101
|
Jun JC, Rathore A, Younas H, Gilkes D, Polotsky VY. Hypoxia-Inducible Factors and Cancer. CURRENT SLEEP MEDICINE REPORTS 2017. [PMID: 28944164 DOI: 10.1007/s40675-017-0062-7.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
PURPOSE OF REVIEW Hypoxia inducible factors (HIFs) mediate the transcription of hundreds of genes that allow cells to adapt to hypoxic environments. In this review, we summarize the current state of knowledge about mechanisms of HIF activation in cancer, as well as downstream cancer-promoting consequences such as altered substrate metabolism, angiogenesis, and cell differentiation. In addition, we examine the proposed relationship between respiratory-related hypoxia, HIFs, and cancer. RECENT FINDINGS HIFs are increased in many forms of cancer, and portend a poor prognosis and response to therapy. CONCLUSION HIFs play a critical role in various stages of carcinogenesis. HIF and its transcription targets may be useful as biomarkers of disease and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Jonathan C Jun
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Aman Rathore
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Haris Younas
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Daniele Gilkes
- Division of Breast Cancer, Department of Oncology, Johns Hopkins University, Baltimore, MD
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
102
|
Barratt SL, Blythe T, Jarrett C, Ourradi K, Shelley-Fraser G, Day MJ, Qiu Y, Harper S, Maher TM, Oltean S, Hames TJ, Scotton CJ, Welsh GI, Bates DO, Millar AB. Differential Expression of VEGF-A xxx Isoforms Is Critical for Development of Pulmonary Fibrosis. Am J Respir Crit Care Med 2017; 196:479-493. [PMID: 28661183 DOI: 10.1164/rccm.201603-0568oc] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Fibrosis after lung injury is related to poor outcome, and idiopathic pulmonary fibrosis (IPF) can be regarded as an exemplar. Vascular endothelial growth factor (VEGF)-A has been implicated in this context, but there are conflicting reports as to whether it is a contributory or protective factor. Differential splicing of the VEGF-A gene produces multiple functional isoforms including VEGF-A165a and VEGF-A165b, a member of the inhibitory family. To date there is no clear information on the role of VEGF-A in IPF. OBJECTIVES To establish VEGF-A isoform expression and functional effects in IPF. METHODS We used tissue sections, plasma, and lung fibroblasts from patients with IPF and control subjects. In a bleomycin-induced lung fibrosis model we used wild-type MMTV mice and a triple transgenic mouse SPC-rtTA+/-TetoCre+/-LoxP-VEGF-A+/+ to conditionally induce VEGF-A isoform deletion specifically in the alveolar type II (ATII) cells of adult mice. MEASUREMENTS AND MAIN RESULTS IPF and normal lung fibroblasts differentially expressed and responded to VEGF-A165a and VEGF-A165b in terms of proliferation and matrix expression. Increased VEGF-A165b was detected in plasma of progressing patients with IPF. In a mouse model of pulmonary fibrosis, ATII-specific deficiency of VEGF-A or constitutive overexpression of VEGF-A165b inhibited the development of pulmonary fibrosis, as did treatment with intraperitoneal delivery of VEGF-A165b to wild-type mice. CONCLUSIONS These results indicate that changes in the bioavailability of VEGF-A sourced from ATII cells, namely the ratio of VEGF-Axxxa to VEGF-Axxxb, are critical in development of pulmonary fibrosis and may be a paradigm for the regulation of tissue repair.
Collapse
Affiliation(s)
| | - Thomas Blythe
- 1 Academic Respiratory Unit, School of Clinical Sciences
| | | | | | - Golda Shelley-Fraser
- 2 Department of Histopathology, Cheltenham and Gloucestershire NHS Trust, Cheltenham, United Kingdom
| | | | | | | | - Toby M Maher
- 5 NIHR Respiratory Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Sebastian Oltean
- 6 Department of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Thomas J Hames
- 7 University of Exeter Medical School, Exeter, United Kingdom; and
| | - Chris J Scotton
- 7 University of Exeter Medical School, Exeter, United Kingdom; and
| | | | - David O Bates
- 8 Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ann B Millar
- 1 Academic Respiratory Unit, School of Clinical Sciences
| |
Collapse
|
103
|
Dynamics and implications of circulating anti-angiogenic VEGF-A 165b isoform in patients with ST-elevation myocardial infarction. Sci Rep 2017; 7:9962. [PMID: 28855597 PMCID: PMC5577291 DOI: 10.1038/s41598-017-10505-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis is crucial to restore microvascular perfusion in the jeopardized myocardium in the weeks following reperfused ST-segment elevation myocardial infarction (STEMI). (VEGF)-A165b, an anti-angiogenic factor, has been identified as a regulator of vascularization; however, it has not been previously implicated in acute myocardial infarction. We sought to investigate the dynamics of circulating VEGF-A165b and its association with cardiac magnetic resonance-derived infarct size and left ventricular ejection fraction (LVEF). 50 STEMI patients and 23 controls were included. Compared with control individuals, serum VEGF-A165b was elevated in STEMI patients prior to primary percutaneous coronary intervention (PCI). Following PCI, serum VEGF-A165b increased further, reaching a maximum level at 24 h and decreased one month after reperfusion. VEGF-A165b levels at 24 h were associated with a large infarct size and inversely related to LVEF. VEGF-A165b expression was increased in myocardial infarct areas from patients with previous history of AMI. An ex vivo assay using serum from STEMI patients showed that neutralization of VEGF-A165b increased tubulogenesis. Overall, the study suggests that VEGF-A165b might play a deleterious role after AMI as an inhibitor of angiogenesis in the myocardium. Accordingly, neutralization of VEGF-A165b could represent a novel pro-angiogenic therapy for reperfusion of myocardium in STEMI.
Collapse
|
104
|
Biselli-Chicote PM, Biselli JM, Cunha BR, Castro R, Maniglia JV, Neto DDS, Tajara EH, Góis Filho JFD, Fukuyama EE, Pavarino ÉC, Goloni-Bertollo EM. Overexpression of Antiangiogenic Vascular Endothelial Growth Factor Isoform and Splicing Regulatory Factors in Oral, Laryngeal and Pharyngeal Squamous Cell Carcinomas. Asian Pac J Cancer Prev 2017; 18:2171-2177. [PMID: 28843252 PMCID: PMC5697477 DOI: 10.22034/apjcp.2017.18.8.2171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Overexpression of proangiogenic vascular endothelial growth factor A family VEGFAxxx is associated with tumor growth and metastasis. The role of the alternatively spliced antiangiogenic family VEGFAxxxb is poorly investigated in head and neck squamous cell carcinomas (HNSCCs). The antiangiogenic isoform binds to bevacizumab and its expression level could influence the treatment response and progression-free survival. In this study, the relative expression of VEGFAxxx and VEGFA165b isoforms and splicing regulatory factors genes was investigated in a series of HNSCCs. Methods: VEGFAxxx, VEGFA165b, SRSF6, SRSF5, SRSF1 and SRPK1 gene expression was quantified by quantitative real time PCR in 53 tissue samples obtained by surgery from HNSCC patients. Protein expression was evaluated by immunohistochemistry. Results: VEGFAxxx and VEGFA165b were overexpressed in HNSCCs. Elevated protein expression was also confirmed. However, VEGFA isoforms demonstrated differential expression according to anatomical sites. VEGFAxxx was overexpressed in pharyngeal tumors while the VEGFA165b isoform was up-regulated in oral tumors. The VEGFA165b isoform was also positively correlated with expression of the splicing regulatory genes SRSF1, SRSF6 and SRSF5. Conclusions: We concluded that VEGFAxxx and VEGFA165b isoforms are overexpressed in HNSCCs and the splicing regulatory factors SRSF1, SRSF6, SRSF5 and SRPK1 may contribute to alternative splicing of the VEGFA gene. The findings for the differential expression of the antiangiogenic isoform in HNSCCs could facilitate effective therapeutic strategies for the management of these tumors.
Collapse
|
105
|
Pruszko M, Milano E, Forcato M, Donzelli S, Ganci F, Di Agostino S, De Panfilis S, Fazi F, Bates DO, Bicciato S, Zylicz M, Zylicz A, Blandino G, Fontemaggi G. The mutant p53-ID4 complex controls VEGFA isoforms by recruiting lncRNA MALAT1. EMBO Rep 2017; 18:1331-1351. [PMID: 28652379 PMCID: PMC5538427 DOI: 10.15252/embr.201643370] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022] Open
Abstract
The abundant, nuclear-retained, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been associated with a poorly differentiated and aggressive phenotype of mammary carcinomas. This long non-coding RNA (lncRNA) localizes to nuclear speckles, where it interacts with a subset of splicing factors and modulates their activity. In this study, we demonstrate that oncogenic splicing factor SRSF1 bridges MALAT1 to mutant p53 and ID4 proteins in breast cancer cells. Mutant p53 and ID4 delocalize MALAT1 from nuclear speckles and favor its association with chromatin. This enables aberrant recruitment of MALAT1 on VEGFA pre-mRNA and modulation of VEGFA isoforms expression. Interestingly, VEGFA-dependent expression signatures associate with ID4 expression specifically in basal-like breast cancers carrying TP53 mutations. Our results highlight a key role for MALAT1 in control of VEGFA isoforms expression in breast cancer cells expressing gain-of-function mutant p53 and ID4 proteins.
Collapse
Affiliation(s)
- Magdalena Pruszko
- Department of Molecular Biology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, PAS, Warsaw, Poland
| | - Elisa Milano
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Mattia Forcato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Federica Ganci
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Silvia Di Agostino
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Simone De Panfilis
- Centre for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - David O Bates
- Division of Cancer and Stem Cells, Cancer Biology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Maciej Zylicz
- Department of Molecular Biology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Alicja Zylicz
- Department of Molecular Biology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| | - Giulia Fontemaggi
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute "Regina Elena", Rome, Italy
| |
Collapse
|
106
|
Reina-Torres E, Wen JC, Liu KC, Li G, Sherwood JM, Chang JYH, Challa P, Flügel-Koch CM, Stamer WD, Allingham RR, Overby DR. VEGF as a Paracrine Regulator of Conventional Outflow Facility. Invest Ophthalmol Vis Sci 2017; 58:1899-1908. [PMID: 28358962 PMCID: PMC5374885 DOI: 10.1167/iovs.16-20779] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Vascular endothelial growth factor (VEGF) regulates microvascular endothelial permeability, and the permeability of Schlemm's canal (SC) endothelium influences conventional aqueous humor outflow. We hypothesize that VEGF signaling regulates outflow facility. Methods We measured outflow facility (C) in enucleated mouse eyes perfused with VEGF-A164a, VEGF-A165b, VEGF-D, or inhibitors to VEGF receptor 2 (VEGFR-2). We monitored VEGF-A secretion from human trabecular meshwork (TM) cells by ELISA after 24 hours of static culture or cyclic stretch. We used immunofluorescence microscopy to localize VEGF-A protein within the TM of mice. Results VEGF-A164a increased C in enucleated mouse eyes. Cyclic stretch increased VEGF-A secretion by human TM cells, which corresponded to VEGF-A localization in the TM of mice. Blockade of VEGFR-2 decreased C, using either of the inhibitors SU5416 or Ki8751 or the inactive splice variant VEGF-A165b. VEGF-D increased C, which could be blocked by Ki8751. Conclusions VEGF is a paracrine regulator of conventional outflow facility that is secreted by TM cells in response to mechanical stress. VEGF affects facility via VEGFR-2 likely at the level of SC endothelium. Disruption of VEGF signaling in the TM may explain why anti-VEGF therapy is associated with decreased outflow facility and sustained ocular hypertension.
