Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Nat Commun ; 11(1): 445, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31974352

ABSTRACT

The number of patients diagnosed with chronic bile duct disease is increasing and in most cases these diseases result in chronic ductular scarring, necessitating liver transplantation. The formation of ductular scaring affects liver function; however, scar-generating portal fibroblasts also provide important instructive signals to promote the proliferation and differentiation of biliary epithelial cells. Therefore, understanding whether we can reduce scar formation while maintaining a pro-regenerative microenvironment will be essential in developing treatments for biliary disease. Here, we describe how regenerating biliary epithelial cells express Wnt-Planar Cell Polarity signalling components following bile duct injury and promote the formation of ductular scars by upregulating pro-fibrogenic cytokines and positively regulating collagen-deposition. Inhibiting the production of Wnt-ligands reduces the amount of scar formed around the bile duct, without reducing the development of the pro-regenerative microenvironment required for ductular regeneration, demonstrating that scarring and regeneration can be uncoupled in adult biliary disease and regeneration.


Subject(s)
Bile Duct Diseases/pathology , Cholangitis, Sclerosing/pathology , Cicatrix/pathology , Wnt Signaling Pathway , Animals , Axin Protein/genetics , Axin Protein/metabolism , Bile Duct Diseases/chemically induced , Bile Duct Diseases/metabolism , Bile Ducts/cytology , Cell Polarity , Cholangitis, Sclerosing/metabolism , Cicatrix/metabolism , Disease Models, Animal , Epithelial Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4/metabolism , Male , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pyridines/toxicity , Wnt Signaling Pathway/drug effects , Wnt-5a Protein/metabolism
2.
RNA Biol ; 16(12): 1672-1681, 2019 12.
Article in English | MEDLINE | ID: mdl-31432737

ABSTRACT

Vascular endothelial growth factor (VEGF)-A is differentially spliced to give two functionally different isoform families; pro-angiogenic, pro-permeability VEGF-Axxx and anti-angiogenic, anti-permeability VEGF-Axxxb. VEGF-A splicing is dysregulated in several pathologies, including cancer, diabetes, and peripheral arterial disease. The bichromatic VEGF-A splicing-sensitive fluorescent reporter harboured in a transgenic mouse is a novel approach to investigate the splicing patterns of VEGF-A in vivo. We generated a transgenic mouse harbouring a splicing-sensitive fluorescent reporter designed to mimic VEGF-A terminal exon splicing (VEGF8ab) by insertion into the ROSA26 genomic locus. dsRED expression denotes proximal splice site selection (VEGF-Axxx) and eGFP expression denotes distal splice site selection (VEGF-Axxxb). We investigated the tissue-specific expression patterns in the eye, skeletal muscle, cardiac muscle, kidney, and pancreas, and determined whether the splicing pattern could be manipulated in the same manner as endogenous VEGF-A by treatment with the SRPK1 inhibitor SPHINX 31. We confirmed expression of both dsRED and eGFP in the eye, skeletal muscle, cardiac muscle, kidney, and pancreas, with the highest expression of both fluorescent proteins observed in the exocrine pancreas. The ratio of dsRED and eGFP matched that of endogenous VEGF-Axxx and VEGF-Axxxb. Treatment of the VEGF8ab mice with SPHINX 31 increased the mRNA and protein eGFP/dsRED ratio in the exocrine pancreas, mimicking endogenous VEGF-A splicing. The VEGF-A exon 8 splicing-sensitive fluorescent reporter mouse is a novel tool to assess splicing regulation in the individual cell-types and tissues, which provides a useful screening process for potentially therapeutic splicing regulatory compounds in vivo.


Subject(s)
Alternative Splicing , Animals, Genetically Modified , Genes, Reporter , Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Exons , Eye/metabolism , Genetic Loci , Green Fluorescent Proteins/metabolism , Introns , Kidney/metabolism , Luminescent Proteins/metabolism , Mice , Muscle, Skeletal/metabolism , Myocardium/metabolism , Pancreas/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
J Physiol ; 596(16): 3675-3693, 2018 08.
Article in English | MEDLINE | ID: mdl-29774557

