"VSports在线直播" Skip to main content
The American Journal of Pathology logoLink to The American Journal of Pathology
. 2013 Sep;183(3):918–929. doi: 10.1016/j.ajpath.2013.05.031

"VSports在线直播" VEGF-A165b Is an Endogenous Neuroprotective Splice Isoform of Vascular Endothelial Growth Factor A in Vivo and in Vitro

"VSports在线直播" Nicholas Beazley-Long , Jing Hua , Thomas Jehle , Richard P Hulse , Rick Dersch §, Christina Lehrling , Heather Bevan , Yan Qiu , VSports app下载 - Wolf A Lagrèze , "VSports手机版" David Wynick §, Amanda J Churchill , Patrick Kehoe (V体育平台登录) , Steven J Harper , David O Bates ∗,, Lucy F Donaldson ‡,∗∗
PMCID: PMC3763768  PMID: 23838428

Abstract

Vascular endothelial growth factor (VEGF) A is generated as two isoform families by alternative RNA splicing, represented by VEGF-A165a and VEGF-A165b. These isoforms have opposing actions on vascular permeability, angiogenesis, and vasodilatation. The proangiogenic VEGF-A165a isoform is neuroprotective in hippocampal, dorsal root ganglia, and retinal neurons, but its propermeability, vasodilatatory, and angiogenic properties limit its therapeutic usefulness. In contrast, a neuroprotective effect of endogenous VEGF-A165b on neurons would be advantageous for neurodegenerative pathologies. Endogenous expression of human and rat VEGF-A165b was detected in hippocampal and cortical neurons. VEGF-A165b formed a significant proportion of total VEGF-A in rat brain VSports注册入口. Recombinant human VEGF-A165b exerted neuroprotective effects in response to multiple insults, including glutamatergic excitotoxicity in hippocampal neurons, chemotherapy-induced cytotoxicity of dorsal root ganglion neurons, and retinal ganglion cells (RGCs) in rat retinal ischemia-reperfusion injury in vivo. Neuroprotection was dependent on VEGFR2 and MEK1/2 activation but not on p38 or phosphatidylinositol 3–kinase activation. Recombinant human VEGF-A165b is a neuroprotective agent that effectively protects both peripheral and central neurons in vivo and in vitro through VEGFR2, MEK1/2, and inhibition of caspase-3 induction. VEGF-A165b may be therapeutically useful for pathologies that involve neuronal damage, including hippocampal neurodegeneration, glaucoma diabetic retinopathy, and peripheral neuropathy. The endogenous nature of VEGF-A165b expression suggests that non–isoform-specific inhibition of VEGF-A (for antiangiogenic reasons) may be damaging to retinal and sensory neurons.


Vascular endothelial growth factor (VEGF) A, originally described as a potent vascular permeability and growth factor for endothelial cells, is up-regulated in the brain during stroke and ischemic episodes1 and has been linked with many neuronal diseases. The most widely studied isoform of VEGF-A, VEGF-A165a, is up-regulated in hypoxia, induces increased vascular permeability in neuronal vasculature, and can stimulate angiogenesis after ischemic episodes. The resulting edema and hyperemia can be damaging, but VEGF-A165a has also been found to have direct anticytotoxic effects on neurons, raising the possibility that it may act as an endogenous neuroprotective agent in neurodegenerative pathologies. VEGF-A exerts neurotrophic (survival) and neurotropic (neurogenesis and axon outgrowth) actions, which, although initially thought to be a function of increased angiogenesis and perfusion after neuronal injury,2 are now appreciated as direct effects of VEGF-A on neurons V体育官网入口.

The vegfa gene encodes numerous products by differential splicing, but not all isoforms exert the same effects. 3 Alternative splicing of exon 8 leads to two functionally distinct families: the proangiogenic VEGF-Axxxa family and the counteracting VEGF-Axxxb family VSports在线直播. 4,5 VEGF-A165b prevents the VEGF-A165a effects on increased vascular permeability, blood vessel growth, and vasodilatation. 4–7.

The therapeutic potential of VEGF-A and anti–VEGF-A treatments are now widely recognized, and effective anti–VEGF-A treatments are available in ophthalmology8 and oncology V体育2025版. 9 The finding that VEGF-A is implicated in neuronal disorders (eg, Alzheimer disease, Parkinson disease, Huntington disease, diabetic neuropathy, and amyotrophic lateral sclerosis10) provides a rationale for the use of VEGF-A as a therapeutic agent in neurodegenerative conditions. Although this rationale is supported by preclinical evidence,11 the identification of the VEGF-Axxxb family requires reexamination of VEGF-A isoforms in these contexts to allow for the clear evidence that VEGF-A splicing variants are not functionally equivalent3 and to determine whether augmentation of the proangiogenic isoform family (VEGF-Axxxa) alone may have deleterious effects (eg, in occult malignancy and carcinoma in situ).

The neuroprotective profile of the exon 8 alternatively spliced isoforms VEGF-Axxxb remains unexplored. Interestingly, VEGF-Axxxb isoforms do not exhibit the vascular effects seen with VEGF-Axxxa isoforms, such as a sustained increase in capillary permeability or hypotension VSports. 5,12 The lack of these potential adverse effects may make VEGF-Axxxb isoforms more amenable as therapeutic agents in neurodegenerative diseases.

We therefore tested the hypothesis that VEGF-A165b is neuroprotective for central and peripheral neurons VSports app下载. We found that VEGF-A165b is expressed in central neurons and is neuroprotective in vitro and in vivo. This finding indicates that VEGF-A165b may prove to be a suitable therapeutic agent in neurodegenerative disorders, exhibiting fewer adverse effects than VEGF-A165a.

Materials and Methods

All reagents were sourced from Sigma-Aldrich (Dorset, UK) unless otherwise stated. Antibodies were sourced from R&D Systems (Carlsbad, CA) [αVEGF-A165b (monoclonal antibody [mAb] 3045), α-caspase-3, αTrkA, and αNF-200] or Cell Signaling Technology (Danver, MA) [phospho-p44/42 mitogen-activated protein kinase (MAPK) (9106) and p44/42 MAPK (9102)]. Recombinant human (rh) VEGF-A165b was provided by Philogene (New York, NY) or from R&D Systems. Computer-aided analysis of immunohistochemistry (IHC), retinal Fluorogold staining, and cytotoxicity was performed using Macintosh computers running public domain ImageJ version 1.46 plus Cell Counter Plugin (NIH, Bethesda, MD; available at http://rsb.info.nih.gov/nih-image).

Protein Assessment by IHC and ELISA

VEGF-A protein was localized in fixed or frozen tissue sections using standard immunohistochemical/immunofluorescent techniques13 or was measured in tissue extracts by commercially available ELISA using a VEGFxxxb-specific capture antibody and a pan-VEGF detection antibody for VEGFxxxb isoforms or two pan-VEGF antibodies for total VEGF, as previously described.5 Total VEGF-A and VEGF-A165b were detected using validated, commercially available antibodies. The VEGF-A165b antibody detects the unique C-terminal of the alternatively spliced VEGF-Axxxb family.5,14 Difficulties in the accurate detection of endogenous VEGF isoforms in rodents have been highlighted recently15 in that the use of mouse antibodies to mouse tissues can detect proteins of similar size to the VEGF165b homodimer and heterodimer. These bands are also found if the primary antibody is omitted, indicating that anti-mouse secondary antibodies detect mouse IgG. The use of human and rat tissue sidesteps this issue because the anti-mouse IgG does not detect human or rat IgG. To confirm specificity of staining a nonspecific mouse IgG control was used at the same concentration and under the same conditions, a second negative control where the primary antibody was incubated for 6 hours at 4°C in eight times the concentration of rhVEGF165b as a peptide block before continuing the IHC under the same conditions, and a positive control where the primary antibody was incubated for 6 hours at 4°C in eight times the concentration of rhVEGF165a before continuing the IHC under the same conditions. Human embryonic and adult tissues were obtained under ethical approval by the North Bristol National Health Service Trust or the University of Leiden. All procedures using animals were performed in accordance with the United Kingdom Animals (Scientific Procedures) Act 1986 and with University of Bristol Ethical Review Panel approval.