Collapse
Affiliation(s)
- Ester Reina-Torres
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Joanne C Wen
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Katy C Liu
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Guorong Li
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Joseph M Sherwood
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Jason Y H Chang
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Pratap Challa
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Cassandra M Flügel-Koch
- Department of Anatomy II, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - R Rand Allingham
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
107
|
Ye X, Abou-Rayyah Y, Bischoff J, Ritchie A, Sebire NJ, Watts P, Churchill AJ, Bates DO. Altered ratios of pro- and anti-angiogenic VEGF-A variants and pericyte expression of DLL4 disrupt vascular maturation in infantile haemangioma. J Pathol 2017; 239:139-51. [PMID: 26957058 PMCID: PMC4869683 DOI: 10.1002/path.4715] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 02/27/2016] [Accepted: 03/03/2016] [Indexed: 12/26/2022]
Abstract
Infantile haemangioma (IH), the most common neoplasm in infants, is a slowly resolving vascular tumour. Vascular endothelial growth factor A (VEGF‐A), which consists of both the pro‐ and anti‐angiogenic variants, contributes to the pathogenesis of IH. However, the roles of different VEGF‐A variants in IH progression and its spontaneous involution is unknown. Using patient‐derived cells and surgical specimens, we showed that the relative level of VEGF‐A165b was increased in the involuting phase of IH and the relative change in VEGF‐A isoforms may be dependent on endothelial differentiation of IH stem cells. VEGFR signalling regulated IH cell functions and VEGF‐A165b inhibited cell proliferation and the angiogenic potential of IH endothelial cells in vitro and in vivo. The inhibition of angiogenesis by VEGF‐A165b was associated with the extent of VEGF receptor 2 (VEGFR2) activation and degradation and Delta‐like ligand 4 (DLL4) expression. These results indicate that VEGF‐A variants can be regulated by cell differentiation and are involved in IH progression. We also demonstrated that DLL4 expression was not exclusive to the endothelium in IH but was also present in pericytes, where the expression of VEGFR2 is absent, suggesting that pericyte‐derived DLL4 may prevent sprouting during involution, independently of VEGFR2. Angiogenesis in IH therefore appears to be controlled by DLL4 within the endothelium in a VEGF‐A isoform‐dependent manner, and in perivascular cells in a VEGF‐independent manner. The contribution of VEGF‐A isoforms to disease progression also indicates that IH may be associated with altered splicing. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xi Ye
- Ophthalmology Unit, School of Clinical Sciences, University of Bristol, UK.,Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, UK
| | | | - Joyce Bischoff
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, MA, USA
| | - Alison Ritchie
- Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, UK
| | - Neil J Sebire
- Histopathology, Great Ormond Street Hospital, London, UK
| | | | - Amanda J Churchill
- Ophthalmology Unit, School of Clinical Sciences, University of Bristol, UK
| | - David O Bates
- Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, UK
| |
Collapse
|
108
|
Kilpatrick LE, Friedman-Ohana R, Alcobia DC, Riching K, Peach CJ, Wheal AJ, Briddon SJ, Robers MB, Zimmerman K, Machleidt T, Wood KV, Woolard J, Hill SJ. Real-time analysis of the binding of fluorescent VEGF 165a to VEGFR2 in living cells: Effect of receptor tyrosine kinase inhibitors and fate of internalized agonist-receptor complexes. Biochem Pharmacol 2017; 136:62-75. [PMID: 28392095 PMCID: PMC5457915 DOI: 10.1016/j.bcp.2017.04.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/04/2017] [Indexed: 01/01/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an important mediator of angiogenesis. Here we have used a novel stoichiometric protein-labeling method to generate a fluorescent variant of VEGF (VEGF165a-TMR) labeled on a single cysteine within each protomer of the antiparallel VEGF homodimer. VEGF165a-TMR has then been used in conjunction with full length VEGFR2, tagged with the bioluminescent protein NanoLuc, to undertake a real time quantitative evaluation of VEGFR2 binding characteristics in living cells using bioluminescence resonance energy transfer (BRET). This provided quantitative information on VEGF-VEGFR2 interactions. At longer incubation times, VEGFR2 is internalized by VEGF165a-TMR into intracellular endosomes. This internalization can be prevented by the receptor tyrosine kinase inhibitors (RTKIs) cediranib, sorafenib, pazopanib or vandetanib. In the absence of RTKIs, the BRET signal is decreased over time as a consequence of the dissociation of agonist from the receptor in intracellular endosomes and recycling of VEGFR2 back to the plasma membrane.
Collapse
Affiliation(s)
- Laura E Kilpatrick
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Diana C Alcobia
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Chloe J Peach
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Amanda J Wheal
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Stephen J Briddon
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | | - Jeanette Woolard
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.
| | - Stephen J Hill
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
109
|
Vascular endothelial growth factor-A 165b ameliorates outer-retinal barrier and vascular dysfunction in the diabetic retina. Clin Sci (Lond) 2017; 131:1225-1243. [PMID: 28341661 PMCID: PMC5450016 DOI: 10.1042/cs20170102] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/09/2017] [Accepted: 03/24/2017] [Indexed: 01/11/2023]
Abstract
Diabetic retinopathy (DR) is one of the leading causes of blindness in the developed world. Characteristic features of DR are retinal neurodegeneration, pathological angiogenesis and breakdown of both the inner and outer retinal barriers of the retinal vasculature and retinal pigmented epithelial (RPE)–choroid respectively. Vascular endothelial growth factor (VEGF-A), a key regulator of angiogenesis and permeability, is the target of most pharmacological interventions of DR. VEGF-A can be alternatively spliced at exon 8 to form two families of isoforms, pro- and anti-angiogenic. VEGF-A165a is the most abundant pro-angiogenic isoform, is pro-inflammatory and a potent inducer of permeability. VEGF-A165b is anti-angiogenic, anti-inflammatory, cytoprotective and neuroprotective. In the diabetic eye, pro-angiogenic VEGF-A isoforms are up-regulated such that they overpower VEGF-A165b. We hypothesized that this imbalance may contribute to increased breakdown of the retinal barriers and by redressing this imbalance, the pathological angiogenesis, fluid extravasation and retinal neurodegeneration could be ameliorated. VEGF-A165b prevented VEGF-A165a and hyperglycaemia-induced tight junction (TJ) breakdown and subsequent increase in solute flux in RPE cells. In streptozotocin (STZ)-induced diabetes, there was an increase in Evans Blue extravasation after both 1 and 8 weeks of diabetes, which was reduced upon intravitreal and systemic delivery of recombinant human (rh)VEGF-A165b. Eight-week diabetic rats also showed an increase in retinal vessel density, which was prevented by VEGF-A165b. These results show rhVEGF-A165b reduces DR-associated blood–retina barrier (BRB) dysfunction, angiogenesis and neurodegeneration and may be a suitable therapeutic in treating DR.
Collapse
|
110
|
Mechanisms regulating angiogenesis underlie seasonal control of pituitary function. Proc Natl Acad Sci U S A 2017; 114:E2514-E2523. [PMID: 28270617 DOI: 10.1073/pnas.1618917114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Seasonal changes in mammalian physiology, such as those affecting reproduction, hibernation, and metabolism, are controlled by pituitary hormones released in response to annual environmental changes. In temperate zones, the primary environmental cue driving seasonal reproductive cycles is the change in day length (i.e., photoperiod), encoded by the pattern of melatonin secretion from the pineal gland. However, although reproduction relies on hypothalamic gonadotrophin-releasing hormone output, and most cells producing reproductive hormones are in the pars distalis (PD) of the pituitary, melatonin receptors are localized in the pars tuberalis (PT), a physically and functionally separate part of the gland. How melatonin in the PT controls the PD is not understood. Here we show that melatonin time-dependently acts on its receptors in the PT to alter splicing of vascular endothelial growth factor (VEGF). Outside the breeding season (BS), angiogenic VEGF-A stimulates vessel growth in the infundibulum, aiding vascular communication among the PT, PD, and brain. This also acts on VEGF receptor 2 (VEGFR2) expressed in PD prolactin-producing cells known to impair gonadotrophin secretion. In contrast, in the BS, melatonin releases antiangiogenic VEGF-Axxxb from the PT, inhibiting infundibular angiogenesis and diminishing lactotroph (LT) VEGFR2 expression, lifting reproductive axis repression in response to shorter day lengths. The time-dependent, melatonin-induced differential expression of VEGF-A isoforms culminates in alterations in gonadotroph function opposite to those of LTs, with up-regulation and down-regulation of gonadotrophin gene expression during the breeding and nonbreeding seasons, respectively. These results provide a mechanism by which melatonin can control pituitary function in a seasonal manner.
Collapse
|
111
|
RNA-Sequencing data supports the existence of novel VEGFA splicing events but not of VEGFA xxxb isoforms. Sci Rep 2017; 7:58. [PMID: 28246395 PMCID: PMC5427905 DOI: 10.1038/s41598-017-00100-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/06/2017] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor (VEGFA), a pivotal regulator of angiogenesis and valuable therapeutic target, is characterised by alternative splicing which generates three principal isoforms, VEGFA121, VEGFA165 and VEGFA189. A second set of anti-angiogenic isoforms termed VEGFAxxxb that utilise an alternative splice site in the final exon have been widely reported, with mRNA detection based principally upon RT-PCR assays. We sought confirmation of the existence of the VEGFAxxxb isoforms within the abundant RNA sequencing data available publicly. Whilst sequences derived specifically from each of the canonical VEGFA isoforms were present in many tissues, there were no sequences derived from VEGFAxxxb isoforms. Sequencing of approximately 50,000 RT-PCR products spanning the exon 7–8 junction in 10 tissues did not identify any VEGFAxxxb transcripts. The absence or extremely low expression of these transcripts in vivo indicates that VEGFAxxxb isoforms are unlikely to play a role in normal physiology. Our analyses also revealed multiple novel splicing events supported by more reads than previously reported for VEGFA145 and VEGFA148 isoforms, including three from novel first exons consistent with existing transcription start site data. These novel VEGFA isoforms may play significant roles in specific cell types.