ABSTRACT

KEY POINTS: Diabetes is thought to induce neuropathic pain through activation of dorsal horn sensory neurons in the spinal cord. Here we explore the impact of hyperglycaemia on the blood supply supporting the spinal cord and chronic pain development. In streptozotocin-induced diabetic rats, neuropathic pain is accompanied by a decline in microvascular integrity in the dorsal horn. Hyperglycaemia-induced degeneration of the endothelium in the dorsal horn was associated with a loss in vascular endothelial growth factor (VEGF)-A165 b expression. VEGF-A165 b treatment prevented diabetic neuropathic pain and degeneration of the endothelium in the spinal cord. Using an endothelial-specific VEGFR2 knockout transgenic mouse model, the loss of endothelial VEGFR2 signalling led to a decline in vascular integrity in the dorsal horn and the development of hyperalgesia in VEGFR2 knockout mice. This highlights that vascular degeneration in the spinal cord could be a previously unidentified factor in the development of diabetic neuropathic pain. ABSTRACT: Abnormalities of neurovascular interactions within the CNS of diabetic patients is associated with the onset of many neurological disease states. However, to date, the link between the neurovascular network within the spinal cord and regulation of nociception has not been investigated despite neuropathic pain being common in diabetes. We hypothesised that hyperglycaemia-induced endothelial degeneration in the spinal cord, due to suppression of vascular endothelial growth factor (VEGF)-A/VEGFR2 signalling, induces diabetic neuropathic pain. Nociceptive pain behaviour was investigated in a chemically induced model of type 1 diabetes (streptozotocin induced, insulin supplemented; either vehicle or VEGF-A165 b treated) and an inducible endothelial knockdown of VEGFR2 (tamoxifen induced). Diabetic animals developed mechanical allodynia and heat hyperalgesia. This was associated with a reduction in the number of blood vessels and reduction in Evans blue extravasation in the lumbar spinal cord of diabetic animals versus age-matched controls. Endothelial markers occludin, CD31 and VE-cadherin were downregulated in the spinal cord of the diabetic group versus controls, and there was a concurrent reduction of VEGF-A165 b expression. In diabetic animals, VEGF-A165 b treatment (biweekly i.p., 20 ng g-1 ) restored normal Evans blue extravasation and prevented vascular degeneration, diabetes-induced central neuron activation and neuropathic pain. Inducible knockdown of VEGFR2 (tamoxifen treated Tie2CreERT2 -vegfr2flfl mice) led to a reduction in blood vessel network volume in the lumbar spinal cord and development of heat hyperalgesia. These findings indicate that hyperglycaemia leads to a reduction in the VEGF-A/VEGFR2 signalling cascade, resulting in endothelial dysfunction in the spinal cord, which could be an undiscovered contributing factor to diabetic neuropathic pain.


Subject(s)
Diabetes Complications/etiology , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Diabetic Neuropathies/etiology , Hyperalgesia/etiology , Neuralgia/etiology , Spinal Cord/pathology , Animals , Cells, Cultured , Diabetes Complications/metabolism , Diabetes Complications/pathology , Diabetes Complications/prevention & control , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Angiopathies/prevention & control , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Diabetic Neuropathies/prevention & control , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hyperalgesia/prevention & control , Male , Mice , Mice, Knockout , Mice, Transgenic , Microvessels/physiopathology , Neuralgia/metabolism , Neuralgia/pathology , Neuralgia/prevention & control , Rats , Rats, Sprague-Dawley , Spinal Cord/blood supply , Spinal Cord/metabolism , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/physiology
4.
Oncogene ; 34(33): 4311-9, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-25381816

ABSTRACT

Angiogenesis is required for tumour growth and is induced principally by vascular endothelial growth factor A (VEGF-A). VEGF-A pre-mRNA is alternatively spliced at the terminal exon to produce two families of isoforms, pro- and anti-angiogenic, only the former of which is upregulated in prostate cancer (PCa). In renal epithelial cells and colon cancer cells, the choice of VEGF splice isoforms is controlled by the splicing factor SRSF1, phosphorylated by serine-arginine protein kinase 1 (SRPK1). Immunohistochemistry staining of human samples revealed a significant increase in SRPK1 expression both in prostate intra-epithelial neoplasia lesions as well as malignant adenocarcinoma compared with benign prostate tissue. We therefore tested the hypothesis that the selective upregulation of pro-angiogenic VEGF in PCa may be under the control of SRPK1 activity. A switch in the expression of VEGF165 towards the anti-angiogenic splice isoform, VEGF165b, was seen in PC-3 cells with SRPK1 knockdown (KD). PC-3 SRPK1-KD cells resulted in tumours that grew more slowly in xenografts, with decreased microvessel density. No effect was seen as a result of SRPK1-KD on growth, proliferation, migration and invasion capabilities of PC-3 cells in vitro. Small-molecule inhibitors of SRPK1 switched splicing towards the anti-angiogenic isoform VEGF165b in PC-3 cells and decreased tumour growth when administered intraperitoneally in an orthotopic mouse model of PCa. Our study suggests that modulation of SRPK1 and subsequent inhibition of tumour angiogenesis by regulation of VEGF splicing can alter prostate tumour growth and supports further studies for the use of SRPK1 inhibition as a potential anti-angiogenic therapy in PCa.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/metabolism , RNA Splicing/drug effects , Vascular Endothelial Growth Factor A/metabolism
5.
Neurobiol Dis ; 71: 245-59, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25151644

ABSTRACT

Vascular endothelial growth factor-A (VEGF-A) is best known as a key regulator of the formation of new blood vessels. Neutralization of VEGF-A with anti-VEGF therapy e.g. bevacizumab, can be painful, and this is hypothesized to result from a loss of VEGF-A-mediated neuroprotection. The multiple vegf-a gene products consist of two alternatively spliced families, typified by VEGF-A165a and VEGF-A165b (both contain 165 amino acids), both of which are neuroprotective. Under pathological conditions, such as in inflammation and cancer, the pro-angiogenic VEGF-A165a is upregulated and predominates over the VEGF-A165b isoform. We show here that in rats and mice VEGF-A165a and VEGF-A165b have opposing effects on pain, and that blocking the proximal splicing event - leading to the preferential expression of VEGF-A165b over VEGF165a - prevents pain in vivo. VEGF-A165a sensitizes peripheral nociceptive neurons through actions on VEGFR2 and a TRPV1-dependent mechanism, thus enhancing nociceptive signaling. VEGF-A165b blocks the effect of VEGF-A165a. After nerve injury, the endogenous balance of VEGF-A isoforms switches to greater expression of VEGF-Axxxa compared to VEGF-Axxxb, through an SRPK1-dependent pre-mRNA splicing mechanism. Pharmacological inhibition of SRPK1 after traumatic nerve injury selectively reduced VEGF-Axxxa expression and reversed associated neuropathic pain. Exogenous VEGF-A165b also ameliorated neuropathic pain. We conclude that the relative levels of alternatively spliced VEGF-A isoforms are critical for pain modulation under both normal conditions and in sensory neuropathy. Altering VEGF-Axxxa/VEGF-Axxxb balance by targeting alternative RNA splicing may be a new analgesic strategy.


