUSION These results show that the anti-angiogenic VEGF(xxx)b isoforms are highly expressed in adult and developing renal cortex, and suggest that the VEGF(xxx)b family plays a role in glomerular maturation and podocyte protection by regulating the pro-angiogenic pro-permeability properties of VEGF(xxx) isoforms.
Collapse
Affiliation(s)
- Heather S. Bevan
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| | | | - Yan Qiu
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| | - Japke A.E. Polman
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden,The Netherlands
| | - Rebecca R. Foster
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| | - Justin Yem
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| | - Ali Nishikawa
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| | | | - Steven J. Harper
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| | | | - David O. Bates
- Microvascular Research Laboratories, Department of Physiology, University of Bristol, Bristol, UK
| |
Collapse
|
278
|
Freitas-Andrade M, Carmeliet P, Stanimirovic DB, Moreno M. VEGFR-2-mediated increased proliferation and survival in response to oxygen and glucose deprivation in PlGF knockout astrocytes. J Neurochem 2008; 107:756-67. [DOI: 10.1111/j.1471-4159.2008.05660.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
279
|
Abstract
The physiology of microvessels limits the growth and development of tumours. Tumours gain nutrients and excrete waste through growth-associated microvessels. New anticancer therapies target this microvasculature by inhibiting vascular endothelial growth factor A (VEGF-A) splice isoforms that promote microvessel growth. However, certain VEGF-A splice isoforms in normal tissues inhibit growth of microvessels. Thus, it is the VEGF-A isoform balance, which is controlled by mRNA splicing, that orchestrates angiogenesis. Here, we highlight the functional differences between the pro-angiogenic and the anti-angiogenic VEGF-A isoform families and the potential to harness the synthetic capacity of cancer cells to produce factors that inhibit, rather than aid, cancer growth.
Collapse
Affiliation(s)
- Steven J Harper
- Microvascular Research Laboratories, Department of Physiology & Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK.
| | | |
Collapse
|
280
|
Nowak DG, Woolard J, Amin EM, Konopatskaya O, Saleem MA, Churchill AJ, Ladomery MR, Harper SJ, Bates DO. Expression of pro- and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors. J Cell Sci 2008; 121:3487-95. [PMID: 18843117 PMCID: PMC2613349 DOI: 10.1242/jcs.016410] [Citation(s) in RCA: 254] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor A (VEGFA; hereafter referred to as VEGF) is a key regulator of physiological and pathological angiogenesis. Two families of VEGF isoforms are generated by alternate splice-site selection in the terminal exon. Proximal splice-site selection (PSS) in exon 8 results in pro-angiogenic VEGFxxx isoforms (xxx is the number of amino acids), whereas distal splice-site selection (DSS) results in anti-angiogenic VEGFxxxb isoforms. To investigate control of PSS and DSS, we investigated the regulation of isoform expression by extracellular growth factor administration and intracellular splicing factors. In primary epithelial cells VEGFxxxb formed the majority of VEGF isoforms (74%). IGF1, and TNFalpha treatment favoured PSS (increasing VEGFxxx) whereas TGFbeta1 favoured DSS, increasing VEGFxxxb levels. TGFbeta1 induced DSS selection was prevented by inhibition of p38 MAPK and the Clk/sty (CDC-like kinase, CLK1) splicing factor kinase family, but not ERK1/2. Clk phosphorylates SR protein splicing factors ASF/SF2, SRp40 and SRp55. To determine whether SR splicing factors alter VEGF splicing, they were overexpressed in epithelial cells, and VEGF isoform production assessed. ASF/SF2, and SRp40 both favoured PSS, whereas SRp55 upregulated VEGFxxxb (DSS) isoforms relative to VEGFxxx. SRp55 knockdown reduced expression of VEGF165b. Moreover, SRp55 bound to a 35 nucleotide region of the 3'UTR immediately downstream of the stop codon in exon 8b. These results identify regulation of splicing by growth and splice factors as a key event in determining the relative pro-versus anti-angiogenic expression of VEGF isoforms, and suggest that p38 MAPK-Clk/sty kinases are responsible for the TGFbeta1-induced DSS selection, and identify SRp55 as a key regulatory splice factor.