Collapse
|
112
|
Jun JC, Rathore A, Younas H, Gilkes D, Polotsky VY. Hypoxia-Inducible Factors and Cancer. CURRENT SLEEP MEDICINE REPORTS 2017; 3:1-10. [PMID: 28944164 DOI: 10.1007/s40675-017-0062-7] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Hypoxia inducible factors (HIFs) mediate the transcription of hundreds of genes that allow cells to adapt to hypoxic environments. In this review, we summarize the current state of knowledge about mechanisms of HIF activation in cancer, as well as downstream cancer-promoting consequences such as altered substrate metabolism, angiogenesis, and cell differentiation. In addition, we examine the proposed relationship between respiratory-related hypoxia, HIFs, and cancer. RECENT FINDINGS HIFs are increased in many forms of cancer, and portend a poor prognosis and response to therapy. CONCLUSION HIFs play a critical role in various stages of carcinogenesis. HIF and its transcription targets may be useful as biomarkers of disease and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Jonathan C Jun
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Aman Rathore
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Haris Younas
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Daniele Gilkes
- Division of Breast Cancer, Department of Oncology, Johns Hopkins University, Baltimore, MD
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
113
|
Majumder S, Advani A. VEGF and the diabetic kidney: More than too much of a good thing. J Diabetes Complications 2017; 31:273-279. [PMID: 27836681 DOI: 10.1016/j.jdiacomp.2016.10.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/18/2016] [Indexed: 02/06/2023]
Abstract
Over a decade and a half has passed since the publication of early reports hinting at a pathogenetic role for vascular endothelial growth factor ("VEGF") in the development of diabetic kidney disease. In diabetic rats, renal mRNA levels of the VEGF-A isoform were upregulated and administration of a VEGF-A neutralizing antibody attenuated albuminuria: VEGF was "bad" in diabetic nephropathy. Since that time, our understanding of the complexity of the renal VEGF system has advanced. Unlike its experimental counterpart, human diabetic nephropathy is associated with diminished VEGF-A levels and experience in the oncological setting has taught us that VEGF blocking therapy can cause adverse renal effects in patients. Correspondingly, investigational studies in cultured cells and rodent models have demonstrated that the biological effects of the VEGF system are dependent not only on the amount of VEGF, but also the type of VEGF, its sites of action and the prevailing milieu. Here we reflect back on the discoveries that have been made since those initial reports that shone the spotlight on the importance of the VEGF system in the diabetic kidney and we consider that the role of VEGF in diabetic nephropathy extends well beyond being "too much of a good thing".
Collapse
Affiliation(s)
- Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
114
|
Lapeyre-Prost A, Terme M, Pernot S, Pointet AL, Voron T, Tartour E, Taieb J. Immunomodulatory Activity of VEGF in Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:295-342. [PMID: 28215534 DOI: 10.1016/bs.ircmb.2016.09.007] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability of tumor cells to escape tumor immunosurveillance contributes to cancer development. Factors produced in the tumor microenvironment create "tolerizing" conditions and thereby help the tumor to evade antitumoral immune responses. VEGF-A, already known for its major role in tumor vessel growth (neoangiogenesis), was recently identified as a key factor in tumor-induced immunosuppression. In particular, VEGF-A fosters the proliferation of immunosuppressive cells, limits T-cell recruitment into tumors, and promotes T-cell exhaustion. Antiangiogenic therapies have shown significant efficacy in patients with a variety of solid tumors, preventing tumor progression by limiting tumor-induced angiogenesis. VEGF-targeting therapies have also been shown to modulate the tumor-induced immunosuppressive microenvironment, enhancing Th1-type T-cell responses and increasing tumor infiltration by T cells. The immunomodulatory properties of VEGF-targeting therapies open up new perspectives for cancer treatment, especially through strategies combining antiangiogenic drugs with immunotherapy. Preclinical models and early clinical studies of these combined approaches have given promising results.
Collapse
Affiliation(s)
- A Lapeyre-Prost
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France
| | - M Terme
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France.
| | - S Pernot
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'hépatogastroentérologie et d'oncologie digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - A-L Pointet
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'hépatogastroentérologie et d'oncologie digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - T Voron
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service de chirurgie digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - E Tartour
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'immunologie biologique. Hôpital Européen Georges Pompidou, Paris, France
| | - J Taieb
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'hépatogastroentérologie et d'oncologie digestive, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
115
|
Ganta VC, Choi M, Kutateladze A, Annex BH. VEGF165b Modulates Endothelial VEGFR1-STAT3 Signaling Pathway and Angiogenesis in Human and Experimental Peripheral Arterial Disease. Circ Res 2016; 120:282-295. [PMID: 27974423 DOI: 10.1161/circresaha.116.309516] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 11/22/2016] [Accepted: 12/14/2016] [Indexed: 01/25/2023]
Abstract
RATIONALE Atherosclerotic-arterial occlusions decrease tissue perfusion causing ischemia to lower limbs in patients with peripheral arterial disease (PAD). Ischemia in muscle induces an angiogenic response, but the magnitude of this response is frequently inadequate to meet tissue perfusion requirements. Alternate splicing in the exon-8 of vascular endothelial growth factor (VEGF)-A results in production of proangiogenic VEGFxxxa isoforms (VEGF165a, 165 for the 165 amino acid product) and antiangiogenic VEGFxxxb (VEGF165b) isoforms. OBJECTIVE The antiangiogenic VEGFxxxb isoforms are thought to antagonize VEGFxxxa isoforms and decrease activation of VEGF receptor-2 (VEGFR2), hereunto considered the dominant receptor in postnatal angiogenesis in PAD. Our data will show that VEGF165b inhibits VEGFR1 signal transducer and activator of transcription (STAT)-3 signaling to decrease angiogenesis in human and experimental PAD. METHODS AND RESULTS In human PAD versus control muscle biopsies, VEGF165b: (1) is elevated, (2) is bound higher (versus VEGF165a) to VEGFR1 not VEGFR2, and (3) levels correlated with decreased VEGFR1, not VEGFR2, activation. In experimental PAD, delivery of an isoform-specific monoclonal antibody to VEGF165b versus control antibody enhanced perfusion in animal model of severe PAD (Balb/c strain) without activating VEGFR2 signaling but with increased VEGFR1 activation. Receptor pull-down experiments demonstrate that VEGF165b inhibition versus control increased VEGFR1-STAT3 binding and STAT3 activation, independent of Janus-activated kinase-1)/Janus-activated kinase-2. Using VEGFR1+/- mice that could not increase VEGFR1 after ischemia, we confirm that VEGF165b decreases VEGFR1-STAT3 signaling to decrease perfusion. CONCLUSIONS Our results indicate that VEGF165b prevents activation of VEGFR1-STAT3 signaling by VEGF165a and hence inhibits angiogenesis and perfusion recovery in PAD muscle.
Collapse
Affiliation(s)
- Vijay Chaitanya Ganta
- From the Cardiovascular Research Center (V.C.G., M.C., B.H.A.), Department of Biology (A.K.), and Department of Cardiovascular Medicine, University of Virginia, Charlottesville (B.H.A.)
| | - Min Choi
- From the Cardiovascular Research Center (V.C.G., M.C., B.H.A.), Department of Biology (A.K.), and Department of Cardiovascular Medicine, University of Virginia, Charlottesville (B.H.A.)
| | - Anna Kutateladze
- From the Cardiovascular Research Center (V.C.G., M.C., B.H.A.), Department of Biology (A.K.), and Department of Cardiovascular Medicine, University of Virginia, Charlottesville (B.H.A.)
| | - Brian H Annex
- From the Cardiovascular Research Center (V.C.G., M.C., B.H.A.), Department of Biology (A.K.), and Department of Cardiovascular Medicine, University of Virginia, Charlottesville (B.H.A.).
| |
Collapse
|
116
|
Chu LH, Ganta VC, Choi MH, Chen G, Finley SD, Annex BH, Popel AS. A multiscale computational model predicts distribution of anti-angiogenic isoform VEGF 165b in peripheral arterial disease in human and mouse. Sci Rep 2016; 6:37030. [PMID: 27853189 PMCID: PMC5113071 DOI: 10.1038/srep37030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/24/2016] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is the growth of new blood vessels from pre-existing microvessels. Peripheral arterial disease (PAD) is caused by atherosclerosis that results in ischemia mostly in the lower extremities. Clinical trials including VEGF-A administration for therapeutic angiogenesis have not been successful. The existence of anti-angiogenic isoform (VEGF165b) in PAD muscle tissues is a potential cause for the failure of therapeutic angiogenesis. Experimental measurements show that in PAD human muscle biopsies the VEGF165b isoform is at least as abundant if not greater than the VEGF165a isoform. We constructed three-compartment models describing VEGF isoforms and receptors, in human and mouse, to make predictions on the secretion rate of VEGF165b and the distribution of various isoforms throughout the body based on the experimental data. The computational results are consistent with the data showing that in PAD calf muscles secrete mostly VEGF165b over total VEGF. In the PAD calf compartment of human and mouse models, most VEGF165a and VEGF165b are bound to the extracellular matrix. VEGF receptors VEGFR1, VEGFR2 and Neuropilin-1 (NRP1) are mostly in ‘Free State’. This study provides a computational model of VEGF165b in PAD supported by experimental measurements of VEGF165b in human and mouse, which gives insight of VEGF165b in therapeutic angiogenesis and VEGF distribution in human and mouse PAD model.
Collapse
Affiliation(s)
- Liang-Hui Chu
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Vijay Chaitanya Ganta
- Cardiovascular Medicine, Department of Medicine, and the Robert M. Berne Cardiovascular Research Center University of Virginia School of Medicine, Charlottesville, VA 22901, United States
| | - Min H Choi
- Cardiovascular Medicine, Department of Medicine, and the Robert M. Berne Cardiovascular Research Center University of Virginia School of Medicine, Charlottesville, VA 22901, United States
| | - George Chen
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Brian H Annex
- Cardiovascular Medicine, Department of Medicine, and the Robert M. Berne Cardiovascular Research Center University of Virginia School of Medicine, Charlottesville, VA 22901, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
117
|
Association between levels of anti-angiogenic isoform of vascular endothelial growth factor A and pulmonary hypertension. Int J Cardiol 2016; 222:416-420. [DOI: 10.1016/j.ijcard.2016.07.277] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/14/2016] [Accepted: 07/30/2016] [Indexed: 01/01/2023]
|
118
|
Simão S, Santos DF, Silva GA. Aliskiren decreases oxidative stress and angiogenic markers in retinal pigment epithelium cells. Angiogenesis 2016; 20:175-181. [DOI: 10.1007/s10456-016-9526-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/27/2016] [Indexed: 12/17/2022]
|
119
|
Xin H, Zhong C, Nudleman E, Ferrara N. Evidence for Pro-angiogenic Functions of VEGF-Ax. Cell 2016; 167:275-284.e6. [PMID: 27662093 DOI: 10.1016/j.cell.2016.08.054] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/07/2016] [Accepted: 08/19/2016] [Indexed: 01/21/2023]
Abstract
The VEGF-A isoforms play a crucial role in vascular development, and the VEGF signaling pathway is a clinically validated therapeutic target for several pathological conditions. Alternative mRNA splicing leads to the generation of multiple VEGF-A isoforms, including VEGF165. A recent study reported the presence of another isoform, VEGF-Ax, arising from programmed readthrough translation. Compared to VEGF165, VEGF-Ax has a 22-amino-acid extension in the COOH terminus and has been reported to function as a negative regulator of VEGF signaling in endothelial cells, with potent anti-angiogenic effects. Here, we show that, contrary to the earlier report, VEGF-Ax stimulates endothelial cell mitogenesis, angiogenesis, as well as vascular permeability. Accordingly, VEGF-Ax induces phosphorylation of key tyrosine residues in VEGFR-2. Notably, VEGF-Ax was less potent than VEGF165, consistent with its impaired binding to the VEGF co-receptor neuropilin-1.