Subject(s)
Antibodies/therapeutic use , DNA, Recombinant/genetics , Neuralgia/metabolism , Neuralgia/therapy , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A , Animals , Antibodies/pharmacology , Benzofurans , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Ganglia, Spinal/cytology , Hyperalgesia/metabolism , Male , Mice , Mice, Transgenic , Neural Conduction/genetics , Pain Measurement , Pain Threshold/physiology , Quinolines , RNA, Messenger/genetics , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
6.
Br J Cancer ; 111(3): 477-85, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25010863

ABSTRACT

BACKGROUND: Current therapies for metastatic melanoma are targeted either at cancer mutations driving growth (e.g., vemurafenib) or immune-based therapies (e.g., ipilimumab). Tumour progression also requires angiogenesis, which is regulated by VEGF-A, itself alternatively spliced to form two families of isoforms, pro- and anti-angiogenic. Metastatic melanoma is associated with a splicing switch to pro-angiogenic VEGF-A, previously shown to be regulated by SRSF1 phosphorylation by SRPK1. Here, we show a novel approach to preventing angiogenesis-targeting splicing factor kinases that are highly expressed in melanomas. METHODS: We used RT-PCR, western blotting and immunohistochemistry to investigate SRPK1, SRSF1 and VEGF expression in tumour cells, and in vivo xenograft assays to investigate SRPK1 knockdown and inhibition in vivo. RESULTS: In both uveal and cutaneous melanoma cell lines, SRPK1 was highly expressed, and inhibition of SRPK1 by knockdown or with pharmacological inhibitors reduced pro-angiogenic VEGF expression maintaining the production of anti-angiogenic VEGF isoforms. Both pharmacological SRPK1 inhibitors and SRPK1 knockdown reduced growth of human melanomas in vivo, but neither affected cell proliferation in vitro. CONCLUSIONS: These results suggest that selective blocking of pro-angiogenic isoforms by inhibiting splice-site selection with SRPK1 inhibitors reduces melanoma growth. SRPK1 inhibitors may be used as therapeutic agents.


Subject(s)
Melanoma/metabolism , Neovascularization, Pathologic/metabolism , Protein Serine-Threonine Kinases/metabolism , Skin Neoplasms/metabolism , Vascular Endothelial Growth Factor A/physiology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Melanoma/drug therapy , Melanoma/secondary , Mice , Mice, Nude , Molecular Targeted Therapy , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Piperidines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Splicing , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Tumor Burden , Xenograft Model Antitumor Assays
7.
Oncogene ; 33(46): 5311-8, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-24336324

ABSTRACT

The immense majority of genes are alternatively spliced and there are many isoforms specifically associated with cancer progression and metastasis. The splicing pattern of specific isoforms of numerous genes is altered as cells move through the oncogenic process of gaining proliferative capacity, acquiring angiogenic, invasive, antiapoptotic and survival properties, becoming free from growth factor dependence and growth suppression, altering their metabolism to cope with hypoxia, enabling them to acquire mechanisms of immune escape, and as they move through the epithelial-mesenchymal and mesenchymal-epithelial transitions and metastasis. Each of the 'hallmarks of cancer' is associated with a switch in splicing, towards a more aggressive invasive cancer phenotype. The choice of isoforms is regulated by several factors (signaling molecules, kinases, splicing factors) currently being identified systematically by a number of high-throughput, independent and unbiased methodologies. Splicing factors are de-regulated in cancer, and in some cases are themselves oncogenes or pseudo-oncogenes and can contribute to positive feedback loops driving cancer progression. Tumour progression may therefore be associated with a coordinated splicing control, meaning that there is the potential for a relatively small number of splice factors or their regulators to drive multiple oncogenic processes. The understanding of how splicing contributes to the various phenotypic traits acquired by tumours as they progress and metastasise, and in particular how alternative splicing is coordinated, can and is leading to the development of a new class of anticancer therapeutics-the alternative-splicing inhibitors.


Subject(s)
Alternative Splicing , Neoplasms/genetics , Oncogene Proteins/genetics , Signal Transduction/genetics , Antineoplastic Agents/therapeutic use , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Oncogene Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/drug effects
8.
Reproduction ; 143(4): 501-11, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22232745

ABSTRACT

Angiogenesis and vascular regression are critical for the female ovulatory cycle. They enable progression and regression of follicular development, and corpora lutea formation and regression. Angiogenesis in the ovary occurs under the control of the vascular endothelial growth factor-A (VEGFA) family of proteins, which are generated as both pro-(VEGF(165)) and anti(VEGF(165)b)-angiogenic isoforms by alternative splicing. To determine the role of the VEGF(165)b isoforms in the ovulatory cycle, we measured VEGF(165)b expression in marmoset ovaries by immunohistochemistry and ELISA, and used transgenic mice over-expressing VEGF(165)b in the ovary. VEGF(165)b was expressed in the marmoset ovaries in granulosa cells and theca, and the balance of VEGF(165)b:VEGF(165) was regulated during luteogenesis. Mice over-expressing VEGF(165)b in the ovary were less fertile than wild-type littermates, had reduced secondary and tertiary follicles after mating, increased atretic follicles, fewer corpora lutea and generated fewer embryos in the oviduct after mating, and these were more likely not to retain the corona radiata. These results indicate that the balance of VEGFA isoforms controls follicle progression and luteogenesis, and that control of isoform expression may regulate fertility in mammals, including in primates.