Excitotoxicity

Glutamate-induced hippocampal neuronal excitotoxicity was assessed in cultures of neonatal hippocampal neurons from twelve 2-day-old CD1 mouse pups as previously described.16 Neurons were cultured on polylysine-coated glass coverslips in 6 well plates (37°C, ambient oxygen, and 5% CO2, 400,000 cells per plate) in neuronal growth media plus B-27 supplement (GIBCO-Invitrogen, Paisley, UK) with penicillin and streptomycin and 1% bovine serum albumin. After 24 hours, cultures were supplemented with 10 μg/mL of 5-fluoro-2′-deoxyuridine to inhibit growth of nonpostmitotic satellite cells. Excitotoxicity assays were started on day 10 of culture. Cultures were exposed to 3 mmol/L l-glutamic acid for 24 hours in the presence of VEGF-A165b (0.01, 0.1, 1, and 10 nmol/L) or 50 nmol/L galanin (Bachem). To determine the mechanism of VEGF-A165b–mediated effects, additional cultures were incubated with VEGF-A165b in the presence of VEGF-A receptor and downstream signaling molecule inhibitors PTK787 (Novartis, New York, NY), SU5416 (Sugen, New York, NY), ZM323881 (AstraZeneca, Wilmington, DE), and PD98059 (Calbiochem, Billerica, MA). Cytotoxicity was assessed by a trained observer masked to treatment using a Live/Dead Viability/Cytotoxicity Kit (Molecular Probes-Invitrogen, Paisley, UK). The cells were fixed and nuclei stained (Hoechst 33258; 1:2300) and mounted to a glass slide. For quantitation purposes, only cells with positive nuclear staining with Hoechst 33258 were counted. Ten random images of each coverslip were taken.

Chemotherapy-Induced Cytotoxicity

Oxaliplatin-induced dorsal root ganglion (DRG) neuronal death was assessed in primary cultures of adult rat DRG neurons. DRG was dissected and enzymatically and mechanically dissociated, and the cells were plated onto poly-l-lysine- and laminin-coated coverslips at approximately 2850 cells/cm2 in Ham's F12 with N2 supplement (Gibco 17502) +0.3% bovine serum albumin. A total of 30 μg/mL of 5-fluoro-2′-deoxyuridine was added to prevent nonpostmitotic cells from proliferating. DRG neurons were cultured for 2 days before overnight pretreatment with test compounds (2.5 nmol/L rhVEGF-A165b, 10 nmol/L rhPlGF, 0 to 100 μg/mL of anti-VEGF165b, 0 to 100 μg/mL of mouse IgG, 10 nmol/L ZM323881, or vehicle). Neurons were then treated with oxaliplatin for 24 hours with test agent or vehicle. Neurons were treated with PrestoBlue Cell Viability Reagent (Molecular Probes; A-13261) for detection of cell viability and absorbance read on a fluorescence plate-reader (PerkinElmer, Waltham, MA), treated with trypan blue for detection of viable cells and trypan blue–positive percentage determined by manual counting, or fixed in 4% paraformaldehyde and subjected to immunofluorescence for the detection of activated caspase-3 (200 ng/mL of Rab mAb overnight; Cell Signaling). Neurons were identified by co-staining with markers NeuN (MAB377; Millipore, Billerica, MA), βIII-tubulin (TU20; Abcam, Cambridge, UK), or neuron-specific enolase 1. The percentage of caspase-3–positive neurons were analyzed by a trained observer masked to treatment. In separate experiments, VEGFR2 expression was detected using a rabbit VEGFR2 mAb (1:500, overnight, 50b11; Cell Signaling). The primary antibody was replaced by concentration- and species-matched IgG to control for each immunofluorescence experiment.

Western Immunoblotting

Rat immortalized sensory neuronal cells (50B1117) were used for protein analysis after VEGF-A165b treatment because of the larger protein content available from these cells compared with that from primary cultures. They were grown to approximately 80% confluence, differentiated with 75 μmol/L forskolin (catalog no. F6886) for 24 hours, and treated with ± .1 nmol/L rhVEGF-A165b, directly to the media for 10 minutes. Protein was extracted in radioimmunoprecipitation assay buffer with protease inhibitor cocktail plus 2 mmol/L NaVO4, separated by SDS-PAGE (12%), and blotted onto polyvinylidene difluoride membrane. This was blocked in 2% bovine serum albumin Tris-buffered saline–Tween (0.05%) for 1 hour, and anti-p44/42 MAPK (Rab polyclonal antibody, no. 9102, 1 in 500; Cell Signaling) and anti–phospho-p44/42 (mouse mAb, no. 9106, 1 in 250; Cell Signaling) were added in blocking solution and incubated overnight at 4°C. Fluorescent secondary antibodies (LI-COR, Lincoln, NE) were added 1 in 10,000 in blocking solution for 1 hour at room temperature and fluorescence visualized on a LI-COR Odyssey.

Neurite Outgrowth

The effect of VEGF-A165b on neurite outgrowth in cultured rat primary DRG neurons was assessed as previously described.18 Adult DRG neurons were cultured with or without 1 nmol/L VEGF-A165b for 24 hours and the length of neurite outgrowth assessed.

RGC Death in Vivo

A total of 27 male Wister rats (250 to 350 g; Charles River, Wilmington, MA) were used to assess the effect of VEGF-A165b on RGC death after ischemia. One week before ischemia/reperfusion injury, RGCs were retrogradely labeled with Fluorogold using a previously described method.19,20 Briefly, Fluorogold (4% in PBS; Fluorochrome Inc, Denver, CO) was injected into bilateral superior colliculi (0.6 μL at 4.2-mm depth and 0.7 μL at 4.7-mm depth) under isoflurane inhalation anesthesia (1-minute induction at 5% in oxygen, reduced to 3% for maintenance) and sterotaxic guidance. Fluorogold is retrogradely transported in RGCs, and somatic labeling is maintained for at least 3 weeks.21 Animals were randomly assigned to 1 of 3 groups: 12 animals received an intravitreal injection of 10 ng of VEGF-A165b in 5 μL of HBSS in one eye, seven received the same volume of HBSS, and eight were untreated. Twenty-four hours after intraocular injection, animals were subject to retinal ischemia as previously described.19,20 Under isoflurane anesthesia and stereotaxic guidance, the pupil was dilated (1% tropicamide and 2.5% phenylepherine eye drops), and anterior chamber pressure was maintained for 60 minutes with 0.9% NaCl infusion that elevated the intraocular pressure to 120 mm Hg. The cessation of the retinal blood flow was observed using direct funduscopy during the procedure. Reperfusion of retinal vessels was observed as intraocular pressure reduced after cessation of perfusion. Twenty-four hours after ischemia, retinas from three animals from each group were stained for activated caspase-3 immunofluorescence to assess the extent of ischemic damage.22 Caspase-3–positive cells were counted independently in the retinal ganglion and inner nuclear layers. The remaining animals were euthanized with isoflurane overdose 10 days after ischemia and the numbers of surviving Fluorogold-labeled neurons counted. Cell counts were performed by an operator masked to treatment.