Collapse
Affiliation(s)
- Hong Xin
- University of California, San Diego, 3855 Health Sciences Drive #0819, La Jolla, CA 92093, USA
| | - Cuiling Zhong
- University of California, San Diego, 3855 Health Sciences Drive #0819, La Jolla, CA 92093, USA
| | - Eric Nudleman
- University of California, San Diego, 3855 Health Sciences Drive #0819, La Jolla, CA 92093, USA
| | - Napoleone Ferrara
- University of California, San Diego, 3855 Health Sciences Drive #0819, La Jolla, CA 92093, USA.
| |
Collapse
|
120
|
Niderla-Bielińska J, Ciszek B, Jankowska-Steifer E, Flaht-Zabost A, Gula G, Radomska-Leśniewska DM, Ratajska A. Mouse Proepicardium Exhibits a Sprouting Response to Exogenous Proangiogenic Growth Factors in vitro. J Vasc Res 2016; 53:83-93. [DOI: 10.1159/000448685] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/24/2016] [Indexed: 11/19/2022] Open
|
121
|
Tousi A, Hasanpour H, Soheilian M. Intravitreal Injection of Bevacizumab in Primary Vitrectomy to Decrease the Rate of Retinal Redetachment: A Randomized Pilot Study. J Ophthalmic Vis Res 2016; 11:271-6. [PMID: 27621784 PMCID: PMC5000529 DOI: 10.4103/2008-322x.188390] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Purpose: To evaluate the effect of intravitreal bevacizumab (IVB) as a surgical adjunct in prevention of proliferative vitreoretinopathy (PVR) after retinal detachment surgery. Methods: In this controlled, randomized pilot study, 27 patients with primary retinal detachment undergoing pars plana deep vitrectomy were included. Of these, 12 received IVB at the end of procedure. The anatomic success and best corrected visual acuity (BCVA) were compared to the control group at months 3 and 6 postoperatively. Results: At three month follow-up, 3 of 11 eyes (27.3%) had detached retinas in the IVB group versus 6 of 12 (50.0%) in the control group (P = 0.40). At six-month follow-up, 3 of 10 eyes (30%) had detached retinas in the IVB group versus 3 in 8 (37.5%) in the control group (P > 0.99). Mean logMAR BCVA improved significantly in both groups relative to baseline, but did not show a significant difference at three-and six-month follow-ups between the two groups. Conclusion: Our preliminary results show neither a benefit nor any harm from intervention in both anatomic and visual outcomes. Our results support conducting additional studies to evaluate the effect of intravitreal bevacizumab on postoperative PVR.
Collapse
Affiliation(s)
- Adib Tousi
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hasanpour
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soheilian
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
122
|
Jazwa A, Florczyk U, Grochot-Przeczek A, Krist B, Loboda A, Jozkowicz A, Dulak J. Limb ischemia and vessel regeneration: Is there a role for VEGF? Vascul Pharmacol 2016; 86:18-30. [PMID: 27620809 DOI: 10.1016/j.vph.2016.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/24/2016] [Accepted: 09/07/2016] [Indexed: 11/18/2022]
Abstract
Vascular endothelial growth factor (VEGF), as an endothelial cell-specific mitogen, is crucial for new blood vessels formation. Atherosclerosis affecting the cardiovascular system causes ischemia and functio laesa in tissues supplied by the occluded vessels. When such a situation occurs in the lower extremities, it causes critical limb ischemia (CLI) often requiring leg amputation. Low oxygen tension leads to upregulation of hypoxia-regulated genes (i.e. VEGF), that should help to restore the impaired blood flow. In CLI these rescue mechanisms are, however, often inefficient. Moreover, there are many contradictory reports showing either induction, no changes or even down-regulation of VEGF in specimens taken from patients with CLI, as well as in samples collected from animals subjected to hindlimb ischemia. Additionally, taking into account numerous experimental and clinical data demonstrating rather insufficient therapeutic potential of VEGF, we called into question the role of this protein in limb ischemia and vessel regeneration. In this review we are also summarizing several aspects which can influence VEGF expression and its measurement in the ischemic tissues.
Collapse
Affiliation(s)
- Agnieszka Jazwa
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Urszula Florczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
123
|
Hulse RP, Drake RAR, Bates DO, Donaldson LF. The control of alternative splicing by SRSF1 in myelinated afferents contributes to the development of neuropathic pain. Neurobiol Dis 2016; 96:186-200. [PMID: 27616424 PMCID: PMC5113660 DOI: 10.1016/j.nbd.2016.09.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023] Open
Abstract
Neuropathic pain results from neuroplasticity in nociceptive neuronal networks. Here we demonstrate that control of alternative pre-mRNA splicing, through the splice factor serine-arginine splice factor 1 (SRSF1), is integral to the processing of nociceptive information in the spinal cord. Neuropathic pain develops following a partial saphenous nerve ligation injury, at which time SRSF1 is activated in damaged myelinated primary afferent neurons, with minimal found in small diameter (IB4 positive) dorsal root ganglia neurons. Serine arginine protein kinase 1 (SRPK1) is the principal route of SRSF1 activation. Spinal SRPK1 inhibition attenuated SRSF1 activity, abolished neuropathic pain behaviors and suppressed central sensitization. SRSF1 was principally expressed in large diameter myelinated (NF200-rich) dorsal root ganglia sensory neurons and their excitatory central terminals (vGLUT1+ve) within the dorsal horn of the lumbar spinal cord. Expression of pro-nociceptive VEGF-Axxxa within the spinal cord was increased after nerve injury, and this was prevented by SRPK1 inhibition. Additionally, expression of anti-nociceptive VEGF-Axxxb isoforms was elevated, and this was associated with reduced neuropathic pain behaviors. Inhibition of VEGF receptor-2 signaling in the spinal cord attenuated behavioral nociceptive responses to mechanical, heat and formalin stimuli, indicating that spinal VEGF receptor-2 activation has potent pro-nociceptive actions. Furthermore, intrathecal VEGF-A165a resulted in mechanical and heat hyperalgesia, whereas the sister inhibitory isoform VEGF-A165b resulted in anti-nociception. These results support a role for myelinated fiber pathways, and alternative pre-mRNA splicing of factors such as VEGF-A in the spinal processing of neuropathic pain. They also indicate that targeting pre-mRNA splicing at the spinal level could lead to a novel target for analgesic development.
Collapse
Affiliation(s)
- Richard P Hulse
- Cancer Biology, School of Medicine, University of Nottingham, Nottingham, NG7 7UH, United Kingdom; School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom.
| | - Robert A R Drake
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - David O Bates
- Cancer Biology, School of Medicine, University of Nottingham, Nottingham, NG7 7UH, United Kingdom; School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Lucy F Donaldson
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom; School of Life Sciences and Arthritis Research UK Pain Centre, University of Nottingham, Nottingham NG7 7UH, United Kingdom.
| |
Collapse
|
124
|
Andreucci E, Bianchini F, Biagioni A, Del Rosso M, Papucci L, Schiavone N, Magnelli L. Roles of different IRES-dependent FGF2 isoforms in the acquisition of the major aggressive features of human metastatic melanoma. J Mol Med (Berl) 2016; 95:97-108. [PMID: 27558498 DOI: 10.1007/s00109-016-1463-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 08/09/2016] [Accepted: 08/15/2016] [Indexed: 11/29/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is involved in many physiological and pathological processes. Fgf2 deregulation contributes to the acquisition of malignant features of melanoma and other cancers. FGF2 is an alternative translation product expressed as five isoforms, a low-molecular-weight (18 KDa) and four high-molecular-weight (22, 22.5, 24, 34 KDa) isoforms, with different subcellular distributions. An internal ribosomal entry site (IRES) in its mRNA controls the translation of all the isoforms with the exception for the cap-dependent 34 KDa. The 18-KDa isoform has been extensively studied, while very few is known about the roles of high molecular weight isoforms. FGF2 is known to promote melanoma development and progression. To disclose the differential contribution of FGF2 isoforms in melanoma, we forced the expression of IRES-dependent low-molecular-weight (LMW, 18 KDa) and high-molecular-weight (HMW, 22, 22.5, 24 KDa) isoforms in a human metastatic melanoma cell line. This comparative study highlights that, while LMW isoform confers stem-like features to melanoma cells and promotes angiogenesis, HMW isoforms induce higher migratory ability and contribute to tumor perfusion by promoting vasculogenic mimicry (VM) when endothelial cell-driven angiogenesis is lacking. To conclude, FGF2 isoforms mainly behave in specific, antithetical manners, but can cooperate in different steps of tumor progression, providing melanoma cells with major malignant features. KEY MESSAGE FGF2 is an alternative translation product expressed as different isoforms termed LMW and HMW. FGF2 is involved in melanoma development and progression. HMW FGF2 isoforms enhance in vitro motility of melanoma cells. LMW FGF2 confers stem-like features and increases in vivo metastasization. LMW FGF2 promotes angiogenesis while HMW FGF2 induces vasculogenic mimicry.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| | - Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| |
Collapse
|
125
|
Lange C, Storkebaum E, de Almodóvar CR, Dewerchin M, Carmeliet P. Vascular endothelial growth factor: a neurovascular target in neurological diseases. Nat Rev Neurol 2016; 12:439-54. [PMID: 27364743 DOI: 10.1038/nrneurol.2016.88] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain function critically relies on blood vessels to supply oxygen and nutrients, to establish a barrier for neurotoxic substances, and to clear waste products. The archetypal vascular endothelial growth factor, VEGF, arose in evolution as a signal affecting neural cells, but was later co-opted by blood vessels to regulate vascular function. Consequently, VEGF represents an attractive target to modulate brain function at the neurovascular interface. On the one hand, VEGF is neuroprotective, through direct effects on neural cells and their progenitors and indirect effects on brain perfusion. In accordance, preclinical studies show beneficial effects of VEGF administration in neurodegenerative diseases, peripheral neuropathies and epilepsy. On the other hand, pathologically elevated VEGF levels enhance vessel permeability and leakage, and disrupt blood-brain barrier integrity, as in demyelinating diseases, for which blockade of VEGF may be beneficial. Here, we summarize current knowledge on the role and therapeutic potential of VEGF in neurological diseases.
Collapse
Affiliation(s)
- Christian Lange
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| | - Erik Storkebaum
- Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149 Muenster, Germany.,Faculty of Medicine, University of Muenster, Roentgenstrasse 20, D-48149 Muenster, Germany
| | | | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| |
Collapse
|
126
|
Abstract
Vascular endothelial growth factor (VEGF) plays a fundamental role in angiogenesis and endothelial cell biology, and has been the subject of intense study as a result. VEGF acts via a diverse and complex range of signaling pathways, with new targets constantly being discovered. This review attempts to summarize the current state of knowledge regarding VEGF cell signaling in endothelial and cardiovascular biology, with a particular emphasis on its role in angiogenesis.