Subject(s)
Fertility , Ovary/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Callithrix , Down-Regulation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Ovary/growth & development , Pregnancy
9.
BJOG ; 118(10): 1253-61, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21668772

ABSTRACT

OBJECTIVE: Pre-eclampsia is diagnosed by hypertension and proteinuria, probably caused by endothelial dysfunction, resulting in symptoms including oedema, inflammation and altered metabolism. Vascular endothelial growth factor A (VEGF-A) is detected at higher concentrations in plasma from patients with pre-eclampsia than in plasma from normotensive pregnant patients when determined by radioimmunoassay. This study tested the hypothesis that circulating VEGF-A in pre-eclamptic plasma is biologically active in vivo, and aimed to identify specific isoforms responsible for this activity. DESIGN: Plasma from pre-eclamptic (n = 17) and normotensive (n = 10) pregnant women was perfused into Rana mesenteric microvessels, and the subsequent change in microvascular permeability was measured using a single-vessel perfusion micro-occlusion technique. RESULTS: Pre-eclamptic but not normotensive plasma resulted in a 5.25 ± 0.8-fold acute increase in vascular permeability (P = 0.0003). This increase could be blocked by the incubation of plasma with bevacizumab, an antibody to VEGF-A (n = 7; P = 0012), and by VEGF-A receptor inhibition by SU5416 at doses specific to VEGF-A receptor-1 (VEGFR1), but not by the VEGF-A receptor-2 inhibitor, ZM323881. Although VEGF(165) b levels were not significantly altered in the PET samples, the increase in permeability was also inhibited by incubation of pre-eclamptic plasma with an inhibitory monoclonal antibody specific for VEGF165b (n=6; P<0.01), or by the addition of placental growth factor 1 (PlGF-1; n = 3; P < 0.001). PlGF-1 was detected at lower concentrations in pre-eclamptic plasma than in normotensive plasma. CONCLUSIONS: These findings suggest that circulating VEGF-A levels in pre-eclampsia are biologically active because of a loss of repression of VEGFR1 signalling by PlGF-1, and VEGF165b may be involved in the increased vascular permeability of pre-eclampsia.


Subject(s)
Capillary Permeability/physiology , Pre-Eclampsia/blood , Vascular Endothelial Growth Factor A/blood , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Biological Assay , Female , Humans , Membrane Proteins/blood , Pregnancy , Quinazolines/pharmacology , Ranidae , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
10.
Curr Eye Res ; 36(4): 328-35, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21309690

ABSTRACT

PURPOSE: Alternative splicing of the last exon (exon 8) of vascular endothelial growth factor (VEGF) pre-mRNA is a key element in the balance of pro- and anti-angiogenic VEGF isoforms in exudative age-related macular degeneration (exAMD) and proliferative diabetic retinopathy (PDR). Three splicing factors, SRp40, ASF/SF2, and SRp55 are predicted to control alternative splicing by binding to exonic splice enhancers (ESE) in VEGF exon 8. This pilot study examines whether there is an association between angiogenic eye disease and splicing factor polymorphisms, and whether there are sequence variations in the alternative splice sites of the VEGF gene. MATERIALS AND METHODS: A case:control pilot study comparing 163 individuals with angiogenic eye disease (94 exAMD and 69 PDR patients) with 95 age-matched controls. Splicing factor polymorphisms were genotyped by Restriction Fragment Length Polymorphism (RFLP) and sequencing, and the VEGF alternatively spliced region was assessed by denaturing High Performance Liquid Chromatography (dHPLC) using a transgenomic WAVE heteroduplex analyzer. RESULTS: No variations were observed in the alternatively spliced region of VEGF exon 8. ASF/SF2 polymorphisms showed no association with exAMD or PDR. For PDR, we observed a trend in SRp40 (rs6573908) where the 5136CC genotype was more frequent in controls (p = 0.0517) and a significant association of the SRp55 (rs2235611), where the 2994C allele was more common in the PDR group (p = 0.03). This remained strong, but not significant, after logistic regression for age, sex, disease type, and duration (p = 0.06). CONCLUSIONS: The lack of variation in the VEGF alternatively spliced region suggests the importance of sequence conservation in this area in maintaining the balance of pro- and anti-angiogenic VEGF isoforms. The link between PDR and the SRp55 2994 polymorphism suggests a disease-specific association between factors controlling VEGF splicing and ocular angiogenesis.