Collapse
Affiliation(s)
- Dawid G. Nowak
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - Jeanette Woolard
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - Elianna Mohamed Amin
- Centre for Research in Biomedicine, Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol BS16 1QY, UK
| | - Olga Konopatskaya
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - Moin A. Saleem
- Department of Clinical Sciences at North Bristol (Academic Renal Unit), Paul O’Gorman Lifeline Centre, Southmead Hospital, Bristol BS10 5NB, UK
| | - Amanda J. Churchill
- Department of Ophthalmology, Bristol Eye Hospital, University of Bristol, Lower Maudlin Street, Bristol BS1 2LX, UK
| | - Michael R. Ladomery
- Centre for Research in Biomedicine, Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol BS16 1QY, UK
| | - Steven J. Harper
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - David O. Bates
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| |
Collapse
|
281
|
Akhavani MA, Sivakumar B, Paleolog EM, Kang N. Angiogenesis and plastic surgery. J Plast Reconstr Aesthet Surg 2008; 61:1425-37. [PMID: 18835232 DOI: 10.1016/j.bjps.2008.05.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 03/27/2008] [Accepted: 05/29/2008] [Indexed: 01/23/2023]
Abstract
SUMMARY Angiogenesis, the formation of new blood vessels from an existing vascular bed, is a normal physiological process which also underpins many--apparently unrelated--pathological states. It is an integral factor in determining the success or failure of many procedures in plastic and reconstructive surgery. As a result, the ability to control the process would be of great therapeutic benefit. To appreciate the potential benefits and limitations of recent advances in our understanding of angiogenesis, it is important to comprehend the basic physiology of blood vessel formation. This review aims to summarise current knowledge of the way in which angiogenesis is controlled and to look at how disordered vessel development results in pathology relevant to plastic surgery. Through this we hope to provide a comprehensive overview of the recent advances in angiogenesis as they relate to plastic surgery, particularly the promotion of flap survival, tendon healing, nerve regeneration, fracture healing and ulcer treatments.
Collapse
|
282
|
Zbytek B, Carlson JA, Granese J, Ross J, Mihm MC, Slominski A. Current concepts of metastasis in melanoma. EXPERT REVIEW OF DERMATOLOGY 2008; 3:569-585. [PMID: 19649148 PMCID: PMC2601641 DOI: 10.1586/17469872.3.5.569] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The main cause of death in melanoma patients is widespread metastases. Staging of melanoma is based on the primary tumor thickness, ulceration, lymph node and distant metastases. Metastases develop in regional lymph nodes, as satellite or in-transit lesions, or in distant organs. Lymph flow and chemotaxis is responsible for the homing of melanoma cells to different sites. Standard pathologic evaluation of sentinel lymph nodes fails to find occult melanoma in a significant proportion of cases. Detection of small numbers of malignant melanoma cells in these and other sites, such as adjacent to the primary site, bone marrow or the systemic circulation, may be enhanced by immunohistochemistry, reverse transcription PCR, evaluation of lymphatic vessel invasion and proteomics. In the organs to which melanoma cells metastasize, extravasation of melanoma cells is regulated by adhesion molecules, matrix metalloproteases, chemokines and growth factors. Melanoma cells may travel along external vessel lattices. After settling in the metastatic sites, melanoma cells develop mechanisms that protect them against the attack of the immune system. It is thought that one of the reasons why melanoma cells are especially resistant to killing is the fact that melanocytes (cells from which melanoma cells derive) are resistant to such noxious factors as ultraviolet light and reactive oxygen species. Targeted melanoma therapies are, so far, largely unsuccessful, and new ones, such as adjuvant inhibition of melanogenesis, are under development.
Collapse
Affiliation(s)
- Blazej Zbytek
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163, USA, Tel.: +1 901 448 6300, Fax: +1 901 448 6979,
| | - J Andrew Carlson
- Department of Pathology and Laboratory Medicine, Albany Medical Center, 47 New Scotland Avenue, Albany, NY, USA, Tel.: +1 518 262 8099, Fax: +1 518 262 8092,
| | - Jacqueline Granese
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163, USA, Tel.: +1 901 448 6300, Fax: +1 901 448 6979,
| | - Jeffrey Ross
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY, 47 New Scotland Avenue, Albany, NY, USA, Tel.: +1 518 262 5461, Fax: +1 518 262 8092,
| | - Martin C Mihm
- Dermatopathology Unit, Massachusetts General Hospital, 55 Fruit Street, WRN 225, Boston, MA 02114, USA, Tel.: +1 617 726 2967, Fax: +1 617 726 7474,
| | - Andrzej Slominski
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163, USA, Tel.: +1 901 448 6300, Fax: +1 901 448 6979,
| |
Collapse
|
283
|
Carcereny Costa E, Viñolas Segarra N, Gascón Vilaplana P. Angiogenesis inhibitors in the treatment of non-small-cell lung cancer (NSCLC). Clin Transl Oncol 2008; 10:198-203. [PMID: 18411192 DOI: 10.1007/s12094-008-0182-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lung cancer is a leading cause of death worldwide and, although some progress has been made in its treatment, the results remain poor. Better knowledge in tumour biology has allowed us to design anti-target drugs and incorporate them in the treatment of non-small-cell lung cancer (NSCLC). One of the most widely used targeted approaches in this type of tumour has been the inhibition of angiogenesis. Several strategies blocking the VEGF pathway, either at the ligand or recepor level, have been studied and developed. In this review, we present an up-to-date analysis of the current inhibitors of angiogenesis in the treatment of NSCLC.