Partial Saphenous Nerve Injury–Induced Cell Death

Partial saphenous nerve injury (PSNI) on C57/Bl6 mice was performed as previously described23 with the mice under isofluorane anesthesia (2% to 3% in oxygen). An incision (approximately 5 mm) was made in the inguinal fossa region of the right hind leg. Fifty percent of the saphenous nerve was tightly ligated using a size 6.0 sterile silk suture, and the wound was closed with size 4.0 sterile silk suture. Animals in the sham surgery group (n = 5) underwent anesthesia and surgery that involved solely an incision in the inguinal fossa region of the right hind limb. Each PSNI experimental group received biweekly VEGF-A165b (n = 6, 20 ng/g) or PBS vehicle (n = 16, 200 μL). Sham surgery involved the exact same procedure, except without the tight ligation of the saphenous nerve. Controls in the sham surgery group (n = 5) received i.p. PBS at the same times. Injections were given immediately after surgery, and animals were sacrificed after 10 days by overdose of anesthetic and perfuse fixation with 4% paraformaldehyde. DRG sections (10 μm thick) were dissected out, frozen, and stained for activating transcription factor 3 (ATF3, cat no sc188; Santa Cruz, Santa Cruz, CA).

Statistical Analysis

Numbers of repetitions and animals are given in the text and/or figures. All data were analyzed using t-tests (two groups) or one-way analysis of variance and either Student's Newman Keuls post hoc test (when all columns are compared with each other) or Bonferroni post hoc tests when a priori specific tests were used, unless data were ordinal or obviously nongaussian, in which case nonparametric equivalents were used.

Results

IHC of human brain samples demonstrated that VEGF-A165b was strongly expressed in the human hippocampal region (Figure 1A), throughout all three pyramidal regions (CA1, CA2, and CA3) (Figure 1B), and the dentate gyrus (Figure 1C), with neurons also being expressed in occasional neurons scattered through the cortex. The use of a mouse IgG at the same concentration or preincubation of the primary antibody with recombinant VEGF165b demonstrated no significant staining (Figure 1, D and E). Preincubation with recombinant VEGF165a, however, did not block the staining (Figure 1F). To quantify expression, protein was extracted from rat brains and VEGF-Axxxb and total VEGF-A levels measured by ELISA. VEGF-Axxxb levels averaged 45% and 41% of total VEGF-A in the cortex and hippocampus, respectively (Figure 1, G and H).

Figure 1. (VSports在线直播)

Figure 1

VEGF-A165b is expressed in human and rat hippocampus. Human cortical sections from the Human Tissue Authority–licensed South West Dementia Brain Bank were stained with an anti–VEGF-A165b antibody. Hippocampal staining in CA1 (A), CA2 (B), CA3, and dentate gyrus (DG; C). D: Negative control mouse IgG staining of the hippocampus. High-power view of DG stained with mouse IgG (right panel). E: Anti-VEGF165b antibody preincubated with rhVEGF165b reveals no staining of the hippocampus. High-power view (right panel). F: Anti-VEGF165b antibody preincubated with rhVEGF165a reveals similar staining of the hippocampus. G: Protein was extracted from cortex and hippocampus dissected from rat brains (n = 3) and subjected to ELISA for VEGF-A and VEGF-Axxxb. VEGF-Axxxa levels were estimated from the difference between total VEGF-A and VEGF-Axxxb. H: Percentage of total VEGF-A that is VEGF-Axxxb.

Because VEGF-Axxxa splice variants are known to be neuroprotective, we assessed the effectiveness of VEGF-Axxxb splice variants as neuroprotective agents in hippocampal neurons, central nervous system neurons that are particularly vulnerable to damage.

RhVEGF-A165b reversed glutamate-induced hippocampal neuronal death in a concentration-dependent manner (Figure 2, A and B) and was no less potent than galanin, which has previously been described as a potent hippocampal neuroprotective agent16 (Figure 2B). At 10 nmol/L, the maximum VEGF-A165b concentration used, cell death was the same as untreated cells, revealing complete inhibition of excitotoxicity. VEGF-A165b is a weak partial agonist at VEGF-A receptor 2 (VEGFR2, also known as Flk1) in vitro.24 In hippocampal neurons, the neuroprotective action of VEGF-A165b was dependent on VEGFR2 activation (Figure 2C), as has been described for VEGF-A165a.25 The partial reversal of excitotoxic cell death seen in the presence of VEGF-A165b was blocked by the 100 nmol/L VEGFR blockers PTK787 (PTK, blocks both VEGFR1 and VEGFR226) and 10 nmol/L ZM323881 (ZM, blocks VEGFR227) but was unaffected by SU5146 (SU, at 100 nmol/L has specificity for VEGFR112). Hippocampal neurons in culture expressed VEGFR2 (Figure 2D). Hippocampal neuroprotection elicited by VEGF-A165b was dependent on downstream signaling of the VEGFR2 through the p44/42 MAPK pathway (Figure 3A), as it was also blocked by the MEK2 inhibitor, 10 μmol/L PD98059, as is also the case for VEGF-A165a.28 Neuroprotection was not affected by blockade of either p38 MAPK by SB203580 or phosphatidylinositol 3–kinase (PI3K) with LY294002 (Figure 3, A and B). Treatment of 50B11 neurons with VEGF-A165b significantly increased p44/42 MAPK phosphorylation (Figure 3C), which was blocked by the inhibitor PD98059 (Figure 3D).

Figure 2.

Figure 2

VEGF-A165b is protective against glutamate-induced hippocampal neuronal excitotoxicity. A: Pseudocolored image of cultured hippocampal neurons exposed to glutamic acid, with and without VEGF-A165b or galanin. Cells co-stained with Hoechst 33258 nuclear stain (blue) and Dead stain (red) are purple and represent approximately 75% of glutamic acid–treated neurons under control conditions. Also note effect of treatment with 10 nmol/L VEGF-A165b on excitotoxicity in neurons co-incubated with 3 mmol/L glutamate. B: VEGF-A165b has a concentration-dependent inhibitory effect on glutamatergic excitotoxicity in hippocampal neurons (P < 0.0001, n = 4, analysis of variance plus Newman-Keuls post hoc test, P < 0.01, ∗∗P < 0.001). C: Effect of VEGF-A receptor inhibitors on the hippocampal neuroprotective effect of VEGF-A165b [100 nmol/L PTK787 (nonspecific VEGFR2 antagonist), 10 nmol/L ZM323881 (VEGFR2-specific antagonist), and 100 nmol/L SU5416 (VEGFR1-specific antagonist); P < 0.05 compared with control, ∗∗P < 0.01, n = 4 per group]. D: VEGFR2 (red) expression in β3-tubulin–positive (green) neurons. Nuclei are stained blue. Higher-power individual color images (right panel) reveal membrane staining of the VEGFR2 outside tubulin.

Figure 3.