Subject(s)
Alternative Splicing/genetics , Choroidal Neovascularization/genetics , Polymorphism, Single Nucleotide , Retinal Neovascularization/genetics , Vascular Endothelial Growth Factor A/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Chromatography, High Pressure Liquid , Diabetic Retinopathy/genetics , Exons/genetics , Female , Genotype , Heteroduplex Analysis , Humans , Macular Degeneration/genetics , Male , Middle Aged , Nuclear Proteins/genetics , Phosphoproteins/genetics , Pilot Projects , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Protein Isoforms/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Young Adult
11.
Br J Cancer ; 101(7): 1183-93, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19707198

ABSTRACT

BACKGROUND: The key mediator of new vessel formation in cancer and other diseases is VEGF-A. VEGF-A exists as alternatively spliced isoforms - the pro-angiogenic VEGF(xxx) family generated by exon 8 proximal splicing, and a sister family, termed VEGF(xxx)b, exemplified by VEGF(165)b, generated by distal splicing of exon 8. However, it is unknown whether this anti-angiogenic property of VEGF(165)b is a general property of the VEGF(xxx)b family of isoforms. METHODS: The mRNA and protein expression of VEGF(121)b was studied in human tissue. The effect of VEGF(121)b was analysed by saturation binding to VEGF receptors, endothelial migration, apoptosis, xenograft tumour growth, pre-retinal neovascularisation and imaging of biodistribution in tumour-bearing mice with radioactive VEGF(121)b. RESULTS: The existence of VEGF(121)b was confirmed in normal human tissues. VEGF(121)b binds both VEGF receptors with similar affinity as other VEGF isoforms, but inhibits endothelial cell migration and is cytoprotective to endothelial cells through VEGFR-2 activation. Administration of VEGF(121)b normalised retinal vasculature by reducing both angiogenesis and ischaemia. VEGF(121)b reduced the growth of xenografted human colon tumours in association with reduced microvascular density, and an intravenous bolus of VEGF(121)b is taken up into colon tumour xenografts. CONCLUSION: Here we identify a second member of the family, VEGF(121)b, with similar properties to those of VEGF(165)b, and underline the importance of the six amino acids of exon 8b in the anti-angiogenic activity of the VEGF(xxx)b isoforms.


Subject(s)
Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/physiology , Aged , Aged, 80 and over , Alternative Splicing , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Colon/chemistry , Colonic Neoplasms/chemistry , Endothelial Cells/physiology , Female , Humans , Male , Mice , Middle Aged , Neoplasms/pathology , Protein Isoforms , Tissue Distribution , Vascular Endothelial Growth Factor A/pharmacokinetics
12.
Br J Cancer ; 98(8): 1366-79, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18349829

ABSTRACT

Bevacizumab, an anti-vascular endothelial growth factor (VEGF-A) antibody, is used in metastatic colorectal carcinoma (CRC) treatment, but responses are unpredictable. Vascular endothelial growth factor is alternatively spliced to form proangiogenic VEGF(165) and antiangiogenic VEGF(165)b. Using isoform-specific enzyme-linked immunosorbent assay and quantitative polymerase chain reaction, we found that over 90% of the VEGF in normal colonic tissue was VEGF(xxx)b, but there was a variable upregulation of VEGF(xxx) and downregulation of VEGF(xxx)b in paired human CRC samples. Furthermore, cultured colonic adenoma cells expressed predominantly VEGF(xxx)b, whereas colonic carcinoma cells expressed predominantly VEGF(xxx). However, adenoma cells exposed to hypoxia switched their expression from predominantly VEGF(xxx)b to predominantly VEGF(xxx). VEGF(165)b overexpression in LS174t colon cancer cells inhibited colon carcinoma growth in mouse xenograft models. Western blotting and surface plasmon resonance showed that VEGF(165)b bound to bevacizumab with similar affinity as VEGF(165). However, although bevacizumab effectively inhibited the rapid growth of colon carcinomas expressing VEGF(165), it did not affect the slower growth of tumours from colonic carcinoma cells expressing VEGF(165)b. Both bevacizumab and anti-VEGF(165)b-specific antibodies were cytotoxic to colonic epithelial cells, but less so to colonic carcinoma cells. These results show that the balance of antiangiogenic to proangiogenic isoforms switches to a variable extent in CRC, regulates tumour growth rates and affects the sensitivity of tumours to bevacizumab by competitive binding. Together with the identification of an autocrine cytoprotective role for VEGF(165)b in colonic epithelial cells, these results indicate that bevacizumab treatment of human CRC may depend upon this balance of VEGF isoforms.


Subject(s)
Angiogenesis Inhibitors/physiology , Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/physiology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Mice , Protein Isoforms , RNA Splicing , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors
13.
Br J Cancer ; 97(2): 223-30, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17595666

ABSTRACT

Malignant melanoma is the most lethal of the skin cancers and the UK incidence is rising faster than that of any other cancer. Angiogenesis - the growth of new vessels from preexisting vasculature - is an absolute requirement for tumour survival and progression beyond a few hundred microns in diameter. We previously described a class of anti-angiogenic isoforms of VEGF, VEGF(xxx)b, that inhibit tumour growth in animal models, and are downregulated in some cancers, but have not been investigated in melanoma. To determine whether VEGF(xxx)b expression was altered in melanoma, PCR and immunohistochemistry of archived human tumour samples were used. In normal epidermis and in a proportion of melanoma samples, VEGF(xxx)b staining was seen. Some melanomas had much weaker staining. Subsequent examination revealed that expression was significantly reduced in primary melanoma samples (both horizontal and vertical growth phases) from patients who subsequently developed tumour metastasis compared with those who did not (analysis of variance (ANOVA) P<0.001 metastatic vs nonmetastatic), irrespective of tumour thickness, while the surrounding epidermis showed no difference in expression. Staining for total VEGF expression showed staining in metastatic and nonmetastatic melanomas, and normal epidermis. An absence of VEGF(xxx)b expression appears to predict metastatic spread in patients with primary melanoma. These results suggest that there is a switch in splicing as part of the metastatic process, from anti-angiogenic to pro-angiogenic VEGF isoforms. This may form part of a wider metastatic splicing phenotype.