Collapse
Affiliation(s)
- Enric Carcereny Costa
- Servei d'Oncologia Mèdica, Hospital Clínic (ICMHO), Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.
| | | | | |
Collapse
|
284
|
Rennel ES, Hamdollah-Zadeh MA, Wheatley ER, Magnussen A, Schüler Y, Kelly SP, Finucane C, Ellison D, Cebe-Suarez S, Ballmer-Hofer K, Mather S, Stewart L, Bates DO, Harper SJ. Recombinant human VEGF165b protein is an effective anti-cancer agent in mice. Eur J Cancer 2008; 44:1883-94. [PMID: 18657413 PMCID: PMC2565644 DOI: 10.1016/j.ejca.2008.05.027] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 05/18/2008] [Accepted: 05/21/2008] [Indexed: 01/19/2023]
Abstract
Tumour growth is dependent on angiogenesis, the key mediator of which is vascular endothelial growth factor-A (VEGF-A). VEGF-A exists as two families of alternatively spliced isoforms - pro-angiogenic VEGF(xxx) generated by proximal, and anti-angiogenic VEGF(xxx)b by distal splicing of exon 8. VEGF(165)b inhibits angiogenesis and is downregulated in tumours. Here, we show for the first time that administration of recombinant human VEGF(165)b inhibits colon carcinoma tumour growth and tumour vessel density in nude mice, with a terminal plasma half-life of 6.2h and directly inhibited angiogenic parameters (endothelial sprouting, orientation and structure formation) in vitro. Intravenous injection of (125)I-VEGF(165)b demonstrated significant tumour uptake lasting at least 24h. No adverse effects on liver function or haemodynamics were observed. These results indicate that injected VEGF(165)b was taken up into the tumour as an effective anti-angiogenic cancer therapy, and provide proof of principle for the development of this anti-angiogenic growth factor splice isoform as a novel cancer therapy.
Collapse
Affiliation(s)
- Emma S. Rennel
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, United Kingdom
| | - Maryam A. Hamdollah-Zadeh
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, United Kingdom
| | - Edward R. Wheatley
- Cancer Research Technologies, Cruciform Building, Gower Street, London, United Kingdom
| | - Anette Magnussen
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, United Kingdom
| | - Yvonne Schüler
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, United Kingdom
| | - Sara P. Kelly
- Department of Physiology and Pharmacology, Medical School, University Walk, Bristol BS8 1TD, United Kingdom
| | - Ciara Finucane
- Nuclear Medicine Research Lab, Barts and the London School of Medicine, Queen Mary, London, United Kingdom
| | - David Ellison
- Nuclear Medicine Research Lab, Barts and the London School of Medicine, Queen Mary, London, United Kingdom
| | - Stephanie Cebe-Suarez
- Paul Scherrer Institute, Laboratory of Biomolecular Research, Molecular Cell Biology, 5232 Villigen-PSI, Switzerland
| | - Kurt Ballmer-Hofer
- Paul Scherrer Institute, Laboratory of Biomolecular Research, Molecular Cell Biology, 5232 Villigen-PSI, Switzerland
| | - Stephen Mather
- Nuclear Medicine Research Lab, Barts and the London School of Medicine, Queen Mary, London, United Kingdom
| | - Lorna Stewart
- Cancer Research Technologies, Cruciform Building, Gower Street, London, United Kingdom
| | - David O. Bates
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, United Kingdom
| | - Steven J. Harper
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, United Kingdom
| |
Collapse
|
285
|
Díaz R, Peña C, Silva J, Lorenzo Y, García V, García JM, Sánchez A, Espinosa P, Yuste R, Bonilla F, Domínguez G. p73 isoforms affect VEGF, VEGF165b and PEDF expression in human colorectal tumors: VEGF165b downregulation as a marker of poor prognosis. Int J Cancer 2008; 123:1060-7. [DOI: 10.1002/ijc.23619] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
286
|
Nagy JA, Dvorak AM, Dvorak HF. VEGF-A and the induction of pathological angiogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 2:251-75. [PMID: 18039100 DOI: 10.1146/annurev.pathol.2.010506.134925] [Citation(s) in RCA: 314] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tumors, wounds, and chronic inflammatory disorders generate a new vascular supply by a process known as pathological angiogenesis. Whereas formation of the normal blood vasculature requires the interaction of many different agonists and inhibitors, including vascular endothelial growth factor-A (VEGF-A) and other members of the vascular permeability factor/VEGF family, pathological angiogenesis is a cruder, simpler process that can be replicated by a single VEGF-A isoform, VEGF-A(164/5). VEGF-A(164/5) induces the formation of several distinctly different types of new blood vessels that differ from normal blood vessels with respect to organization, structure, and function. Elucidating the properties of these new vessels has led to a better understanding of angiogenesis and will hopefully lead to new approaches to antiangiogenic therapy.