Figure 3

The effect of intracellular kinase–pharmacological inhibition on VEGF-A165b neuroprotection. A: Representative images of hippocampal neurons subjected to the live/dead cell viability stain after 24-hour treatment with 3 mmol/L l-glutamic acid in the presence of test compounds or respective vehicles. Neurons treated in culture media alone (neurobasal media) or l-glutamic acid plus 2.5 nmol/L VEGF-A165b with or without 10 μmol/L SB203580 (p38 MAPK inhibitor) or 15 μmol/L LY294002 (PI3K inhibitor) maintained neurite projections (arrows). In the presence of l-glutamic acid plus VEGF-A165b plus 10 μmol/L PD098059 (MEK1/2 inhibitor) neurons retracted neurites (arrowheads). Scale bar = 25 μm. B: The percentage of red-stained (dead) nuclei per total nuclei stained was calculated. More neurons died when treated with the MEK1/2 inhibitor plus VEGF-A165b (PD) than when treated with VEGF-A165b and vehicle (control). Neither p38 MAPK (SB) nor PI3K (LY) inhibition had any effect on the neuroprotection exerted by VEGF-A165b. Data are means ± SEM, n = 3 of 4 one-way analysis of variance plus Bonferroni post hoc comparison; increase of cell death in glutamic acid plus inhibitor/vehicle over media plus inhibitor/vehicle (white bars) compared with increase of cell death in glutamic acid plus VEGF-A165b and inhibitor/vehicle (black bars), P < 0.05, ∗∗P < 0.01. C: The level of phosphorylated p44/42 MAPK detected by immunoblotting was increased in cultured 50B11 neurons treated with 0.1 nmol/L rhVEGF165b compared with control (n = 3). D: PD98059 blocked phosphorylation of p42/p44 MAPK induced by VEGF165b in 50B11 neurons (P < 0.05, ∗∗P < 0.01 compared with untreated).

VEGF-A165b also exerted a neuroprotective action on retinal neurons in vivo (Figure 4). In retinal ischemia a reduction in retrograde transport of fluorescent tracer was seen (Figure 4A). This ischemic neuronal loss was reversed by prior intraocular VEGF-A165b treatment (Figure 4A). To determine whether this was due to apoptosis, retinae were stained for active caspase-3 (Figure 4B). Uninjected and HBSS-injected ischemic eyes revealed significant retinal neuronal loss through apoptosis, as assessed by Fluorogold labeling of live neurons (Figure 4, C and D) and caspase-3 staining respectively (Figure 4E). The reduction in neuronal retrograde transport was significantly reversed by VEGF-A165b (Figure 4, A, C, and D), when compared with the uninjected contralateral eye and reduced apoptosis in both RGCs and inner nuclear layer cells (Figure 4, B and D).

Figure 4.

Figure 4

VEGF-A165b protects retinal neurons from ischemia induced cell death in vivo. A: Pseudo-colored fluorescent images of retinal cells revealing the contralateral nonischemic retina, ischemic eye injected with HBSS, or ischemic eye injected with VEGF-A165b. B: Staining of retinae of HBSS- or VEGF-A165b–injected rats for activated caspase 3 (red) and nuclei (blue). C: Live RGC counts were significantly lower in ischemic eyes compared with nonischemic eyes. VEGF-A165b treatment resulted in more viable RGCs (n = 8 HBSS, n = 12 VEGF-A165b, P < 0.001, analysis of variance plus Bonferroni post hoc test). VEGF-A165b treatment increased the numbers of live Fluorogold-labeled retinal cells compared with the HBSS and control untreated eyes, which can be clearly seen in D. The ratio of RGCs per field in the ischemic and nonischemic eyes were compared. E: Neuroprotection by VEGF-A165b was mediated through an inhibition of apoptosis, as indicated by a reduction in active caspase-3 staining, in both the RGCs and inner nuclear layer (INL). Data are means ± SEM. ∗∗P < 0.01, ∗∗∗P < 0.001 compared with contralateral and †††P < 0.001 compared with HBSS.

VEGF-A165a has been found to be cytoprotective against injury induced by a variety of cellular insults to neurons.29 To determine whether neuroprotection was confined to central neurons or also affected peripheral neurons, cultured primary DRG neurons were investigated. The addition of 1 nmol/L VEGF-A165b significantly increased the length of neurite outgrowth during 24 hours from 41.7 ± 2.2 μm to 73.8 ± 20 μm (P < 0.05, U-test) but did not affect the number of DRG neurons in treated and control cultures, indicating a neurotrophic action. A total of 2.5 nmol/L VEGF-A165b significantly prevented an increase in activated caspase-3 in primary DRG neurons induced by 24 hours of treatment with the chemotherapeutic oxaliplatin. Treatment with oxaliplatin alone (5, 10, or 20 μg/mL) significantly increased the percentage of caspase-3–positive DRG neurons compared with untreated neurons, and VEGF-A165b prevented this (Figure 5, A and B).

Figure 5.

Figure 5

VEGF-A165b is cytoprotective for primary sensory neurons. A: Effect of VEGF-A165b in a model of chemotherapeutic-induced neurotoxicity. Primary adult rat DRG cultures were treated with increasing concentrations (0, 5, 10, and 20 μg/mL) of the chemotherapeutic oxaliplatin for 24 hours with or without 2.5 nmol/L VEGF-A165b (after 16 hours of pretreatment). The percentage of activated caspase-3–positive NeuN-positive cells was determined after immunofluorescence analysis. The percentage of neurons positive for activated caspase-3 was determined after treatment with 0 to 20 μg/mL of oxaliplatin for 24 hours with or without 2.5 nmol/L VEGF-A165b. VEGF-A165b treatment inhibited oxaliplatin-induced caspase-3 expression. Data are means ± SEM (n = 3). B: Representative images of NeuN-positive cells after treatment. NeuN-negative cells (only blue or blue and red) were not counted. Arrows signify activated caspase-3 detected both around and in neuronal nuclei. C: Primary DRG neurons were pretreated with concentrations of a neutralizing VEGF165b antibody for 20 hours before oxaliplatin treatment (0, 10 μg/mL; 24 hours). IgG content was equalized for the groups by addition of control mouse IgG. Pretreatment with anti-VEGF165b did not affect the proportion of live neurons without oxaliplatin treatment. The percentage of live neurons was significantly decreased by anti-VEGF165b after oxaliplatin treatment. D: Cell viability was measured by PrestoBlue absorbance from DRG neurons treated as above. Oxaliplatin treatment significantly decreased the viability without anti-VEGF165b pretreatment compared with control. VEGF165b pretreatment reduced the viability further still compared with control, which was significant from oxaliplatin treatment without anti-VEGF165b. Data are means ± SEM, n = 6, one-way analysis of variance plus Bonferroni. P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001, compared with no oxaliplatin plus IgG control; P < 0.05 compared with oxaliplatin IgG control.

To determine whether VEGF165b was acting as an endogenous neuroprotective factor for DRG, we treated cultured primary rat DRG cells with an antibody to VEGF165b with or without the toxic chemotherapeutic oxaliplatin. In the absence of oxaliplatin, neither anti-VEGF165b antibody (means ± SEM, 98% ± 14%, 103% ± 12%, and 95% ± 6.6% of control, respectively) nor anti–pan VEGF antibody (93% ± 10%, 98% ± 16%, and 95% ± 12%) had any effect at 1, 10, or 100 μg/mL, respectively. In contrast, when combined with oxaliplatin anti-VEGF165b treatment resulted in a significant increase in the proportion of dead cells after 24 hours compared with mouse IgG (Figure 5C). As a more sensitive measure we also used the PrestoBlue cell viability assay where absorption is proportional to mitochondrial metabolic activity and can be taken as a measure of cell viability, in which oxaliplatin also resulted in significant reduction in cell viability, which was further reduced by the addition of anti-VEGF165b (Figure 5D).