Collapse
Affiliation(s)
- Ian Evans
- Centre for Cardiovascular Biology and Medicine, Division of Medicine, University College London, Rayne Building, 5 University Street, London, WC1E 6JF, UK,
| |
Collapse
|
127
|
Kamarulzaman EE, Vanderesse R, Gazzali AM, Barberi-Heyob M, Boura C, Frochot C, Shawkataly O, Aubry A, Wahab HA. Molecular modelling, synthesis and biological evaluation of peptide inhibitors as anti-angiogenic agent targeting neuropilin-1 for anticancer application. J Biomol Struct Dyn 2016; 35:26-45. [PMID: 26766582 DOI: 10.1080/07391102.2015.1131196] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor (VEGF) and its co-receptor neuropilin-1 (NRP-1) are important targets of many pro-angiogenic factors. In this study, nine peptides were synthesized and evaluated for their molecular interaction with NRP-1 and compared to our previous peptide ATWLPPR. Docking study showed that the investigated peptides shared the same binding region as shown by tuftsin known to bind selectively to NRP-1. Four pentapeptides (DKPPR, DKPRR, TKPPR and TKPRR) and a hexapeptide CDKPRR demonstrated good inhibitory activity against NRP-1. In contrast, peptides having arginine residue at sites other than the C-terminus exhibited low activity towards NRP-1 and this is confirmed by their inability to displace the VEGF165 binding to NRP-1. Docking study also revealed that replacement of carboxyl to amide group at the C-terminal arginine of the peptide did not affect significantly the binding interaction to NRP-1. However, the molecular affinity study showed that these peptides have marked reduction in the activity against NRP-1. Pentapeptides having C-terminal arginine showed strong interaction and good inhibitory activity with NRP thus may be a good template for anti-angiogenic targeting agent.
Collapse
Affiliation(s)
- Ezatul E Kamarulzaman
- a School of Pharmaceutical Sciences , Universiti Sains Malaysia , 11800 Penang , Malaysia.,b LCPM, UMR-CNRS 7375, Université de Lorraine, ENSIC , 1 Rue Grandville, F-54000 Nancy , France
| | - Régis Vanderesse
- b LCPM, UMR-CNRS 7375, Université de Lorraine, ENSIC , 1 Rue Grandville, F-54000 Nancy , France
| | - Amirah M Gazzali
- b LCPM, UMR-CNRS 7375, Université de Lorraine, ENSIC , 1 Rue Grandville, F-54000 Nancy , France
| | - Muriel Barberi-Heyob
- c CRAN, UMR-CNRS 7039 , Campus Science, BP 70239, F-54506 Vandœuvre-lès-Nancy , France
| | - Cédric Boura
- c CRAN, UMR-CNRS 7039 , Campus Science, BP 70239, F-54506 Vandœuvre-lès-Nancy , France
| | - Céline Frochot
- d LRGP , UMR-CNRS 7274, Université de Lorraine, ENSIC , 1 Rue Grandville, F-54000 Nancy , France
| | - Omar Shawkataly
- e Chemical Sciences Programme , School of Distance Education, Universiti Sains Malaysia , 11800 Penang , Malaysia
| | - André Aubry
- b LCPM, UMR-CNRS 7375, Université de Lorraine, ENSIC , 1 Rue Grandville, F-54000 Nancy , France
| | - Habibah A Wahab
- a School of Pharmaceutical Sciences , Universiti Sains Malaysia , 11800 Penang , Malaysia.,f Malaysian Institute of Pharmaceuticals and Nutraceuticals, Ministry of Science, Technology and Innovation , Jalan Bukit Gambir, 11800 Penang , Malaysia
| |
Collapse
|
128
|
Wang H. Anti-VEGF therapy in the management of retinopathy of prematurity: what we learn from representative animal models of oxygen-induced retinopathy. Eye Brain 2016; 8:81-90. [PMID: 28539803 PMCID: PMC5398744 DOI: 10.2147/eb.s94449] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Retinopathy of prematurity (ROP) remains a leading cause of childhood blindness, affecting infants born prematurely. ROP is characterized by the onset of delayed physiological retinal vascular development (PRVD) and followed by pathologic neovascularization into the vitreous instead of the retina, called intravitreal neovascularization (IVNV). Therefore, the therapeutic strategy for treating ROP is to promote PRVD and inhibit or prevent IVNV. Vascular endothelial growth factor (VEGF) plays an important role in the pathogenesis of ROP. There is a growing body of studies testing the use of anti-VEGF agents as a treatment for ROP. Intravitreal anti-VEGF treatment for ROP has potential advantages compared with laser photocoagulation, the gold standard for the treatment of severe ROP; however, intravitreal anti-VEGF treatment has been associated with reactivation of ROP and suppression of systemic VEGF that may affect body growth and organ development in preterm infants. Therefore, it is important to understand the role of VEGF in PRVD and IVNV. This review includes the current knowledge of anti-VEGF treatment for ROP from animal models of oxygen-induced retinopathy (OIR), highlighting the importance of VEGF inhibition by targeting retinal Müller cells, which inhibits IVNV and permits PRVD. The signaling events involved in mediating VEGF expression and promoting VEGF-mediated angiogenesis, including hypoxia-dependent signaling, erythropoietin/erythropoietin receptor-, oxidative stress-, beta-adrenergic receptor-, integrin-, Notch/Delta-like ligand 4- and exon guidance molecules-mediated signaling pathways, are also discussed.
Collapse
Affiliation(s)
- Haibo Wang
- Department of Ophthalmology, John A Moran Eye Center, The University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
129
|
NAGASAKI MASAHIRO, KONDO SEIJI, MUKUDAI YOSHIKI, KAMATANI TAKAAKI, AKIZUKI AYAKO, YASO ATSUSHI, SHIMANE TOSHIKAZU, SHIROTA TATSUO. Clinicopathological implications of vascular endothelial growth factor 165b expression in oral squamous cell carcinoma stroma. Oncol Rep 2016; 36:573-81. [DOI: 10.3892/or.2016.4826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/05/2016] [Indexed: 11/05/2022] Open
|
130
|
Schuster C, Akslen LA, Straume O. Expression of Heat Shock Protein 27 in Melanoma Metastases Is Associated with Overall Response to Bevacizumab Monotherapy: Analyses of Predictive Markers in a Clinical Phase II Study. PLoS One 2016; 11:e0155242. [PMID: 27166673 PMCID: PMC4864228 DOI: 10.1371/journal.pone.0155242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/25/2016] [Indexed: 11/27/2022] Open
Abstract
The aim of this study was to identify potential predictive biomarkers in 35 patients with metastatic melanoma treated with anti-angiogenic bevacizumab monotherapy in a clinical phase II study. The immunohistochemical expression of various angiogenic factors in tissues from primary melanomas and metastases as well as their concentration in blood samples were examined. Strong expression of Heat Shock Protein 27 (HSP27) in metastases correlated significantly with complete or partial response to bevacizumab (p = 0.044). Furthermore, clinical benefit, i.e., complete or partial response or stable disease for at least 6 months, was more frequent in patients with strong expression of HSP27 in primary tumors (p = 0.046). Tissue expression of vascular endothelial growth factor (VEGF-A), its splicing variant VEGF165b or basic fibroblast growth factor (bFGF) did not correlate with response, and the concentration of HSP27, VEGF-A or bFGF measured in blood samples before treatment did not show predictive value. Further, microvessel density, proliferating microvessel density and presence of glomeruloid microvascular proliferations were assessed in sections of primary tumors and metastases. Microvessel density in primary melanomas was significantly higher in patients with clinical benefit than in non-responders (p = 0.042). In conclusion, our findings suggest that strong HSP27 expression in melanoma metastases predicts response to bevacizumab treatment.
Collapse
Affiliation(s)
- Cornelia Schuster
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- * E-mail: (OS); (LAA)
| | - Oddbjørn Straume
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- * E-mail: (OS); (LAA)
| |
Collapse
|
131
|
Zhao M, Xie WK, Bai YJ, Huang LZ, Wang B, Liang JH, Yin H, Li XX, Shi X. Expression of Total Vascular Endothelial Growth Factor and the Anti-angiogenic VEGF 165 b Isoform in the Vitreous of Patients with Retinopathy of Prematurity. Chin Med J (Engl) 2016; 128:2505-9. [PMID: 26365970 PMCID: PMC4725572 DOI: 10.4103/0366-6999.164937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background: This study was to examine the expression of total vascular endothelial growth factor (VEGF) and the anti-angiogenic VEGF165b isoform in the vitreous body of retinopathy of prematurity (ROP) patients, and to further study the role of the VEGF splicing in the development of ROP. Methods: This was a prospective clinical laboratory investigation study. All patients enrolled received standard ophthalmic examination with stage 4 ROP that required vitrectomy to collect the vitreous samples. The control samples were from congenital cataract patients. The expression of total VEGF and the anti-angiogenic VEGF165b were measured by enzyme-linked immunosorbent assay. Results were analyzed statistically using nonparametric tests. Results: The total VEGF level was markedly elevated in ROP samples while VEGF165b was markedly decreased compared to control group. The relative protein expression level of VEGF165b isoform was significantly decreased in ROP patients which were correlated with the ischemia-induced neovascularization. Conclusions: There was a switch of VEGF splicing from anti-angiogenic to pro-angiogenic family in ROP patients. A specific inhibitor that more selectively targets VEGF165and controls the VEGF splicing between pro- and anti-angiogenic families might be a more effective therapy for ROP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xuan Shi
- Department of Ophthalmology, Peking University People's Hospital, Beijing 100044; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100044; Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
| |
Collapse
|
132
|
VEGF121 and VEGF165 differentially promote vessel maturation and tumor growth in mice and humans. Cancer Gene Ther 2016; 23:125-32. [DOI: 10.1038/cgt.2016.12] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/07/2016] [Accepted: 03/09/2016] [Indexed: 12/31/2022]
|
133
|
Raimondi C, Brash JT, Fantin A, Ruhrberg C. NRP1 function and targeting in neurovascular development and eye disease. Prog Retin Eye Res 2016; 52:64-83. [PMID: 26923176 PMCID: PMC4854174 DOI: 10.1016/j.preteyeres.2016.02.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/19/2022]
Abstract
Neuropilin 1 (NRP1) is expressed by neurons, blood vessels, immune cells and many other cell types in the mammalian body and binds a range of structurally and functionally diverse extracellular ligands to modulate organ development and function. In recent years, several types of mouse knockout models have been developed that have provided useful tools for experimental investigation of NRP1 function, and a multitude of therapeutics targeting NRP1 have been designed, mostly with the view to explore them for cancer treatment. This review provides a general overview of current knowledge of the signalling pathways that are modulated by NRP1, with particular focus on neuronal and vascular roles in the brain and retina. This review will also discuss the potential of NRP1 inhibitors for the treatment for neovascular eye diseases.