Collapse
Affiliation(s)
- Janice A Nagy
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | |
Collapse
|
287
|
Grisanti S, Tatar O. The role of vascular endothelial growth factor and other endogenous interplayers in age-related macular degeneration. Prog Retin Eye Res 2008; 27:372-90. [PMID: 18621565 DOI: 10.1016/j.preteyeres.2008.05.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Age-related macular degeneration (AMD) is a multifaceted disease characterized by early subclinical changes at the choroidea-retinal pigment epithelium interface. Both the causal and formal pathogenesis of the disease is still puzzling. Similarly, the reason for progression into two distinct late forms which are "geographic atrophy" and "choroidal neovascularization" remains enigmatic. Late changes are usually responsible for the dramatic loss in central function that has a devastating effect on quality of life. In industrialized countries the disease is a major cause for visual disability among persons over 60 years of age. Due to demographic right-shift and increased life expectancy, AMD is not only a medical problem but will have a pronounced socio-economic effect. Neovascular AMD with the development of choroidal neovascularization in the macular area accounts for 80% of the severe loss of visual acuity due to AMD. In the last decades, treatment modes were merely based on the destruction or surgical removal of the neovascular complex. In the present, however, the philosophical approach to treat the disease is changing to a pathology modifying manner. Intelligent targeting of the involved relevant factors and pathways should stop disease progression, reduce complications and improve vision. The first step into this new era has been accomplished with the introduction of antiangiogenic agents. The new agents act either directly on vascular endothelial growth factor (VEGF) or indirectly on its functional cascade. VEGF makes a fundamental contribution to neovascular processes but it also acts in physiological pathways. The main purpose of this review is to summarize its physiological role especially within the eye, the role in the development of AMD and to understand and foresee both the benefits and potential side-effects of the anti-VEGF-based therapy.
Collapse
Affiliation(s)
- Salvatore Grisanti
- Department of Ophthalmology at the University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany.
| | | |
Collapse
|
288
|
Bott RC, Clopton DT, Cupp AS. A proposed role for VEGF isoforms in sex-specific vasculature development in the gonad. Reprod Domest Anim 2008; 43 Suppl 2:310-6. [PMID: 18638140 PMCID: PMC2708602 DOI: 10.1111/j.1439-0531.2008.01179.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Many scientists have expended efforts to determine what regulates development of an indifferent gonad into either a testis or ovary. Expression of Sry and upregulation of Sox9 are factors that initiate formation of the testis-specific pathway to allow for both sex-specific vasculature and seminiferous cord formation. Migration of mesonephric precursors of peritubular myoid cells and endothelial cells into the differentiating testis is a critical step in formation of both of these structures. Furthermore, these events appear to be initiated downstream from Sry expression. Sertoli cell secretion of growth factors acts to attract these mesonephric cells. One hypothesis is that a growth factor specific for these cell linages act in concert to coordinate migration of both peritubular and endothelial cells. A second hypothesis is that several growth factors stimulate migration and differentiation of mesonephric 'stem-like' cells to result in migration and differentiation into several different cell lineages. While the specific mechanism is unclear, several growth factors have been implicated in the initiation of mesonephric cell migration. This review will focus on the proposed mechanisms of a growth factor, Vascular Endothelial Growth Factor, and how different angiogenic and inhibitory isoforms from this single gene may aid in development of testis-specific vascular development.