Staining of cultured DRG neurons for VEGFR2 revealed strong expression on the cell membrane (Figure 6A). To investigate the receptor mediating the neuroprotective response to VEGF-A165b, DRG cultures were treated with 10 nmol/L specific VEGFR2 inhibitor ZM323881 or vehicle during oxaliplatin treatment and stained for NeuN and activated caspase-3 (Figure 6B). Treatment with 20 μg/mL of oxaliplatin, either alone or with ZM323881, induced a significant increase in the percentage of activated caspase-3–positive, NeuN-positive cells compared with media (51.4% ± 3.0% and 55.7% ± 1.9% versus 37.8% ± 1.3% and 42.5% ± 2.6%, respectively). Concurrent treatment with 2.5 nmol/L recombinant human VEGF-A165b without ZM323881 significantly reduced the percentage of activated caspase-3–positive, NeuN-positive cells compared with vehicle (35.4% ± 3.1%) (Figure 6B), and this effect was blocked by treatment with the VEGFR2 inhibitor ZM323881 (54.3% ± 3.3%). To further rule out a role for VEGFR1, cells were treated with placenta growth factor (PlGF), which activates only VEGFR1. This neither affected the oxaliplatin-induced cell death nor prevented its amelioration by VEGF165b (Figure 6B). As an additional control to determine whether VEGF165b was acting through VEGFR2, cells were treated with oxaliplatin and VEGF165b in the presence of either mouse IgG as a control or the VEGFR2 neutralizing antibody DC101. Although VEGFR2 blockade by itself did not increase neuronal caspase activity or affect the oxaliplatin-induced effect, it prevented the VEGF165b-mediated cytoprotection (Figure 6C).

Figure 6. (VSports注册入口)

Figure 6

Neuroprotection is mediated through VEGFR2 not VEGFR1. A: Primary cultured DRG were stained for βIII tubulin (green), VEGFR2 (red), and Hoechst (blue). βIII tubulin–positive cells were also positive for VEGFR2, with the receptor detected in the soma (white arrows) and along cellular projections (arrowheads). Some VEGFR2 detection did not colocalize to βIII tubulin (cyan arrows). The matched-species IgG-negative control confirms the detection of VEGFR2 expression. B: Treatment of DRG cultures with oxaliplatin plus 2.5 nmol/L VEGF-A165b and 10 nmol/L VEGFR2 inhibitor ZM323881 blocked the neuroprotection exerted by VEGF-A165b in the presence of ZM vehicle. Treatment of DRG cultures with 10 nmol/L PlGF, a competitor for VEGFR1, did not affect rhVEGF165b neuroprotection against oxaliplatin. C: Treatment of DRG cultures with oxaliplatin plus 2.5 nmol/L VEGF-A165b and 10 μg/mL of VEGFR2 blocking antibody DC101 blocked the neuroprotection exerted by VEGF-A165b in the presence of rat IgG. Data are means ± SEM, (n = 3), one-way analysis of variance plus Newman Keuls. ∗∗P < 0.01, ∗∗∗P < 0.001, compared with no oxaliplatin; P < 0.05, compared with respective concentration matched vehicle; and ns are compared with respective oxaliplatin without VEGF165b; ‡‡P < 0.01, ‡‡‡P < 0.001, compared with respective oxaliplatin without VEGF165b; §P < 0.05, compared with oxaliplatin and VEGF165b. Scale bar = 50 μm.

To determine whether systemic administration of VEGF-A165b could be neuroprotective, we used a mouse model of peripheral traumatic nerve injury that results in activation of injury-response genes in DRG neurons, such as galanin,30 and ATF3 (Figure 7, A and B). Biweekly treatment with rhVEGF-A165b reduced the intensity of neuronal ATF3 expression in L3/4 DRG compared with sham surgery controls after 10 days (Figure 7C).

Figure 7.

Figure 7

VEGF-A165b is neuroprotective for DRG neurons in vivo. PSNI was performed on anesthetized C57BL/6 mice, test compounds were administered biweekly by i.p. injection after surgery, and L4 DRG was harvested 14 days later. For each DRG a complete cross section profile was analyzed for ATF3 immunofluorescence intensity. A: Low-power representative images of ATF3 immunofluorescence intensities from whole ipsilateral L4 DRG sections and a species- and concentration-matched IgG negative control. B: Mean pixel intensity was calculated for each DRG profile and expressed relative to the mean pixel intensity for L4 DRG in the sham surgery group. PSNI increased ATF3 staining, which was blocked by VEGF-A165b treatment. Results are means ± SEM, n = 3 per group. Statistical analysis was performed with Kruskal-Wallis and Dunn's multiple comparison test: test group ipsilateral DRG intensity versus ipsilateral DRG in the sham surgery group. P < 0.05. C: DRG ipsilateral to the injury was stained for ATF3 and the neuronal marker NeuN to identify nerve injury–induced neuronal ATF3 expression. Scale bar = 500 μm.

Discussion

We report that, like VEGF-A165a, the splice isoform VEGF-A165b is neuroprotective for central and peripheral neurons, in vitro and in vivo, and mediates this neuroprotective effect through VEGFR2 and MEK1/2 activation. In addition to its neuroprotective effects, VEGF-A165b also has neurotrophic actions on neurons.

VEGF-A165a has been found to play a key role in neuronal protection, in addition to its actions on the vasculature, directly protecting motoneurons under conditions of hypoxia, oxidative stress, and serum deprivation.31 Disturbance of VEGF-A expression contributes to the development of amyotrophic lateral sclerosis in humans.29 VEGF-A and its receptors are also expressed in central and peripheral nervous system support cells, such as astrocytes and Schwann cells, thereby also contributing to neuronal survival and growth.10,32

In the central nervous system, VEGF-A165a protects hippocampal, cortical, and cerebellar granule neurons against numerous insults33–36 through VEGFR2, signaling through activation of multiple intracellular pathways, including phospholipase C, PI3K, and MEK1/2,32 whereas effects on supporting cells, such as Schwann cells, and astrocytes, are generally mediated through VEGFR1.10 In contrast, our data indicate that neuroprotection by VEGF-A165b in hippocampal neurons, although also mediated through VEGFR2, involves MEK1/2 but not either PI3K or p38 MAPK. The VEGF-Axxx bisoforms compete for and inhibit VEGF-Axxxa binding at VEGFR2,5,7,37 but the VEGF-Axxxb isoforms are not simply competitive inhibitors of the VEGFR2 because binding of VEGF-A165b results in differential tyrosine residue phosphorylation of VEGFR2.24 Neuroprotective and neurotrophic actions by VEGF-A165a may also involve the VEGF co-receptor neuropilin-1 (NP-1)32; VEGF-A165b binds weakly to NP-1 and does not require NP-1 binding to phosphorylate and activate VEGFR2.24 Our data support the conclusion that the differential VEGFR2 phosphorylation and MEK1/2 activation exerted by VEGF-A165b is sufficient to protect central and peripheral neurons, without NP-1 binding. Our data on both central nervous system and peripheral sensory neurons indicate that the neuroprotective mechanism of VEGF-A165b is like that ascribed to VEGF-A165a in that neuroprotection occurs through prevention of caspase-3 induction.32

In the eye, endogenous VEGF-A is a survival factor for RGCs, protecting against ischemia-reperfusion injury38 and preventing neuronal apoptosis without the necessity for NP-1 binding because VEGF-A120a (which lacks the NP-1 binding domain39) also exerted neuroprotective effects. Our findings indicate that exogenous VEGF-A165b can reduce both RGC and inner nuclear cell loss through VEGFR2 activation. The loss of neuronal retrograde transport is seen clinically in diabetic retinopathy as cotton wool spots under fundus examination, and the loss of ganglion cells contributes to vision loss in glaucoma.40 We have previously found that VEGF-A165b is cytoprotective for endothelial and retinal pigmented epithelial cells,41 whereas it is antiangiogenic in the eye.42 Prevention of the ischemia-induced damage to neurons in the retina by VEGF-A165b would therefore also be a substantial advantage in therapeutic approaches for diabetic retinopathy and glaucoma.