Collapse
Affiliation(s)
- Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
134
|
Sato A, Yoshihisa A, Yokokawa T, Shimizu T, Nakamura Y, Misaka T, Kamioka M, Kaneshiro T, Suzuki S, Kunii H, Saitoh SI, Takeishi Y. The association between circulating anti-angiogenic isoform of vascular endothelial growth factor and clinical profiles in patients with peripheral artery disease. Int J Cardiol 2016; 207:368-9. [PMID: 26820370 DOI: 10.1016/j.ijcard.2016.01.189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 10/22/2022]
Affiliation(s)
- Akihiko Sato
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Akiomi Yoshihisa
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan.
| | - Tetsuro Yokokawa
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Takeshi Shimizu
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Yuichi Nakamura
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Tomofumi Misaka
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Masashi Kamioka
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Takashi Kaneshiro
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Satoshi Suzuki
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Hiroyuki Kunii
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Shu-ichi Saitoh
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
135
|
Amadio M, Govoni S, Pascale A. Targeting VEGF in eye neovascularization: What's new? Pharmacol Res 2016; 103:253-69. [DOI: 10.1016/j.phrs.2015.11.027] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/19/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
|
136
|
Abstract
The rabbit corneal micropocket angiogenesis assay uses the avascular cornea as a substrate canvas to study angiogenesis in vivo. Through the use of standardized slow-release pellets, a predictable angiogenic response is generated over the course of 1-2 weeks and then quantified. Uniform slow-release pellets are prepared by mixing purified angiogenic growth factors such as basic fibroblast growth factor or vascular endothelial growth factor and a synthetic polymer to allow slow release. A micropocket is surgically created in the rabbit cornea under anesthesia and a pellet implanted. On the days later, the angiogenic response is measured and qualified using a slit lamp, as well as the concomitant vascular phenotype or inflammatory features. The results of the assay are used to assess the ability of potential therapeutic molecules to modulate angiogenesis in vivo, both when released locally or given by ocular formulations or through systemic treatment. In this chapter, the experimental details of the rabbit cornea assay and technical implementations to the original protocol are described.
Collapse
Affiliation(s)
- Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Marina Ziche
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| |
Collapse
|
137
|
Lim DK, Wylie RG, Langer R, Kohane DS. Selective binding of C-6 OH sulfated hyaluronic acid to the angiogenic isoform of VEGF(165). Biomaterials 2016; 77:130-138. [PMID: 26588795 PMCID: PMC4735037 DOI: 10.1016/j.biomaterials.2015.10.074] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/26/2015] [Accepted: 10/29/2015] [Indexed: 01/17/2023]
Abstract
Vascular endothelial growth factor 165 (VEGF165) is an important extracellular protein involved in pathological angiogenesis in diseases such as cancer, wet age-related macular degeneration (wet-AMD) and retinitis pigmentosa. VEGF165 exists in two different isoforms: the angiogenic VEGF165a, and the anti-angiogenic VEGF165b. In some angiogenic diseases the proportion of VEGF165b may be equal to or higher than that of VEGF165a. Therefore, developing therapeutics that inhibit VEGF165a and not VEGF165b may result in greater anti-angiogenic activity and therapeutic benefit. To this end, we report the selective binding properties of sulfated hyaluronic acid (s-HA). Selective biopolymers offer several advantages over antibodies or aptamers including cost effective and simple synthesis, and the ability to make nanoparticles or hydrogels for drug delivery applications or VEGF165a sequestration. Limiting sulfation to the C-6 hydroxyl (C-6 OH) in the N-acetyl-glucosamine repeat unit of hyaluronic acid (HA) resulted in a polymer with strong affinity for VEGF165a but not VEGF165b. Increased sulfation beyond the C-6 OH (i.e. greater than 1 sulfate group per HA repeat unit) resulted in s-HA polymers that bound both VEGF165a and VEGF165b. The C-6 OH sulfated HA (Mw 150 kDa) showed strong binding properties to VEGF165a with a fast association rate constant (Ka; 2.8 × 10(6) M(-1) s(-1)), slow dissociation rate constant (Kd; 2.8 × 10(-3) s(-1)) and strong equilibrium binding constant (KD; ∼1.0 nM)), which is comparable to the non-selective VEGF165 binding properties of the commercialized therapeutic anti-VEGF antibody (Avastin(®)). The C-6 OH sulfated HA also inhibited human umbilical vein endothelial cell (HUVEC) survival and proliferation and human dermal microvascular endothelial cell (HMVEC) tube formation. These results demonstrate that the semi-synthetic natural polymer, C-6 OH sulfated HA, may be a promising biomaterial for the treatment of angiogenesis-related disease.
Collapse
Affiliation(s)
- Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk gu, Seoul, South Korea
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Ryan G. Wylie
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, Ontario, L8S 4M1, Canada
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Robert Langer
- David H. Koch Institutes for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
138
|
Sell-Kubiak E, Duijvesteijn N, Lopes MS, Janss LLG, Knol EF, Bijma P, Mulder HA. Genome-wide association study reveals novel loci for litter size and its variability in a Large White pig population. BMC Genomics 2015; 16:1049. [PMID: 26652161 PMCID: PMC4674943 DOI: 10.1186/s12864-015-2273-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/03/2015] [Indexed: 01/11/2023] Open
Abstract
Background In many traits, not only individual trait levels are under genetic control, but also the variation around that level. In other words, genotypes do not only differ in mean, but also in (residual) variation around the genotypic mean. New statistical methods facilitate gaining knowledge on the genetic architecture of complex traits such as phenotypic variability. Here we study litter size (total number born) and its variation in a Large White pig population using a Double Hierarchical Generalized Linear model, and perform a genome-wide association study using a Bayesian method. Results In total, 10 significant single nucleotide polymorphisms (SNPs) were detected for total number born (TNB) and 9 SNPs for variability of TNB (varTNB). Those SNPs explained 0.83 % of genetic variance in TNB and 1.44 % in varTNB. The most significant SNP for TNB was detected on Sus scrofa chromosome (SSC) 11. A possible candidate gene for TNB is ENOX1, which is involved in cell growth and survival. On SSC7, two possible candidate genes for varTNB are located. The first gene is coding a swine heat shock protein 90 (HSPCB = Hsp90), which is a well-studied gene stabilizing morphological traits in Drosophila and Arabidopsis. The second gene is VEGFA, which is activated in angiogenesis and vasculogenesis in the fetus. Furthermore, the genetic correlation between additive genetic effects on TNB and on its variation was 0.49. This indicates that the current selection to increase TNB will also increase the varTNB. Conclusions To the best of our knowledge, this is the first study reporting SNPs associated with variation of a trait in pigs. Detected genomic regions associated with varTNB can be used in genomic selection to decrease varTNB, which is highly desirable to avoid very small or very large litters in pigs. However, the percentage of variance explained by those regions was small. The SNPs detected in this study can be used as indication for regions in the Sus scrofa genome involved in maintaining low variability of litter size, but further studies are needed to identify the causative loci.
Collapse
Affiliation(s)
- E Sell-Kubiak
- Animal Breeding and Genomics Center, Wageningen University, P.O. Box 338, 6700, Wageningen, AH, The Netherlands.
| | - N Duijvesteijn
- Topigs Norsvin Research Center B.V, P.O. Box 43, 6640, Beuningen, AA, The Netherlands.
| | - M S Lopes
- Topigs Norsvin Research Center B.V, P.O. Box 43, 6640, Beuningen, AA, The Netherlands.
| | - L L G Janss
- Department of Molecular Biology and Genetics, Aarhus University, P.O. Box 50, 8830, Tjele, Denmark.
| | - E F Knol
- Topigs Norsvin Research Center B.V, P.O. Box 43, 6640, Beuningen, AA, The Netherlands.
| | - P Bijma
- Animal Breeding and Genomics Center, Wageningen University, P.O. Box 338, 6700, Wageningen, AH, The Netherlands.
| | - H A Mulder
- Animal Breeding and Genomics Center, Wageningen University, P.O. Box 338, 6700, Wageningen, AH, The Netherlands.
| |
Collapse
|
139
|
Sargent KM, Clopton DT, Lu N, Pohlmeier WE, Cupp AS. VEGFA splicing: divergent isoforms regulate spermatogonial stem cell maintenance. Cell Tissue Res 2015; 363:31-45. [PMID: 26553653 DOI: 10.1007/s00441-015-2297-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/24/2015] [Indexed: 12/22/2022]
Abstract
Despite being well-known for regulating angiogenesis in both normal and tumorigenic environments, vascular endothelial growth factor A (VEGFA) has been recently implicated in male fertility, namely in the maintenance of spermatogonial stem cells (SSC). The VEGFA gene can be spliced into multiple distinct isoforms that are either angiogenic or antiangiogenic in nature. Although studies have demonstrated the alternative splicing of VEGFA, including the divergent roles of the two isoform family types, many investigations do not differentiate between them. Data concerning VEGFA in the mammalian testis are limited, but the various angiogenic isoforms appear to promote seminiferous cord formation and to form a gradient across which cells may migrate. Treatment with either antiangiogenic isoforms of VEGFA or with inhibitors to angiogenic signaling impair these processes. Serendipitously, expression of KDR, the primary receptor for both types of VEGFA isoforms, was observed on male germ cells. These findings led to further investigation of the way that VEGFA elicits avascular functions within testes. Following treatment of donor perinatal male mice with either antiangiogenic VEGFA165b or angiogenic VEGFA164 isoforms, seminiferous tubules were less colonized following transplantation with cells from VEGFA165b-treated donors. Thus, VEGFA165b and possibly other antiangiogenic isoforms of VEGFA reduce SSC number either by promoting premature differentiation, inducing cell death, or by preventing SSC formation. Thus, angiogenic isoforms of VEGFA are hypothesized to promote SSC self-renewal, and the divergent isoforms are thought to balance one another to maintain SSC homeostasis in vivo.
Collapse
Affiliation(s)
- Kevin M Sargent
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - Debra T Clopton
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - Ningxia Lu
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - William E Pohlmeier
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, A224i Animal Science Building, 3940 Fair Street, Lincoln, NE 68583-0908, USA.
| |
Collapse
|
140
|
Sargent KM, McFee RM, Spuri Gomes R, Cupp AS. Vascular endothelial growth factor A: just one of multiple mechanisms for sex-specific vascular development within the testis? J Endocrinol 2015; 227:R31-50. [PMID: 26562337 PMCID: PMC4646736 DOI: 10.1530/joe-15-0342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/16/2015] [Indexed: 01/25/2023]
Abstract
Testis development from an indifferent gonad is a critical step in embryogenesis. A hallmark of testis differentiation is sex-specific vascularization that occurs as endothelial cells migrate from the adjacent mesonephros into the testis to surround Sertoli-germ cell aggregates and induce seminiferous cord formation. Many in vitro experiments have demonstrated that vascular endothelial growth factor A (VEGFA) is a critical regulator of this process. Both inhibitors to VEGFA signal transduction and excess VEGFA isoforms in testis organ cultures impaired vascular development and seminiferous cord formation. However, in vivo models using mice which selectively eliminated all VEGFA isoforms: in Sertoli and germ cells (pDmrt1-Cre;Vegfa(-/-)); Sertoli and Leydig cells (Amhr2-Cre;Vegfa(-/-)) or Sertoli cells (Amh-Cre;Vegfa(-/-) and Sry-Cre;Vegfa(-/-)) displayed testes with observably normal cords and vasculature at postnatal day 0 and onwards. Embryonic testis development may be delayed in these mice; however, the postnatal data indicate that VEGFA isoforms secreted from Sertoli, Leydig or germ cells are not required for testis morphogenesis within the mouse. A Vegfa signal transduction array was employed on postnatal testes from Sry-Cre;Vegfa(-/-) versus controls. Ptgs1 (Cox1) was the only upregulated gene (fivefold). COX1 stimulates angiogenesis and upregulates, VEGFA, Prostaglandin E2 (PGE2) and PGD2. Thus, other gene pathways may compensate for VEGFA loss, similar to multiple independent mechanisms to maintain SOX9 expression. Multiple independent mechanism that induce vascular development in the testis may contribute to and safeguard the sex-specific vasculature development responsible for inducing seminiferous cord formation, thus ensuring appropriate testis morphogenesis in the male.