Collapse
Affiliation(s)
- R C Bott
- Animal Science Department, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | | | | |
Collapse
|
289
|
Yunker CK, Golembieski W, Lemke N, Schultz CR, Cazacu S, Brodie C, Rempel SA. SPARC-induced increase in glioma matrix and decrease in vascularity are associated with reduced VEGF expression and secretion. Int J Cancer 2008; 122:2735-43. [PMID: 18350569 PMCID: PMC3644882 DOI: 10.1002/ijc.23450] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 12/27/2007] [Indexed: 11/23/2022]
Abstract
Glioblastomas are heterogeneous tumors displaying regions of necrosis, proliferation, angiogenesis, apoptosis and invasion. SPARC, a matricellular protein that negatively regulates angiogenesis and cell proliferation, but enhances cell deadhesion from matrix, is upregulated in gliomas (Grades II-IV). We previously demonstrated that SPARC promotes invasion while concomitantly decreasing tumor growth, in part by decreasing proliferation of the tumor cells. In other cancer types, SPARC has been shown to influence tumor growth by altering matrix production, and by decreasing angiogenesis via interfering with the VEGF-VEGFR1 signaling pathway. We therefore examined whether the SPARC-induced decrease in glioma tumor growth was also, in part, due to alterations in matrix and/or decreased vascularity, and assessed SPARC-VEGF interactions. The data demonstrate that SPARC upregulates glioma matrix, collagen I is a constituent of the matrix and SPARC promotes collagen fibrillogenesis. Furthermore, SPARC suppressed glioma vascularity, and this was accompanied by decreased VEGF expression and secretion, which was, in part, due to reduced VEGF165 transcript abundance. These data indicate that SPARC modulates glioma growth by altering the tumor microenvironment and by suppressing tumor vascularity through suppression of VEGF expression and secretion. These experiments implicate a novel mechanism, whereby SPARC regulates VEGF function by limiting the available growth factor. Because SPARC is considered to be a therapeutic target for gliomas, a further understanding of its complex signaling mechanisms is important, as targeting SPARC to decrease invasion could undesirably lead to the growth of more vascular and proliferative tumors.
Collapse
Affiliation(s)
- Christopher K Yunker
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - William Golembieski
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Nancy Lemke
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Chad R Schultz
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Simona Cazacu
- William and Karen Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Chaya Brodie
- William and Karen Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| | - Sandra A Rempel
- Barbara Jane Levy Laboratory of Molecular Neuro-Oncology, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford HospitalDetroit, MI
| |
Collapse
|
290
|
Kawamura H, Li X, Harper SJ, Bates DO, Claesson-Welsh L. Vascular endothelial growth factor (VEGF)-A165b is a weak in vitro agonist for VEGF receptor-2 due to lack of coreceptor binding and deficient regulation of kinase activity. Cancer Res 2008; 68:4683-92. [PMID: 18559514 DOI: 10.1158/0008-5472.can-07-6577] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Vascular endothelial growth factor (VEGF)-A165b is a COOH-terminal splice variant of VEGF-A that has been implicated in negative regulation of angiogenesis. We compared the properties of VEGF-A165b with those of VEGF-A121, VEGF-A145, and VEGF-A165. Induction of tyrosine phosphorylation sites in VEGFR-2 differed between the VEGF ligands as determined by tryptic phosphopeptide mapping and by use of phosphosite-specific antibodies. VEGF-A165b was considerably poorer in inducing phosphorylation of the positive regulatory site Y1052 in VEGFR-2. Whereas this did not affect activation of VEGFR-2 in vitro, we show that VEGF-A165b failed to induce vasculogenesis and sprouting angiogenesis in differentiating embryonic stem cells and vascularization of s.c. Matrigel plugs. In addition, the ability of the different VEGF ligands to induce angiogenesis correlated with their abilities to bind the VEGF coreceptor neuropilin 1 (NRP1). Our data indicate that loss of VEGFR-2/NRP1 complex formation and Y1052 phosphorylation contribute to the lack of angiogenic properties of VEGF-A165b.
Collapse
Affiliation(s)
- Harukiyo Kawamura
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | | | | |
Collapse
|
291
|
Korgaonkar SN, Feng X, Ross MD, Lu TC, D'Agati V, Iyengar R, Klotman PE, He JC. HIV-1 upregulates VEGF in podocytes. J Am Soc Nephrol 2008; 19:877-83. [PMID: 18443354 DOI: 10.1681/asn.2007050629] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
HIV-associated nephropathy (HIVAN) is characterized by collapsing FSGS. Because transgenic mice with podocyte-specific overexpression of the vascular endothelial growth factor 164 (VEGF164) isoform also develop collapsing FSGS, we sought to determine whether VEGF plays a role in HIVAN. Compared with controls, immunohistochemistry revealed that kidneys from HIV-1-transgenic mice (Tg26) and from patients with HIVAN had greater expression of both VEGF and its transcriptional regulator, hypoxia-inducible factor 2alpha (HIF-2alpha). Similarly, mRNA and protein levels of VEGF and HIF-2alpha were increased in HIV-infected podocytes in vitro, and this transcriptional upregulation was found to be stimulated by the HIV viral protein Nef in a Src kinase-and Stat3-dependent manner. HIV-1 also upregulated VEGFR2 and its co-receptor neuropilin-1 and suppressed the expression of semaphorin 3a in the podocyte. Exogenous VEGF stimulated proliferation and de-differentiation of podocytes, which are features of collapsing FSGS, and VEGFR2 neutralizing antibodies reversed these features in podocytes infected with HIV-1 or isolated from Tg26 mice. In conclusion, HIV-1 induces VEGF and VEGFR2 expression in podocytes, and this may be a critical step in the pathogenesis of HIVAN.