In hippocampal and peripheral sensory cultured neurons, VEGF-A165b also enhances neurite outgrowth, demonstrating that VEGF-A165b exerts neurotrophic effects, also through VEGFR2. In peripheral sensory and autonomic neurons, VEGF-A165a enhances neuronal neurite outgrowth in culture.11,32,43 VEGFR2 is expressed in both the central and peripheral nervous systems but often in different neuronal populations from VEGF-A proteins.44,45 These different distributions suggest that VEGF-A isoforms exert paracrine actions on neurons through which neuroprotective and neurotrophic effects may be mediated. We demonstrate that VEGF-A165b is a major VEGF-A splice variant commonly found in central and peripheral neurons, where it is well placed to exert such paracrine effects. VEGF-A165b is also expressed in human skin, prostate, and kidney, among other tissues,5,46 where it could also exert paracrine effects on peripheral sensory neurons.

VEGF-A165b has clear inhibitory effects on tumor growth5,14,47 but does not result in hypotension, angiogenesis, or proteinuria.5,6,48 Our data indicate that VEGF-A165b has neuroprotective effects similar to those ascribed to VEGF-A165a, on central and peripheral neurons. VEGF-A–dependent neovascularization is key in the pathophysiology of many conditions, and anti–VEGF-A therapies have entered clinical practice in oncology9 and ophthalmology.8 After preclinical studies of VEGF-A administration in neurodegenerative disease,11,29,49–51 VEGF-A supplementation is now under trial for treatment of neuronal degenerative diseases, for instance in diabetic neuropathy.52 Although effective in some patients, there has to be some concern about the safety profile of these strategies in relation to the potential compromise of nonendothelial tissue and cell function in which VEGF-A has been found to have protective properties, particularly neurons10 and podocytes.53 We report that VEGF-Axxxb can exert similar neuroprotective and neurotropic effects with VEGF-Axxxa both centrally (eg, hippocampal neurons in culture, and retinal neurons in vivo) and peripherally.

With the requirement that therapy for pathological conditions in which VEGF-A has been implicated should not adversely affect the function of the normal vasculature or other cell types, we suggest that VEGF-A165b may be an alternative therapeutic agent in neurodegenerative conditions with fewer adverse vascular effects.

Footnotes

Supported by Wellcome Trust grants 079736 and 058083 and the Richard Bright VEGF-A Research Trust.

N.B.-L. and J.H. contributed equally to this work.

D.O.B. and L.F.D. contributed equally to this work as senior authors.

Current address of J.H., Department of Ophthalmology, Schepens Eye Research Institute, Boston, MA; of D.O.B., Division of Preclinical Oncology, University of Nottingham Queen's Medical Centre, Nottingham, UK.

"VSports手机版" Contributor Information

David O. Bates, Email: david.bates@qiuluzeuv.cn.

Lucy F. Donaldson, Email: lucy.donaldson@qiuluzeuv.cn.