Subject(s)
Angiogenesis Inhibitors/metabolism , Melanoma/pathology , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Alternative Splicing , Angiogenesis Inhibitors/analysis , Angiogenesis Inhibitors/genetics , Down-Regulation , Humans , Melanoma/blood supply , Melanoma/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Prognosis , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Skin/blood supply , Skin/metabolism , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics
14.
Diabetologia ; 50(7): 1423-32, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17476476

ABSTRACT

AIMS/HYPOTHESIS: Rapamycin, part of the immunosuppressive regimen of the Edmonton protocol, has been shown to inhibit vascular endothelial growth factor (VEGF) production and VEGF-mediated survival signalling in tumour cell lines. This study investigates the survival-promoting activities of VEGF in human islets and the effects of rapamycin on islet viability. MATERIALS AND METHODS: Levels of VEGF and its receptors in isolated human islets and whole pancreas was determined by western blotting and immunostaining. Islet viability following VEGF or immunosuppressive drug treatment was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Islet VEGF release was measured by ELISA. Mouse islets infected with an adenovirus expressing the gene for VEGF were transplanted syngeneically into streptozotocin-induced diabetic mice, with blood glucose levels measured three times per week. RESULTS: Isolated human islets produced multiple isoforms of VEGF and VEGF receptors 1, 2 and 3 and the coreceptor neuropilin 1. Exogenous VEGF (10 ng/ml) prevented human islet death induced by serum starvation, which suggests that VEGF can act as a survival factor for human islets. Transplantation of mouse islets infected with a VEGF-expressing adenovirus in a syngeneic model, improved glycaemic control at day 1 post-transplantation (p < 0.05). Rapamycin at 10 and 100 ng/ml significantly reduced islet VEGF release (by 37 +/- 4% and 43 +/- 6%, respectively; p < 0.05) and at 100 ng/ml reduced islet viability (by 36 +/- 9%) and insulin release (by 47 +/- 7%, all vs vehicle-treated controls; p < 0.05). Tacrolimus had no effect on islet VEGF release or viability. CONCLUSIONS/INTERPRETATION: Our data suggest that rapamycin may have deleterious effects on islet survival post-transplantation, both through a direct effect on islet viability and indirectly through blockade of VEGF-mediated revascularisation.


Subject(s)
Immunosuppressive Agents/pharmacology , Islets of Langerhans/pathology , Neoplasms/mortality , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Survival , Cells, Cultured , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neovascularization, Pathologic , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Sirolimus/pharmacology
15.
Oncogene ; 26(21): 2997-3005, 2007 May 10.
Article in English | MEDLINE | ID: mdl-17130836

ABSTRACT

The mechanisms that cause tumors such as melanomas to metastasize into peripheral lymphatic capillaries are poorly defined. Non-mutually-exclusive mechanisms are lymphatic endothelial cell (LEC) chemotaxis and proliferation in response to tumor cells (chemotaxis-lymphangiogenesis hypothesis) or LECs may secrete chemotactic agents that attract cancer cells (chemotactic metastasis hypothesis). Using migration assays, we found evidence supporting both hypotheses. Conditioned medium (CM) from metastatic malignant melanoma (MMM) cell lines attracted LEC migration, consistent with the lymphangiogenesis hypothesis. Conversely, CM from mixed endothelial cells or LECs, but not blood endothelial cells, attracted MMM cells but not non-metastatic melanoma cells, consistent with the chemotactic metastasis hypothesis. MMM cell lines expressed CCR7 receptors for the lymphatic chemokine CCL21 and CCL21 neutralizing antibodies prevented MMM chemotaxis in vitro. To test for chemotactic metastasis in vivo tumor cells were xenotransplanted into nude mice approximately 1 cm from an injected LEC depot. Two different MMM grew directionally towards the LECs, whereas non-metastatic melanomas did not. These observations support the hypothesis that MMM cells grow towards regions of high LEC density owing to chemotactic LEC secretions, including CCL21. This chemotactic metastasis may contribute to the close association between metastasizing tumor cells and peri-tumor lymphatic density and promote lymphatic invasion.