Collapse
|
292
|
Cébe-Suarez S, Grünewald FS, Jaussi R, Li X, Claesson-Welsh L, Spillmann D, Mercer AA, Prota AE, Ballmer-Hofer K. Orf virus VEGF-E NZ2 promotes paracellular NRP-1/VEGFR-2 coreceptor assembly via the peptide RPPR. FASEB J 2008; 22:3078-86. [PMID: 18467594 DOI: 10.1096/fj.08-107219] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factors (VEGFs) interact with the receptor tyrosine kinases (RTKs) VEGFR-1, -2, and -3; neuropilins (NRPs); and heparan sulfate (HS) proteoglycans. VEGF RTKs signal to downstream targets upon ligand-induced tyrosine phosphorylation, while NRPs and HS act as coreceptors that lack enzymatic activity yet modulate signal output by VEGF RTKs. VEGFs exist in various isoforms with distinct receptor specificity and biological activity. Here, a series of mammalian VEGF-A splice variants and orf virus VEGF-Es, as well as chimeric and mutant VEGF variants, were characterized to determine the motifs required for binding to NRP-1 in the absence (VEGF-E) or presence (VEGF-A(165)) of an HS-binding sequence. We identified the carboxyterminal peptides RPPR and DKPRR as the NRP-1 binding motifs of VEGF-E and VEGF-A, respectively. RPPR had significantly higher affinity for NRP-1 than DKPRR. VEGFs containing an RPPR motif promoted HS-independent coreceptor complex assembly between VEGFR-2 and NRP-1, independent of whether these receptors were expressed on the same or separate cells grown in cocultures. Functional studies showed that stable coreceptor assembly by VEGF correlated with its ability to promote vessel formation in an embryoid body angiogenesis assay.
Collapse
Affiliation(s)
- Stéphanie Cébe-Suarez
- Paul Scherrer Institut, Laboratory of Biomolecular Research, Molecular Cell Biology, 5232 Villigen-PSI Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
293
|
Varey AHR, Rennel ES, Qiu Y, Bevan HS, Perrin RM, Raffy S, Dixon AR, Paraskeva C, Zaccheo O, Hassan AB, Harper SJ, Bates DO. VEGF 165 b, an antiangiogenic VEGF-A isoform, binds and inhibits bevacizumab treatment in experimental colorectal carcinoma: balance of pro- and antiangiogenic VEGF-A isoforms has implications for therapy. Br J Cancer 2008; 98:1366-79. [PMID: 18349829 PMCID: PMC2361696 DOI: 10.1038/sj.bjc.6604308] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/31/2008] [Accepted: 02/20/2008] [Indexed: 02/08/2023] Open
Abstract
Bevacizumab, an anti-vascular endothelial growth factor (VEGF-A) antibody, is used in metastatic colorectal carcinoma (CRC) treatment, but responses are unpredictable. Vascular endothelial growth factor is alternatively spliced to form proangiogenic VEGF(165) and antiangiogenic VEGF(165)b. Using isoform-specific enzyme-linked immunosorbent assay and quantitative polymerase chain reaction, we found that over 90% of the VEGF in normal colonic tissue was VEGF(xxx)b, but there was a variable upregulation of VEGF(xxx) and downregulation of VEGF(xxx)b in paired human CRC samples. Furthermore, cultured colonic adenoma cells expressed predominantly VEGF(xxx)b, whereas colonic carcinoma cells expressed predominantly VEGF(xxx). However, adenoma cells exposed to hypoxia switched their expression from predominantly VEGF(xxx)b to predominantly VEGF(xxx). VEGF(165)b overexpression in LS174t colon cancer cells inhibited colon carcinoma growth in mouse xenograft models. Western blotting and surface plasmon resonance showed that VEGF(165)b bound to bevacizumab with similar affinity as VEGF(165). However, although bevacizumab effectively inhibited the rapid growth of colon carcinomas expressing VEGF(165), it did not affect the slower growth of tumours from colonic carcinoma cells expressing VEGF(165)b. Both bevacizumab and anti-VEGF(165)b-specific antibodies were cytotoxic to colonic epithelial cells, but less so to colonic carcinoma cells. These results show that the balance of antiangiogenic to proangiogenic isoforms switches to a variable extent in CRC, regulates tumour growth rates and affects the sensitivity of tumours to bevacizumab by competitive binding. Together with the identification of an autocrine cytoprotective role for VEGF(165)b in colonic epithelial cells, these results indicate that bevacizumab treatment of human CRC may depend upon this balance of VEGF isoforms.