References

  • 1.Kovacs Z., Ikezaki K., Samoto K., Inamura T., Fukui M. VEGF and flt: expression time kinetics in rat brain infarct. Stroke. 1996;27:1865–1872. doi: 10.1161/01.str.27.10.1865. [DOI] [PubMed] [Google Scholar]
  • 2.Hobson M.I., Green C.J., Terenghi G. VEGF enhances intraneural angiogenesis and improves nerve regeneration after axotomy. J Anat. 2000;197(pt 4):591–605. doi: 10.1046/j.1469-7580.2000.19740591.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Harper S.J., Bates D.O. VEGF-A splicing: the key to anti-angiogenic therapeutics? Nat Rev Cancer. 2008;8:880–887. doi: 10.1038/nrc2505. ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bates D.O., Cui T.G., Doughty J.M., Winkler M., Sugiono M., Shields J.D., Peat D., Gillatt D., Harper S.J. VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, is down-regulated in renal cell carcinoma. Cancer Res. 2002;62:4123–4131. [PubMed] [Google Scholar]
  • 5.Woolard J., Wang W.Y., Bevan H.S., Qiu Y., Morbidelli L., Pritchard-Jones R.O., Cui T.G., Sugiono M., Waine E., Perrin R., Foster R., Digby-Bell J., Shields J.D., Whittles C.E., Mushens R.E., Gillatt D.A., Ziche M., Harper S.J., Bates D.O. VEGF165b, an inhibitory vascular endothelial growth factor splice variant: mechanism of action, in vivo effect on angiogenesis and endogenous protein expression. Cancer Res. 2004;64:7822–7835. doi: 10.1158/0008-5472.CAN-04-0934. [DOI] [PubMed] [Google Scholar]
  • 6.Qiu Y., Ferguson J., Oltean S., Neal C.R., Kaura A., Bevan H., Wood E., Sage L.M., Lanati S., Nowak D.G., Salmon A.H., Bates D., Harper S.J. Overexpression of VEGF165b in podocytes reduces glomerular permeability. J Am Soc Nephrol. 2010;21:1498–1509. doi: 10.1681/ASN.2009060617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cebe Suarez S., Pieren M., Cariolato L., Arn S., Hoffmann U., Bogucki A., Manlius C., Wood J., Ballmer-Hofer K. A VEGF-A splice variant defective for heparan sulfate and neuropilin-1 binding shows attenuated signaling through VEGFR-2. Cell Mol Life Sci. 2006;63:2067–2077. doi: 10.1007/s00018-006-6254-9. [DOI (V体育官网入口)] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rosenfeld P.J., Brown D.M., Heier J.S., Boyer D.S., Kaiser P.K., Chung C.Y., Kim R.Y. Ranibizumab for neovascular age-related macular degeneration. N Engl J Med. 2006;355:1419–1431. doi: 10.1056/NEJMoa054481. [DOI] [PubMed] [Google Scholar]
  • 9.Hurwitz H., Fehrenbacher L., Novotny W., Cartwright T., Hainsworth J., Heim W., Berlin J., Baron A., Griffing S., Holmgren E., Ferrara N., Fyfe G., Rogers B., Ross R., Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691. ["V体育ios版" DOI] [PubMed] [Google Scholar]
  • 10.Storkebaum E., Lambrechts D., Carmeliet P. VEGF: once regarded as a specific angiogenic factor, now implicated in neuroprotection. Bioessays. 2004;26:943–954. doi: 10.1002/bies.20092. [DOI] [PubMed] [Google Scholar]
  • 11.Sondell M., Lundborg G., Kanje M. Vascular endothelial growth factor has neurotrophic activity and stimulates axonal outgrowth, enhancing cell survival and Schwann cell proliferation in the peripheral nervous system. J Neurosci. 1999;19:5731–5740. doi: 10.1523/JNEUROSCI.19-14-05731.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Glass C.A., Harper S.J., Bates D.O. The anti-angiogenic VEGF isoform VEGF165b transiently increases hydraulic conductivity, probably through VEGF receptor 1 in vivo. J Physiol. 2006;572:243–257. doi: 10.1113/jphysiol.2005.103127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bevan H.S., van den Akker N.M., Qiu Y., Polman J.A., Foster R.R., Yem J., Nishikawa A., Satchell S.C., Harper S.J., Gittenberger-de Groot A.C., Bates D.O. The alternatively spliced anti-angiogenic family of VEGF isoforms VEGFxxxb in human kidney development. Nephron Physiol. 2008;110:57–67. doi: 10.1159/000177614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Varey A.H., Rennel E.S., Qiu Y., Bevan H.S., Perrin R.M., Raffy S., Dixon A.R., Paraskeva C., Zaccheo O., Hassan A.B., Harper S.J., Bates D.O. VEGF 165 b, an antiangiogenic VEGF-A isoform, binds and inhibits bevacizumab treatment in experimental colorectal carcinoma: balance of pro- and antiangiogenic VEGF-A isoforms has implications for therapy. Br J Cancer. 2008;98:1366–1379. doi: 10.1038/sj.bjc.6604308. [DOI (V体育ios版)] [PMC free article] [PubMed] [Google Scholar]
  • 15.Harris S., Craze M., Newton J., Fisher M., Shima D.T., Tozer G.M., Kanthou C. Do anti-angiogenic VEGF (VEGFxxxb) isoforms exist? a cautionary tale. PLoS ONE. 2012;7:e35231. doi: 10.1371/journal.pone.0035231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Elliott-Hunt C.R., Marsh B., Bacon A., Pope R., Vanderplank P., Wynick D. Galanin acts as a neuroprotective factor to the hippocampus. Proc Natl Acad Sci U S A. 2004;101:5105–5110. doi: 10.1073/pnas.0304823101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen W., Mi R., Haughey N., Oz M., Hoke A. Immortalization and characterization of a nociceptive dorsal root ganglion sensory neuronal line. J Peripher Nerv Syst. 2007;12:121–130. doi: 10.1111/j.1529-8027.2007.00131.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Holmes F.E., Mahoney S., King V.R., Bacon A., Kerr N.C., Pachnis V., Curtis R., Priestley J.V., Wynick D. Targeted disruption of the galanin gene reduces the number of sensory neurons and their regenerative capacity. Proc Natl Acad Sci U S A. 2000;97:11563–11568. doi: 10.1073/pnas.210221897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Selles-Navarro I., Villegas-Perez M.P., Salvador-Silva M., Ruiz-Gomez J.M., Vidal-Sanz M. Retinal ganglion cell death after different transient periods of pressure-induced ischemia and survival intervals: a quantitative in vivo study. Invest Ophthalmol Vis Sci. 1996;37:2002–2014. [PubMed] [Google Scholar]
  • 20.Jehle T., Wingert K., Dimitriu C., Meschede W., Lasseck J., Bach M., Lagreze W.A. Quantification of ischemic damage in the rat retina: a comparative study using evoked potentials, electroretinography, and histology. Invest Ophthalmol Vis Sci. 2008;49:1056–1064. doi: 10.1167/iovs.07-1050. [DOI] [PubMed] [Google Scholar]
  • 21.Ahmed A.K., Sugioka K., Dong K., Yamadori T. A study of double-labeled retinal ganglion cells from the superior colliculus in the developing albino rat. Brain Res Dev Brain Res. 1995;85:71–79. doi: 10.1016/0165-3806(94)00191-2. [DOI] [PubMed] [Google Scholar]
  • 22.Lam T.T., Abler A.S., Tso M.O. Apoptosis and caspases after ischemia-reperfusion injury in rat retina. Invest Ophthalmol Vis Sci. 1999;40:967–975. [PubMed] [Google Scholar]
  • 23.Walczak J.S., Pichette V., Leblond F., Desbiens K., Beaulieu P. Behavioral, pharmacological and molecular characterization of the saphenous nerve partial ligation: a new model of neuropathic pain. Neuroscience. 2005;132:1093–1102. doi: 10.1016/j.neuroscience.2005.02.010. [DOI] [PubMed] [Google Scholar]
  • 24.Kawamura H., Li X., Harper S.J., Bates D.O., Claesson-Welsh L. Vascular endothelial growth factor (VEGF)-A165b is a weak in vitro agonist for VEGF receptor-2 due to lack of coreceptor binding and deficient regulation of kinase activity. Cancer Res. 2008;68:4683–4692. doi: 10.1158/0008-5472.CAN-07-6577. [DOI] [PubMed] [Google Scholar]
  • 25.Matsuzaki H., Tamatani M., Yamaguchi A., Namikawa K., Kiyama H., Vitek M.P., Mitsuda N., Tohyama M. Vascular endothelial growth factor rescues hippocampal neurons from glutamate-induced toxicity: signal transduction cascades. FASEB J. 2001;15:1218–1220. [PubMed (VSports app下载)] [Google Scholar]
  • 26.Wood J.M., Bold G., Buchdunger E., Cozens R., Ferrari S., Frei J., Hofmann F., Mestan J., Mett H., O'Reilly T., Persohn E., Rosel J., Schnell C., Stover D., Theuer A., Towbin H., Wenger F., Woods-Cook K., Menrad A., Siemeister G., Schirner M., Thierauch K.H., Schneider M.R., Drevs J., Martiny-Baron G., Totzke F. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration. Cancer Res. 2000;60:2178–2189. [PubMed] [Google Scholar]
  • 27.Whittles C.E., Pocock T.M., Wedge S.R., Kendrew J., Hennequin L.F., Harper S.J., Bates D.O. ZM323881, a novel inhibitor of vascular endothelial growth factor-receptor-2 tyrosine kinase activity. Microcirculation. 2002;9:513–522. doi: 10.1038/sj.mn.7800164. [DOI] [PubMed] [Google Scholar]
  • 28.Gomes E., Papa L., Hao T., Rockwell P. The VEGFR2 and PKA pathways converge at MEK/ERK1/2 to promote survival in serum deprived neuronal cells. Mol Cell Biochem. 2007;305:179–190. doi: 10.1007/s11010-007-9542-2. [DOI] [PubMed] [Google Scholar]