Collapse
Affiliation(s)
- Kevin M Sargent
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| | - Renee M McFee
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| | - Renata Spuri Gomes
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| | - Andrea S Cupp
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| |
Collapse
|
141
|
Bullock N, Potts J, Simpkin AJ, Koupparis A, Harper SJ, Oxley J, Oltean S. Serine-arginine protein kinase 1 (SRPK1), a determinant of angiogenesis, is upregulated in prostate cancer and correlates with disease stage and invasion. J Clin Pathol 2015; 69:171-5. [PMID: 26500332 PMCID: PMC4752636 DOI: 10.1136/jclinpath-2015-203125] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/16/2015] [Indexed: 01/21/2023]
Abstract
Vascular endothelial growth factor (VEGF) undergoes alternative splicing to produce both proangiogenic and antiangiogenic isoforms. Preferential splicing of proangiogenic VEGF is determined by serine-arginine protein kinase 1 (SRPK1), which is upregulated in a number of cancers. In the present study, we aimed to investigate SRPK1 expression in prostate cancer (PCa) and its association with cancer progression. SRPK1 expression was assessed using immunohistochemistry of PCa tissue extracted from radical prostatectomy specimens of 110 patients. SRPK1 expression was significantly higher in tumour compared with benign tissue (p<0.00001) and correlated with higher pT stage (p=0.004), extracapsular extension (p=0.003) and extracapsular perineural invasion (p=0.008). Interestingly, the expression did not correlate with Gleason grade (p=0.21), suggesting that SRPK1 facilitates the development of a tumour microenvironment that favours growth and invasion (possibly through stimulating angiogenesis) while having little bearing on the morphology or function of the tumour cells themselves.
Collapse
Affiliation(s)
- Nicholas Bullock
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | | | - Andrew J Simpkin
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | | - Steve J Harper
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - Jon Oxley
- Department of Cellular Pathology, Southmead Hospital, Bristol, UK
| | - Sebastian Oltean
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| |
Collapse
|
142
|
Vascular endothelial growth factor-A165b prevents diabetic neuropathic pain and sensory neuronal degeneration. Clin Sci (Lond) 2015. [PMID: 26201024 DOI: 10.1042/cs20150124] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetic peripheral neuropathy affects up to half of diabetic patients. This neuronal damage leads to sensory disturbances, including allodynia and hyperalgesia. Many growth factors have been suggested as useful treatments for prevention of neurodegeneration, including the vascular endothelial growth factor (VEGF) family. VEGF-A is generated as two alternative splice variant families. The most widely studied isoform, VEGF-A165a is both pro-angiogenic and neuroprotective, but pro-nociceptive and increases vascular permeability in animal models. Streptozotocin (STZ)-induced diabetic rats develop both hyperglycaemia and many of the resulting diabetic complications seen in patients, including peripheral neuropathy. In the present study, we show that the anti-angiogenic VEGF-A splice variant, VEGF-A165b, is also a potential therapeutic for diabetic neuropathy. Seven weeks of VEGF-A165b treatment in diabetic rats reversed enhanced pain behaviour in multiple behavioural paradigms and was neuroprotective, reducing hyperglycaemia-induced activated caspase 3 (AC3) levels in sensory neuronal subsets, epidermal sensory nerve fibre loss and aberrant sciatic nerve morphology. Furthermore, VEGF-A165b inhibited a STZ-induced increase in Evans Blue extravasation in dorsal root ganglia (DRG), saphenous nerve and plantar skin of the hind paw. Increased transient receptor potential ankyrin 1 (TRPA1) channel activity is associated with the onset of diabetic neuropathy. VEGF-A165b also prevented hyperglycaemia-enhanced TRPA1 activity in an in vitro sensory neuronal cell line indicating a novel direct neuronal mechanism that could underlie the anti-nociceptive effect observed in vivo. These results demonstrate that in a model of Type I diabetes VEGF-A165b attenuates altered pain behaviour and prevents neuronal stress, possibly through an effect on TRPA1 activity.
Collapse
|
143
|
Rhee SH, Ma EL, Lee Y, Taché Y, Pothoulakis C, Im E. Corticotropin Releasing Hormone and Urocortin 3 Stimulate Vascular Endothelial Growth Factor Expression through the cAMP/CREB Pathway. J Biol Chem 2015; 290:26194-203. [PMID: 26350463 DOI: 10.1074/jbc.m115.678979] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Indexed: 12/15/2022] Open
Abstract
Colonic epithelium is the first line of defense against various pathological offenses in the gut. Previous studies have shown that the peptides of the corticotropin-releasing hormone (CRH) family modulate vascular endothelial growth factor (VEGF)-A production in other cells. Here we sought to investigate whether CRH and urocortin (Ucn) 3 regulate VEGF-A secretion in colonocytes through CRH receptors and to elucidate the underlying mechanism of action. CRH and Ucn 3 significantly increased the expression levels of VEGF-A mRNA and protein through CRH receptor 1 and 2, respectively, in human colonic epithelial cells and primary mouse intestinal epithelial cells. Underlying mechanisms involve activation of adenylyl cyclase with subsequent increase of intracellular cAMP level and increased DNA binding activity of transcription factor CREB on VEGF-A promoter region. Finally, genetic deficiency of CREB decreased intestinal inflammation and VEGF-A expression in a dextran sodium sulfate-induced colitis model. These results show that activation of CRH receptors by CRH ligands stimulates VEGF-A expression in intestinal epithelial cells through the cAMP/CREB pathway. Since VEGF-A boosts inflammatory responses through angiogenesis, these data suggest that CREB may be a key effector of CRH and Ucn 3-dependent inflammatory angiogenesis.
Collapse
Affiliation(s)
- Sang Hoon Rhee
- From the Section of Inflammatory Bowel Disease & Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California 90095 and
| | - Elise L Ma
- From the Section of Inflammatory Bowel Disease & Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California 90095 and
| | - Yunna Lee
- College of Pharmacy, Pusan National University, Busan, 609-735, South Korea
| | - Yvette Taché
- From the Section of Inflammatory Bowel Disease & Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California 90095 and
| | - Charalabos Pothoulakis
- From the Section of Inflammatory Bowel Disease & Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California 90095 and
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan, 609-735, South Korea
| |
Collapse
|
144
|
Bhaskari J, Premalata CS, Shilpa V, Rahul B, Pallavi VR, Ramesh G, Krishnamoorthy L. Vascular endothelial growth factor polymorphisms and a synchronized examination of plasma and tissue expression in epithelial ovarian cancers. Tumour Biol 2015; 37:1017-23. [PMID: 26264619 DOI: 10.1007/s13277-015-3891-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/04/2015] [Indexed: 11/26/2022] Open
Abstract
In this study, we have analyzed six genetic polymorphisms of the VEGF-A gene and correlated the genetic data with plasma and tissue expression of VEGF-A in epithelial ovarian carcinomas. A total of 130 cases including 95 malignant carcinomas, 17 low malignant potential and 18 benign tumours were studied. rs699947, rs833061, rs1570360, rs2010963, rs1413711 and rs3025039 were studied by polymerase chain reaction and restriction fragment length polymorphism (PCR-RFLP). Plasma levels of VEGF-A were estimated by enzyme-linked immunosorbent assay (ELISA) and tissue expression of VEGF-A by immunohistochemistry (IHC). Four polymorphisms of the above excluding rs699947 and rs3025039 showed significant association with malignancy, and we observed the presence of positive correlation between haplotype CCGGCC and increased expression of VEGF-A in both plasma and tissues which also correlated with poor prognosis and recurrence suggesting a probable increase in resistance to treatment in such carriers. Highly upregulated tissue expression of VEGF-A was seen in all epithelial ovarian carcinomas with intensity of expression increasing from benign to malignant cases. ELISA data from our study showed an increase in circulating levels of VEGF-A in malignancies. VEGF-A plasma levels can be employed as a biomarker for high-grade malignancy in epithelial ovarian cancers alongside tissue expression and CA-125 levels. This study is unique due to the fact that a simultaneous analysis of plasma and tissue expression has been demonstrated and is a first such study in epithelial ovarian cancers and representing the Indian population (South-east Asian) synchronized with genetic polymorphism data as well.
Collapse
Affiliation(s)
- J Bhaskari
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - C S Premalata
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka, India
| | - V Shilpa
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - B Rahul
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - V R Pallavi
- Department of Gynec-Oncology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka, India
| | - G Ramesh
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - Lakshmi Krishnamoorthy
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India.
- Department of Biochemistry, Sri Shankara Cancer Hospital and Research Centre, Bangalore, 560004, India.
| |
Collapse
|
145
|
Janardhan B, Vaderhobli S, Bhagat R, Chennagiri Srinivasamurthy P, Venketeshiah Reddihalli P, Gawari R, Krishnamoorthy L. Investigating impact of Vascular Endothelial Growth Factor Polymorphisms in Epithelial Ovarian Cancers: A Study in the Indian Population. PLoS One 2015; 10:e0131190. [PMID: 26158818 PMCID: PMC4497663 DOI: 10.1371/journal.pone.0131190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/30/2015] [Indexed: 01/19/2023] Open
Abstract
Epithelial ovarian cancer is one of the increasingly incident malignancies that is notorious because of its evasiveness for early diagnosis and high mortality rates. Epithelial ovarian cancers are highly dependent on pathologic vasculature and Vascular Endothelial Growth Factor is known to be one of the most efficient angiogenic factors. Polymorphisms of the VEGF gene, in this study, were assessed for association with the malignancy and other clinico-pathological factors. 300 case samples and 320 age and mensus status matched controls were inculcated into the study. rs699947, rs833061, rs1570360, rs2010963, rs1413711 and rs3025039 were the six single nucleotide polymorphisms that were scrutinized. Genotyping was carried out by polymerase chain reaction and restriction fragment length polymorphism. rs 3025039 showed immense promise as a marker for disease aggression and recurrence and a factor for poor prognosis. rs699947 showed least association with the disease and clinico-pathologic factors studied. rs833061, rs 1570360 showed significant association with some clinico-pathological factors such as bilateral affliction of ovaries and post operative CA-125 levels. rs2010963 associated with presence of ascites in higher volumes. The SNPs under consideration showed no formidable linkage in our study samples. A haplotype analysis (excluding rs699947 and rs1413711) revealed 5 frontrunners being present in >85% of the population with TGGC and CGCC associating significantly as protective and risk factors respectively. These haplotypes showed a dose dependent additive effect of their seeming functionality. This study is unique and a first of its kind carried out in the Indian population of South-east Asia.