Collapse
Affiliation(s)
- A H R Varey
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - E S Rennel
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Y Qiu
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - H S Bevan
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - R M Perrin
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - S Raffy
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - A R Dixon
- Department of Surgery, Frenchay Hospital, Bristol, UK
| | - C Paraskeva
- Department of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - O Zaccheo
- Department of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - A B Hassan
- Department of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - S J Harper
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - D O Bates
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
294
|
Rennel ES, Waine E, Guan H, Schüler Y, Leenders W, Woolard J, Sugiono M, Gillatt D, Kleinerman ES, Bates DO, Harper SJ. The endogenous anti-angiogenic VEGF isoform, VEGF165b inhibits human tumour growth in mice. Br J Cancer 2008; 98:1250-7. [PMID: 18349828 PMCID: PMC2359649 DOI: 10.1038/sj.bjc.6604309] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/31/2008] [Accepted: 02/20/2008] [Indexed: 11/08/2022] Open
Abstract
Vascular endothelial growth factor-A is widely regarded as the principal stimulator of angiogenesis required for tumour growth. VEGF is generated as multiple isoforms of two families, the pro-angiogenic family generated by proximal splice site selection in the terminal exon, termed VEGFxxx, and the anti-angiogenic family formed by distal splice site selection in the terminal exon, termed VEGFxxxb, where xxx is the amino acid number. The most studied isoforms, VEGF165 and VEGF165b have been shown to be present in tumour and normal tissues respectively. VEGF165b has been shown to inhibit VEGF- and hypoxia-induced angiogenesis, and VEGF-induced cell migration and proliferation in vitro. Here we show that overexpression of VEGF165b by tumour cells inhibits the growth of prostate carcinoma, Ewing's sarcoma and renal cell carcinoma in xenografted mouse tumour models. Moreover, VEGF165b overexpression inhibited tumour cell-mediated migration and proliferation of endothelial cells. These data show that overexpression of VEGF165b can inhibit growth of multiple tumour types in vivo indicating that VEGF165b has potential as an anti-angiogenic, anti-tumour strategy in a number of different tumour types, either by control of VEGF165b expression by regulation of splicing, overexpression of VEGF165b, or therapeutic delivery of VEGF165b to tumours.
Collapse
Affiliation(s)
- E S Rennel
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - E Waine
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - H Guan
- Division of Pediatrics Research, Unit 87, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Y Schüler
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - W Leenders
- Radboud University Nijmegen Medical Centre, Department of Pathology, Nijmegen, The Netherlands
| | - J Woolard
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - M Sugiono
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - D Gillatt
- Bristol Urological Institute, Southmead Hospital, Westbury-on-Trym, Bristol, UK
| | - E S Kleinerman
- Division of Pediatrics Research, Unit 87, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - D O Bates
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| | - S J Harper
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Science, University of Bristol, Southwell Street, Bristol BS2 8EJ, UK
| |
Collapse
|
295
|
Ritchie W, Granjeaud S, Puthier D, Gautheret D. Entropy measures quantify global splicing disorders in cancer. PLoS Comput Biol 2008; 4:e1000011. [PMID: 18369415 PMCID: PMC2268240 DOI: 10.1371/journal.pcbi.1000011] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Accepted: 01/28/2008] [Indexed: 11/19/2022] Open
Abstract
Most mammalian genes are able to express several splice variants in a phenomenon known as alternative splicing. Serious alterations of alternative splicing occur in cancer tissues, leading to expression of multiple aberrant splice forms. Most studies of alternative splicing defects have focused on the identification of cancer-specific splice variants as potential therapeutic targets. Here, we examine instead the bulk of non-specific transcript isoforms and analyze their level of disorder using a measure of uncertainty called Shannon's entropy. We compare isoform expression entropy in normal and cancer tissues from the same anatomical site for different classes of transcript variations: alternative splicing, polyadenylation, and transcription initiation. Whereas alternative initiation and polyadenylation show no significant gain or loss of entropy between normal and cancer tissues, alternative splicing shows highly significant entropy gains for 13 of the 27 cancers studied. This entropy gain is characterized by a flattening in the expression profile of normal isoforms and is correlated to the level of estimated cellular proliferation in the cancer tissue. Interestingly, the genes that present the highest entropy gain are enriched in splicing factors. We provide here the first quantitative estimate of splicing disruption in cancer. The expression of normal splice variants is widely and significantly disrupted in at least half of the cancers studied. We postulate that such splicing disorders may develop in part from splicing alteration in key splice factors, which in turn significantly impact multiple target genes.