  • 29.Lambrechts D., Storkebaum E., Morimoto M., Del-Favero J., Desmet F., Marklund S.L., Wyns S., Thijs V., Andersson J., van Marion I., Al-Chalabi A., Bornes S., Musson R., Hansen V., Beckman L., Adolfsson R., Pall H.S., Prats H., Vermeire S., Rutgeerts P., Katayama S., Awata T., Leigh N., Lang-Lazdunski L., Dewerchin M., Shaw C., Moons L., Vlietinck R., Morrison K.E., Robberecht W., Van Broeckhoven C., Collen D., Andersen P.M., Carmeliet P. VEGF is a modifier of amyotrophic lateral sclerosis in mice and humans and protects motoneurons against ischemic death. Nat Genet. 2003;34:383–394. doi: 10.1038/ng1211. [DOI] [PubMed] [Google Scholar]
  • 30.Hulse R., Wynick D., Donaldson L.F. Characterization of a novel neuropathic pain model in mice. Neuroreport. 2008;19:825–829. doi: 10.1097/WNR.0b013e328300ee0a. [V体育2025版 - DOI] [PubMed] [Google Scholar]
  • 31.Oosthuyse B., Moons L., Storkebaum E., Beck H., Nuyens D., Brusselmans K., Van Dorpe J., Hellings P., Gorselink M., Heymans S., Theilmeier G., Dewerchin M., Laudenbach V., Vermylen P., Raat H., Acker T., Vleminckx V., Van Den Bosch L., Cashman N., Fujisawa H., Drost M.R., Sciot R., Bruyninckx F., Hicklin D.J., Ince C., Gressens P., Lupu F., Plate K.H., Robberecht W., Herbert J.M., Collen D., Carmeliet P. Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat Genet. 2001;28:131–138. doi: 10.1038/88842. [DOI (VSports注册入口)] [PubMed] [Google Scholar]
  • 32.Zachary I. Neuroprotective role of vascular endothelial growth factor: signalling mechanisms, biological function, and therapeutic potential. Neurosignals. 2005;14:207–221. doi: 10.1159/000088637. [DOI] [PubMed] [Google Scholar]
  • 33.Jin K.L., Mao X.O., Greenberg D.A. Vascular endothelial growth factor rescues HN33 neural cells from death induced by serum withdrawal. J Mol Neurosci. 2000;14:197–203. doi: 10.1385/JMN:14:3:197. [DOI (VSports最新版本)] [PubMed] [Google Scholar]
  • 34.Jin K.L., Mao X.O., Greenberg D.A. Vascular endothelial growth factor: direct neuroprotective effect in in vitro ischemia. Proc Natl Acad Sci U S A. 2000;97:10242–10247. doi: 10.1073/pnas.97.18.10242. [DOI (V体育安卓版)] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nicoletti J.N., Shah S.K., McCloskey D.P., Goodman J.H., Elkady A., Atassi H., Hylton D., Rudge J.S., Scharfman H.E., Croll S.D. Vascular endothelial growth factor is up-regulated after status epilepticus and protects against seizure-induced neuronal loss in hippocampus. Neuroscience. 2008;151:232–241. doi: 10.1061/j.neuroscience.2007.09.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rosenstein J.M., Mani N., Khaibullina A., Krum J.M. Neurotrophic effects of vascular endothelial growth factor on organotypic cortical explants and primary cortical neurons. J Neurosci. 2003;23:11036–11044. doi: 10.1523/JNEUROSCI.23-35-11036.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kawamura H., Li X., Goishi K., van Meeteren L.A., Jakobsson L., Cebe-Suarez S., Shimizu A., Edholm D., Ballmer-Hofer K., Kjellen L., Klagsbrun M., Claesson-Welsh L. Neuropilin-1 in regulation of VEGF-induced activation of p38MAPK and endothelial cell organization. Blood. 2008;112(9):3638–3640. doi: 10.1182/blood-2007-12-125856. [DOI (V体育2025版)] [PMC free article] [PubMed] [Google Scholar]
  • 38.Nishijima K., Ng Y.S., Zhong L., Bradley J., Schubert W., Jo N., Akita J., Samuelsson S.J., Robinson G.S., Adamis A.P., Shima D.T. Vascular endothelial growth factor-A is a survival factor for retinal neurons and a critical neuroprotectant during the adaptive response to ischemic injury. Am J Pathol. 2007;171:53–67. doi: 10.2353/ajpath.2007.061237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kawamura H., Li X., Goishi K., van Meeteren L.A., Jakobsson L., Cebe-Suarez S., Shimizu A., Edholm D., Ballmer-Hofer K., Kjellen L., Klagsbrun M., Claesson-Welsh L. Neuropilin-1 in regulation of VEGF-induced activation of p38MAPK and endothelial cell organization. Blood. 2008;112:3638–3649. doi: 10.1182/blood-2007-12-125856. [DOI (V体育安卓版)] [PMC free article] [PubMed] [Google Scholar]
  • 40.Almasieh M., Wilson A.M., Morquette B., Cueva Vargas J.L., Di Polo A. The molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res. 2012;31:152–181. doi: 10.1016/j.preteyeres.2011.11.002. [DOI] [PubMed] [Google Scholar]
  • 41.Magnussen A.L., Rennel E.S., Hua J., Bevan H.S., Beazley Long N., Lehrling C., Gammons M., Floege J., Harper S.J., Agostini H.T., Bates D.O., Churchill A.J. VEGF-A165b is cytoprotective and antiangiogenic in the retina. Invest Ophthalmol Vis Sci. 2010;51:4273–4281. doi: 10.1167/iovs.09-4296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hua J., Spee C., Kase S., Rennel E.S., Magnussen A.L., Qiu Y., Varey A., Dhayade S., Churchill A.J., Harper S.J., Bates D.O., Hinton D.R. Recombinant human VEGF165b inhibits experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2010;51:4282–4288. doi: 10.1167/iovs.09-4360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lin G., Shindel A.W., Fandel T.M., Bella A.J., Lin C.S., Lue T.F. Neurotrophic effects of brain-derived neurotrophic factor and vascular endothelial growth factor in major pelvic ganglia of young and aged rats. BJU Int. 2010;105:114–120. doi: 10.1111/j.1464-410X.2009.08647.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sondell M., Kanje M. Postnatal expression of VEGF and its receptor flk-1 in peripheral ganglia. Neuroreport. 2001;12:105–108. doi: 10.1097/00001756-200101220-00028. [DOI] [PubMed] [Google Scholar]
  • 45.Licht T., Goshen I., Avital A., Kreisel T., Zubedat S., Eavri R., Segal M., Yirmiya R., Keshet E. Reversible modulations of neuronal plasticity by VEGF. Proc Natl Acad Sci U S A. 2011;108:5081–5086. doi: 10.1073/pnas.1007640108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pritchard-Jones R.O., Dunn D.B., Qiu Y., Varey A.H., Orlando A., Rigby H., Harper S.J., Bates D.O. Expression of VEGF(xxx)b, the inhibitory isoforms of VEGF, in malignant melanoma. Br J Cancer. 2007;97:223–230. doi: 10.1038/sj.bjc.6603839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rennel E., Waine E., Guan H., Schuler Y., Leenders W., Woolard J., Sugiono M., Gillatt D., Kleinerman E., Bates D., Harper S. The endogenous anti-angiogenic VEGF isoform. VEGF165b inhibits human tumour growth in mice. Br J Cancer. 2008;98:1250–1257. doi: 10.1038/sj.bjc.6604309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Rennel E.S., Hamdollah-Zadeh M.A., Wheatley E.R., Magnussen A., Schuler Y., Kelly S.P., Finucane C., Ellison D., Cebe-Suarez S., Ballmer-Hofer K., Mather S., Stewart L., Bates D.O., Harper S.J. Recombinant human VEGF165b protein is an effective anti-cancer agent in mice. Eur J Cancer. 2008;44:1883–1894. doi: 10.1016/j.ejca.2008.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Schratzberger P., Schratzberger G., Silver M., Curry C., Kearney M., Magner M., Alroy J., Adelman L.S., Weinberg D.H., Ropper A.H., Isner J.M. Favorable effect of VEGF gene transfer on ischemic peripheral neuropathy. Nat Med. 2000;6:405–413. doi: 10.1038/74664. [DOI] [PubMed] [Google Scholar]
  • 50.Schratzberger P., Walter D.H., Rittig K., Bahlmann F.H., Pola R., Curry C., Silver M., Krainin J.G., Weinberg D.H., Ropper A.H., Isner J.M. Reversal of experimental diabetic neuropathy by VEGF gene transfer. J Clin Invest. 2001;107:1083–1092. doi: 10.1172/JCI12188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yasuhara T., Shingo T., Muraoka K., Kameda M., Agari T., Wen Ji Y., Hayase H., Hamada H., Borlongan C.V., Date I. Neurorescue effects of VEGF on a rat model of Parkinson's disease. Brain Res. 2005;1053:10–18. doi: 10.1016/j.brainres.2005.05.027. ["V体育ios版" DOI] [PubMed] [Google Scholar]
  • 52.Ropper A.H., Gorson K.C., Gooch C.L., Weinberg D.H., Pieczek A., Ware J.H., Kershen J., Rogers A., Simovic D., Schratzberger P., Kirchmair R., Losordo D. Vascular endothelial growth factor gene transfer for diabetic polyneuropathy: a randomized, double-blinded trial. Ann Neurol. 2009;65:386–393. doi: 10.1002/ana.21675. [V体育ios版 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Foster R.R., Hole R., Anderson K., Satchell S.C., Coward R.J., Mathieson P.W., Gillatt D.A., Saleem M.A., Bates D.O., Harper S.J. Functional evidence that vascular endothelial growth factor may act as an autocrine factor on human podocytes. Am J Physiol Renal Physiol. 2003;284:F1263–F1273. doi: 10.1152/ajprenal.00276.2002. [DOI (V体育平台登录)] [PubMed] [Google Scholar]

Articles from The American Journal of Pathology are provided here courtesy of American Society for Investigative Pathology

RESOURCES (VSports app下载)