Subject(s)
Cell Movement/physiology , Chemokines/physiology , Lymphatic Metastasis/pathology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Animals , Biomarkers, Tumor/analysis , Cells, Cultured , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/pathology , Humans , Ki-67 Antigen/analysis , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Neoplasm Transplantation/pathology
16.
Lymphat Res Biol ; 4(2): 73-82, 2006.
Article in English | MEDLINE | ID: mdl-16808669

ABSTRACT

OBJECTIVE: To characterize vascular endothelial growth factor-C (VEGF-C) protein expression in normal human tissues by immunohistochemistry (IHC). VEGF-C is a growth factor for lymphatic endothelial cells. VEGF-C mRNA and protein are expressed in a variety of cancerous tissues, but the localization of VEGF-C protein in many normal human tissues has not been clearly demonstrated to date. We therefore performed an immunohistochemical survey of the distribution of intracellular VEGF-C protein in a range of normal human tissue types. METHODS: Five microm sections were cut from archived human tissues. Sections were dewaxed, rehydrated, and subjected to microwave pretreatment. They were incubated with VEGF-C antibody before detection with biotinylated secondary antibody using 'Elite' avidin-biotin enzyme complex and diaminobenzidine substrate. The primary antibody recognized the C-terminus of the VEGF-C propeptide that is cleaved before secretion and hence only cellular protein was detected. Negative controls used the same concentration of normal goat IgG. RESULTS: Staining manifested as small punctate cytoplasmic granules. Strong expression was observed in large intestine epithelium, and mammary duct epithelium, skeletal and cardiac muscle, thyroid, ovary, and the prostate. Weaker expression was also detected in the hepatocytes close to the terminal hepatic venules of the liver, vascular smooth muscle, and placenta. No expression was consistently detected in spleen or thymus. CONCLUSIONS: Intracellular VEGF-C protein is widely expressed in many normal human adult tissues. Its expression in cancer is not therefore per se indicative of a prolymphangiogenic change. To demonstrate the latter, a quantitative change in expression level is required.


Subject(s)
Vascular Endothelial Growth Factor C/metabolism , Female , Humans , Immunohistochemistry , Lymphoid Tissue/metabolism , Mesoderm/metabolism , Muscles/metabolism , Ovary/metabolism , Placenta/metabolism , Reference Values
17.
Arterioscler Thromb Vasc Biol ; 26(8): 1768-76, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16763162

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF) exerts many of its effects by stimulating endothelial calcium influx, but little is known about channels mediating VEGF-induced cation entry. The aim of this study was to measure and characterize for the first time the VEGF-activated cation current in human microvascular endothelial cells (HMVECs). METHODS AND RESULTS: Whole-cell patch-clamp recordings were made from HMVECs. During applied voltage ramps, VEGF activated a current that reversed at 0 mV, was sensitive to gadolinium, and required extracellular cations. Noise analysis yielded a single-channel conductance of 27 pS. The current was not dependent on intracellular calcium stores, and was not blocked by inositol triphosphate (IP3) receptor or serine/threonine kinase inhibition but was partially inhibited by flufenamic acid. A similar current was activated by 1-oleoyl-2-acetyl-sn-glycerol (OAG), a membrane-permeant analog of diacylglycerol (DAG). To determine whether VEGF could activate recombinant ion channels with similar properties, we investigated the effect of VEGF on Chinese hamster ovary cells cotransfected with VEGFR2 and the canonical transient receptor potential (TRPC) channels, TRPC3 or TRPC6. VEGF induced a similar current to that described above in VEGFR2-TRPC3 and VEGFR2-TRPC6 cells but not in cells transfected with either cDNA alone. CONCLUSIONS: VEGF activates a receptor-operated cation current in HMVECs and OAG can activate directly a similar current in these cells. VEGF is also able to activate heterologously expressed TRPC3/6 channels through VEGFR2.


Subject(s)
Calcium Channels/drug effects , Calcium Channels/physiology , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , CHO Cells , Cations/metabolism , Cricetinae , Cricetulus , Diglycerides/pharmacology , Electric Conductivity , Humans , Microcirculation , Patch-Clamp Techniques , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , Vascular Endothelial Growth Factor Receptor-2/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Am J Physiol Renal Physiol ; 291(1): F196-207, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16525158

ABSTRACT

Vascular endothelial growth factor (VEGF)-A is an autocrine survival factor for podocytes, which express two VEGF receptors, VEGF-R1 and VEGF-R3. As VEGF-A is not a known ligand for VEGF-R3, the aim of this investigation was to examine whether VEGF-C, a known ligand for VEGF-R3, served a function in podocyte biology and whether this was VEGF-R3 dependent. VEGF-C protein expression was localized to podocytes in contrast to VEGF-D, which was expressed in parietal epithelial cells. Intracellular calcium ([Ca2+]i) experiments demonstrated that VEGF-C induced a 0.74+/-0.09-fold reduction in [Ca2+]i compared with baseline in human conditionally immortalized podocytes (hCIPs; P<0.05, one sample t-test, n=8). Cytotoxicity experiments revealed that in hCIPs VEGF-C reduced cytotoxicity to 81.4+/-1.9% of serum-starved conditions (P<0.001, paired t-test, n=16), similar to VEGF-A (82.8+/-4.5% of serum-starved conditions, P<0.05, paired t-test). MAZ51 (a VEGF-R3 kinase inhibitor) inhibited the VEGF-C-induced reduction in cytotoxicity (106.2+/-2.1% of serum-starved conditions), whereas MAZ51 by itself had no cytotoxic effects on hCIPs. VEGF-C was also shown to induce a 0.5+/-0.13-fold reduction in levels of MAPK phosphorylation compared with VEGF-A and VEGF-A-Mab treatment (P<0.05, ANOVA, n=4), yet had no effect on Akt phosphorylation. Surprisingly, immunoprecipitation studies detected no VEGF-C-induced autophosphorylation of VEGF-R3 in hCIPs but did so in HMVECs. Moreover, SU-5416, a tyrosine kinase inhibitor, blocked the VEGF-C-induced reduction in cytotoxicity (106+/-2.8% of serum-starved conditions) at concentrations specific for VEGF-R1. Together, these results suggest for the first time that VEGF-C acts in an autocrine manner in cultured podocytes to promote survival, although the receptor or receptor complex activated has yet to be elucidated.