Collapse
Affiliation(s)
- Bhaskari Janardhan
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - Shilpa Vaderhobli
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - Rahul Bhagat
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | | | | | - Ramesh Gawari
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
| | - Lakshmi Krishnamoorthy
- Department of Biochemistry, Kidwai Memorial Institute of Oncology, Bangalore, 560029, India
- * E-mail:
| |
Collapse
|
146
|
Zhao YJ, Han HZ, Liang Y, Shi CZ, Zhu QC, Yang J. Alternative splicing of VEGFA, APP and NUMB genes in colorectal cancer. World J Gastroenterol 2015; 21:6550-60. [PMID: 26074693 PMCID: PMC4458765 DOI: 10.3748/wjg.v21.i21.6550] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 02/10/2015] [Accepted: 03/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate alternative splicing in vascular endothelial growth factor A (VEGFA), amyloid beta precursor protein (APP), and Numb homolog (NUMB) in colorectal cancer (CRC). METHODS Real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and PCR-restriction fragment length polymorphism analyses were performed to detect the expression of VEGFA, APP, and NUMB mRNA in 20 CRC tissues and matched adjacent normal tissues, as well as their alternative splicing variants. RESULTS qRT-PCR analysis revealed that the expression of APP, NUMB, and VEGFA165b mRNA were significantly downregulated, while VEGFA mRNA was upregulated, in CRC tissues (all P < 0.05). PCR-restriction fragment length polymorphism analysis revealed that the expression of VEGFA165a/b in CRC tissues was significantly higher than in adjacent normal tissues (P < 0.05). Compared with adjacent normal tissues, the expression of NUMB-PRR(S) in CRC tissues was significantly decreased (P < 0.05), and the expression of NUMB-PRR(L) was increased (P < 0.05). CONCLUSION Alternative splicing of VEGFA, APP, and NUMB may regulate the development of CRC, and represent new targets for its diagnosis, prognosis, and treatment.
Collapse
|
147
|
de Brot S, Ntekim A, Cardenas R, James V, Allegrucci C, Heery DM, Bates DO, Ødum N, Persson JL, Mongan NP. Regulation of vascular endothelial growth factor in prostate cancer. Endocr Relat Cancer 2015; 22:R107-23. [PMID: 25870249 DOI: 10.1530/erc-15-0123] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is the most common malignancy affecting men in the western world. Although radical prostatectomy and radiation therapy can successfully treat PCa in the majority of patients, up to ~30% will experience local recurrence or metastatic disease. Prostate carcinogenesis and progression is typically an androgen-dependent process. For this reason, therapies for recurrent PCa target androgen biosynthesis and androgen receptor function. Such androgen deprivation therapies (ADT) are effective initially, but the duration of response is typically ≤24 months. Although ADT and taxane-based chemotherapy have delivered survival benefits, metastatic PCa remains incurable. Therefore, it is essential to establish the cellular and molecular mechanisms that enable localized PCas to invade and disseminate. It has long been accepted that metastases require angiogenesis. In the present review, we examine the essential role for angiogenesis in PCa metastases, and we focus in particular on the current understanding of the regulation of vascular endothelial growth factor (VEGF) in localized and metastatic PCa. We highlight recent advances in understanding the role of VEGF in regulating the interaction of cancer cells with tumor-associated immune cells during the metastatic process of PCa. We summarize the established mechanisms of transcriptional and post-transcriptional regulation of VEGF in PCa cells and outline the molecular insights obtained from preclinical animal models of PCa. Finally, we summarize the current state of anti-angiogenesis therapies for PCa and consider how existing therapies impact VEGF signaling.
Collapse
Affiliation(s)
- Simone de Brot
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Atara Ntekim
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Ryan Cardenas
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Victoria James
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Cinzia Allegrucci
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - David M Heery
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - David O Bates
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Niels Ødum
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Jenny L Persson
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Nigel P Mongan
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| |
Collapse
|
148
|
Wilkie MD, Emmett MS, Santosh S, Lightbody KA, Lane S, Goodyear PW, Sheard JD, Boyd MT, Pritchard-Jones RO, Jones TM. Relative expression of vascular endothelial growth factor isoforms in squamous cell carcinoma of the head and neck. Head Neck 2015; 38:775-81. [DOI: 10.1002/hed.23959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/02/2014] [Accepted: 12/12/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Mark D. Wilkie
- Department of Molecular and Clinical Cancer Medicine; Liverpool Cancer Research Centre, University of Liverpool; Liverpool United Kingdom
- Department of Otorhinolaryngology - Head and Neck Surgery; University Hospital Aintree; Liverpool United Kingdom
| | - Maxine S. Emmett
- Department of Molecular and Clinical Cancer Medicine; Liverpool Cancer Research Centre, University of Liverpool; Liverpool United Kingdom
| | - Shilpa Santosh
- Department of Pathology; University Hospital Aintree; Liverpool United Kingdom
| | - Kathryn A. Lightbody
- Department of Otorhinolaryngology - Head and Neck Surgery; University Hospital Aintree; Liverpool United Kingdom
| | - Steven Lane
- Department of Biostatistics; University of Liverpool; Liverpool United Kingdom
| | - Paul W. Goodyear
- Department of Otorhinolaryngology - Head and Neck Surgery; University Hospital Aintree; Liverpool United Kingdom
| | - Jon D. Sheard
- Department of Pathology; University Hospital Aintree; Liverpool United Kingdom
| | - Mark T. Boyd
- Department of Molecular and Clinical Cancer Medicine; Liverpool Cancer Research Centre, University of Liverpool; Liverpool United Kingdom
| | - Rowan O. Pritchard-Jones
- Department of Molecular and Clinical Cancer Medicine; Liverpool Cancer Research Centre, University of Liverpool; Liverpool United Kingdom
| | - Terence M. Jones
- Department of Molecular and Clinical Cancer Medicine; Liverpool Cancer Research Centre, University of Liverpool; Liverpool United Kingdom
- Department of Otorhinolaryngology - Head and Neck Surgery; University Hospital Aintree; Liverpool United Kingdom
| |
Collapse
|
149
|
Li X, Gu F, Niu C, Wang Y, Liu Z, Li N, Pan B, He D, Kong J, Zhang S, Wang X, Yao Y, Zheng L. VEGF111b, a C-terminal splice variant of VEGF-A and induced by mitomycin C, inhibits ovarian cancer growth. J Transl Med 2015; 13:164. [PMID: 25990504 PMCID: PMC4480579 DOI: 10.1186/s12967-015-0522-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/06/2015] [Indexed: 11/10/2022] Open
Abstract
Background Alternative splicing of VEGF-A gives rise to two families – the pro-angiogenic VEGFxxx family and the anti-angiogenic VEGFxxxb family that differ by only six amino acids at their C-terminal end. The first verified and widely reported VEGFxxxb family member is VEGF165b, and here VEGF165b is a positive control. Methords VEGF111b mRNA was detected in ovarian cancer cell lines SKOV3 and OVCAR3 by RT-PCR. Western blot was used to detect VEGF111b and VEGF165b protein in the CMs and lysates of OVCAR3 cells. MTT and colony formation assay were used to detect the short-term and long-term proliferation inhibition ability of ovarian cancer cells with VEGF111b overexpression. Cell-cycle analysis was performed to further characterize VEGF111b inhibition effects. VEGF111b signaling on ovarian cancer cells were determined by western blot. The expression levels of Ki67, PCNA, CD31 and VEGF in VEGF111b overexpression xenograft model were detected by immunohistochemistry. Results Under the effect of mitomycin C, we identify a new member of VEGFxxxb family-VEGF111b in ovarian cancer cell lines. SKOV3 and OVCAR cells were transfected with empty lentivirus, VEGF111b or VEGF165b lentivirus. VEGF111b and VEGF165b overexpression inhibits proliferation of the ovarian cancer cells, but inhibition effect of VEGF111b is slightly less efficient than VEGF165b. Cell cycle analysis was further used to elucidate the mechanism involved in the inhibition effect. Further, we detected the expression of VEGF-R2 in SKOV3 and OVCAR3 cells, and shown that VEGF111b might bind to conventional VEGF-R2 with the results of reducing VEGF-R2 tyrosine phosphorylation and downstream signaling to have anti-tumor effects. In vivo VEGF111b overexpression inhibits ovarian cancer growth in xenograft mice. Conclusion Our results show that VEGF111b, as a new member of VEGFxxxb family, with similar properties to VEGF165b, plays potent anti-tumor effect in vitro and in vivo that can target the VEGF-R2 and its signaling pathway to inhibit ovarian tumor growth. This also opens a new avenue for treating ovarian cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0522-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiuli Li
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China.
| | - Fang Gu
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Beijing, China.
| | - Chenguang Niu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - Yuanfen Wang
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China.
| | - Zhongyu Liu
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China.
| | - Na Li
- Department of Obstetrics and Gynecology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Bing Pan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - Dan He
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - Jian Kong
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, China.
| | - Shaobo Zhang
- Department of General Surgery, PLA 180th hospital, Fujian, China.
| | - Xu Wang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - Yuanqing Yao
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
150
|
Mast cells and basophils in inflammatory and tumor angiogenesis and lymphangiogenesis. Eur J Pharmacol 2015; 778:146-51. [PMID: 25941082 DOI: 10.1016/j.ejphar.2015.03.088] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/20/2015] [Accepted: 03/25/2015] [Indexed: 01/08/2023]
Abstract
Angiogenesis, namely, the growth of new blood vessels from pre-existing ones, is an essential process of embryonic development and post-natal growth. In adult life, it may occur in physiological conditions (menstrual cycle and wound healing), during inflammatory disorders (autoimmune diseases and allergic disorders) and in tumor growth. The angiogenic process requires a tightly regulated interaction among different cell types (e.g. endothelial cells and pericytes), the extracellular matrix, several specific growth factors (e.g. VEGFs, Angiopoietins), cytokines and chemokines. Lymphangiogenesis, namely, the growth of new lymphatic vessels, is an important process in tumor development, in the formation of metastasis and in several inflammatory and metabolic disorders. In addition to tumors, several effector cells of inflammation (mast cells, macrophages, basophils, eosinophils, neutrophils, etc.) are important sources of a wide spectrum of angiogenic and lymphangiogenic factors. Human mast cells produce a large array of angiogenic and lymphangiogenic molecules. Primary human mast cells and two mast cell lines constitutively express several isoforms of angiogenic (VEGF-A and VEGF-B) and the two lymphangiogenic factors (VEGF-C and VEGF-D). In addition, human mast cells express the VEGF receptor 1 (VEGFR-1) and 2 (VEGFR-2), the co-receptors neuropilin-1 (NRP1) and -2 (NRP2) and the Tie1 and Tie2 receptors. Immunologically activated human basophils selectively produce VEGF-A and -B, but not VEGF-C and -D. They also release Angiopoietin1 that activates Tie2 on human mast cells. Collectively, these findings indicate that human mast cells and basophils might participate in the complex network involving inflammatory and tumor angiogenesis and lymphangiogenesis.
Collapse
|