Collapse
Affiliation(s)
- William Ritchie
- Université de la Méditerranée, INSERM ERM 206, Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Samuel Granjeaud
- Université de la Méditerranée, INSERM ERM 206, Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Denis Puthier
- Université de la Méditerranée, INSERM ERM 206, Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Daniel Gautheret
- Université Paris-Sud 11, CNRS UMR 8621, Institut de Génétique et Microbiologie, Orsay, France
- * E-mail:
| |
Collapse
|
296
|
Depasquale I, Thompson WD. Prognosis in human melanoma: PAR-1 expression is superior to other coagulation components and VEGF. Histopathology 2008; 52:500-9. [DOI: 10.1111/j.1365-2559.2008.02978.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
297
|
Bello G, Cailotto F, Hanriot D, Kolopp-Sarda MN, Latger-Cannard V, Hess K, Zannad F, Longrois D, Ropars A. C-reactive protein (CRP) increases VEGF-A expression in monocytic cells via a PI3-kinase and ERK 1/2 signaling dependent pathway. Atherosclerosis 2008; 200:286-93. [PMID: 18280482 DOI: 10.1016/j.atherosclerosis.2007.12.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 12/20/2007] [Accepted: 12/21/2007] [Indexed: 11/26/2022]
Abstract
C-reactive protein (CRP) is an independent predictor of atherosclerosis and its complications. Monocytes/macrophages are implicated in this complex disease which is, among other mechanisms, characterised by angiogenesis. The aim of this study was to analyse whether CRP plays a role in VEGF-A regulation by monocytic cells. Our findings show that CRP up-regulates VEGF-A mRNA expression and protein excretion in THP-1 cells in a concentration- and time-dependent manner. Furthermore, we studied the signaling pathway underlying this effect. CRP increases VEGF-A expression via a PI3-kinase and an extracellular-signal-regulated kinase (ERK) 1/2 dependent pathway. Our results suggest that CRP could play a role in the angiogenesis process via immune cells such as monocytes.
Collapse
Affiliation(s)
- Gaëlle Bello
- INSERM, U684, Vandoeuvre les Nancy, F-54500 France
| | | | | | | | | | | | | | | | | |
Collapse
|
298
|
VEGF Signal Tranduction in Angiogenesis. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
299
|
Xie D, Annex BH, Donatucci CF. Growth factors for therapeutic angiogenesis in hypercholesterolemic erectile dysfunction. Asian J Androl 2008; 10:23-7. [DOI: 10.1111/j.1745-7262.2008.00372.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
300
|
Collinson FJ, Hall GD, Perren TJ, Jayson GC. Development of antiangiogenic agents for ovarian cancer. Expert Rev Anticancer Ther 2008; 8:21-32. [PMID: 18095880 DOI: 10.1586/14737140.8.1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Epithelial ovarian cancer (EOC) remains a major source of cancer morbidity and mortality, despite advances in surgical and chemotherapeutic management. The molecular pathways that control angiogenesis have been demonstrated to be key to the pathogenesis of EOC, and have been shown to have prognostic significance. Increased understanding of the pathways and molecules involved in angiogenesis has allowed the identification of a number of targets for antiangiogenic therapies and the development of a variety of antiangiogenic drugs. There is now significant preclinical evidence, and a growing body of clinical data, demonstrating promising results with antiangiogenic drugs in the treatment of EOC. Single-agent VEGF inhibitor response rates in pretreated patients of between 15 and 20% have been reported, with much higher response rates when used in combination with chemotherapeutic agents. These benefits, however, must be balanced with the toxicities associated with these drugs, particularly the more serious ones, such as gastrointestinal perforation. The results of ongoing and future randomized clinical trials will confirm if, and how, antiangiogenic therapies should be integrated into the routine management of EOC. However, critical issues, such as the relative importance of combination remission induction regimens and maintenance therapy, remain poorly defined.
Collapse
Affiliation(s)
- Fiona J Collinson
- Department of Medical Oncology, St James' University Hospital, Leeds, UK.
| | | | | | | |
Collapse
|