Subject(s)
Cell Survival/drug effects , Cell Survival/physiology , Podocytes/drug effects , Podocytes/physiology , Vascular Endothelial Growth Factor C/physiology , Calcium/analysis , Calcium/metabolism , Cell Line , Endothelial Cells/chemistry , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Immunoprecipitation , Indoles/pharmacology , Kidney Glomerulus/chemistry , Mitogen-Activated Protein Kinase Kinases/analysis , Mitogen-Activated Protein Kinase Kinases/physiology , Naphthalenes/pharmacology , Phosphorylation , Podocytes/chemistry , Podocytes/cytology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/physiology , Pyrroles/pharmacology , Receptors, Vascular Endothelial Growth Factor/analysis , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor D/analysis , Vascular Endothelial Growth Factor D/physiology
19.
J Physiol ; 572(Pt 1): 243-57, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16423853

ABSTRACT

Vascular endothelial growth factor (VEGF) is the principal agent that increases microvascular permeability during physiological and pathological angiogenesis. VEGF is differentially spliced to form two families of isoforms: VEGF(xxx), and VEGF(xxx)b. Whereas VEGF(165) stimulates angiogenesis, VEGF(165)b is anti-angiogenic. To determine the effect of VEGF(165)b on permeability, hydraulic conductivity (L(p)) was measured in individually perfused microvessels in the mesentery of frogs and rats. As with VEGF(165), VEGF(165)b increased L(p) in amphibian (2.4 +/- 0.3-fold) and mammalian (1.9 +/- 0.2-fold) mesenteric microvessels. A dose-response relationship showed that VEGF(165)b (EC(50), 0.65 pm) was approximately 25 times more potent than VEGF(165) (EC(50), 16 pm) in amphibian microvessels. VEGF(165) has been shown to increase permeability through VEGF receptor 2 (VEGF-R2) signalling. However, VEGF(165)b increased L(p) of frog vessels to the same extent in the presence of the VEGF-R2 inhibitor ZM323881, indicating that it does not increase permeability via VEGF-R2 signalling, and was inhibited by the VEGF receptor inhibitor SU5416 at doses that are specific for VEGF receptor 1 (VEGF-R1). VEGF(165)b, in contrast to VEGF(165), did not result in a sustained chronic increase in L(p). These results show that although VEGF(165)b is anti-angiogenic in the mesentery, it does signal in endothelial cells in vivo resulting in a transient, but not sustained, increase in microvascular L(p), probably through VEGF-R1.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Capillary Permeability/physiology , Mesentery/blood supply , Mesentery/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Capillary Permeability/drug effects , Dose-Response Relationship, Drug , Kinetics , Male , Mesentery/drug effects , Neovascularization, Physiologic/drug effects , Protein Isoforms/administration & dosage , Ranidae , Rats , Rats, Wistar
20.
Diabetologia ; 48(11): 2422-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16193288

ABSTRACT

AIMS/HYPOTHESIS: Proliferative diabetic retinopathy results from excess blood vessel growth into the vitreous fluid of the eye. Retinal angiogenesis is regulated by expression of vascular endothelial growth factor (VEGF), and many studies have shown that VEGF is critically involved in proliferative diabetic retinopathy. VEGF is alternatively spliced to form the angiogenic (VEGF(xxx)) and potentially anti-angiogenic (VEGF(xxx)b) family of isoforms. The VEGF(xxx)b family is found in normal tissues, but down-regulated in renal and prostate cancer. Previous studies on endogenous expression of VEGF in the eye have not distinguished between the two families of isoforms. METHODS: We measured VEGF(xxx)b isoform expression in normal human eye tissue (lens, sclera, retina and iris) and vitreous fluid using enzyme-linked immunosorbent assay and Western blotting with a VEGF(xxx)b-specific antibody. RESULTS: VEGF(xxx)b protein was expressed in lens, sclera, retina, iris and vitreous fluid. Multiple isoforms were seen, including VEGF(165)b, VEGF(121)b, VEGF(145)b, VEGF(183)b and VEGF(189)b. In non-diabetic patients, 64+/-7% of the VEGF in the vitreous was VEGF(xxx)b (n=18), whereas in diabetic patients only 12.5+/-3.6% of total VEGF was VEGF(xxx)b. CONCLUSIONS/INTERPRETATION: Since VEGF(xxx)b inhibits VEGF(xxx)-induced angiogenesis in a one-to-one stoichiometric manner, these results show that in the eye of diabetic patients VEGF splicing was switched from an anti-angiogenic to a pro-angiogenic environment. This occurred through changes to the ratio of VEGF(xxx):VEGF(xxx)b. Alterations to splicing, and through that to the balance of VEGF isoforms, could therefore be a potential therapeutic strategy for diabetic retinopathy.


Subject(s)
Alternative Splicing , Diabetic Retinopathy/genetics , Eye/blood supply , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/genetics , Diabetic Retinopathy/pathology , Eye/metabolism , Humans , Reference Values , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...