Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Eur Neuropsychopharmacol ; 31: 145-151, 2020 02.
Article in English | MEDLINE | ID: mdl-31902568

ABSTRACT

Vascular endothelial growth factor (VEGF, also known as VEGF-A) is a pleiotropic factor which is expressed by neurons, astrocytes and perivascular macrophages, as well as endothelial cells, in the brain. Recently, VEGF signaling has been implicated in learning and memory, and several clinical and preclinical studies demonstrate that VEGF inhibitors induce cognitive impairment. However, the role of endogenous neuronal VEGF signaling in recognition memory remains unclear. Recently, we have developed mice with forebrain excitatory neuron-specific deletion of VEGF or its receptor, fetal liver kinase 1 (Flk-1) by crossing Camk2a-Cre mice with Vegfaflox/flox and Flk-1flox/flox mice, respectively. Using these conditional knockout mice, the present study addressed the influence of forebrain excitatory neuron-specific deletion of VEGF or Flk-1 on recognition memory in the novel object recognition test. The results show that both short-term (2 h) and long-term (24 h) recognition memory are impaired by neuron-specific deletion of either Flk-1 or VEGF. These findings indicate the physiological importance of endogenous neuronal VEGF-Flk-1 signaling in recognition memory. In addition, the current results also suggest that the impairment of neuronal VEGF-Flk-1 signaling can be a cause of anti-VEGF chemotherapy-induced cognitive impairment.


Subject(s)
Memory Disorders/metabolism , Memory/physiology , Neurons/metabolism , Recognition, Psychology/physiology , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor Receptor-2/deficiency , Animals , Male , Memory Disorders/genetics , Mice , Mice, Knockout , Mice, Transgenic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
2.
J Appl Physiol (1985) ; 127(5): 1360-1369, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31487223

ABSTRACT

Diaphragm dysfunction accompanies cardiopulmonary disease and impaired oxygen delivery. Vascular endothelial growth factor (VEGF) regulates oxygen delivery through angiogenesis, capillary maintenance, and contraction-induced perfusion. We hypothesized that myofiber-specific VEGF deficiency contributes to diaphragm weakness and fatigability. Diaphragm protein expression, capillarity and fiber morphology, mitochondrial respiration and hydrogen peroxide (H2O2) generation, and contractile function were compared between adult mice with conditional gene ablation of skeletal myofiber VEGF (SkmVEGF-/-; n = 12) and littermate controls (n = 13). Diaphragm VEGF protein was ~50% lower in SkmVEGF-/- than littermate controls (1.45 ± 0.65 vs. 3.04 ± 1.41 pg/total protein; P = 0.001). This was accompanied by an ~15% impairment in maximal isometric specific force (F[1,23] = 15.01, P = 0.001) and a trend for improved fatigue resistance (P = 0.053). Mean fiber cross-sectional area and type I fiber cross-sectional area were lower in SkmVEGF-/- by ~40% and ~25% (P < 0.05). Capillary-to-fiber ratio was also lower in SkmVEGF-/- by ~40% (P < 0.05), and thus capillary density was not different. Sarcomeric actin expression was ~30% lower in SkmVEGF-/- (P < 0.05), whereas myosin heavy chain and MAFbx were similar (measured via immunoblot). Mitochondrial respiration, citrate synthase activity, PGC-1α, and hypoxia-inducible factor 1α were not different in SkmVEGF-/- (P > 0.05). However, mitochondrial-derived reactive oxygen species (ROS) flux was lower in SkmVEGF-/- (P = 0.0003). In conclusion, myofiber-specific VEGF gene deletion resulted in a lower capillary-to-fiber ratio, type I fiber atrophy, actin loss, and contractile dysfunction in the diaphragm. In contrast, mitochondrial respiratory function was preserved alongside lower ROS generation, which may play a compensatory role to preserve fatigue resistance in the diaphragm.NEW & NOTEWORTHY Diaphragm weakness is a hallmark of diseases in which oxygen delivery is compromised. Vascular endothelial growth factor (VEGF) modulates muscle perfusion; however, it remains unclear whether VEGF deficiency contributes to the onset of diaphragm dysfunction. Conditional skeletal myofiber VEGF gene ablation impaired diaphragm contractile function and resulted in type I fiber atrophy, a lower number of capillaries per fiber, and contractile protein content. Mitochondrial function was similar and reactive oxygen species flux was lower. Diaphragm VEGF deficiency may contribute to the onset of respiratory muscle weakness.


Subject(s)
Diaphragm/metabolism , Diaphragm/physiopathology , Mitochondria/metabolism , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , Vascular Endothelial Growth Factor A/deficiency , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Muscle Fibers, Skeletal/physiology , Organ Culture Techniques , Vascular Endothelial Growth Factor A/genetics
3.
Artif Cells Nanomed Biotechnol ; 47(1): 1374-1383, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30977418

ABSTRACT

Non-viral nanocarrier affords a platform for drug and siRNA combination, the focus of which is to load drug and siRNA into a single carrier, allowing for co-delivery and a synergistic effect at tumour site. In our previous study, pH-sensitive carboxymethyl chitosan-modified liposomes (CMCS-SiSf-CL) were assembled for sorafenib (Sf) and Cy3-siRNA co-loaded. The present study evaluated in vitro and in vivo co-delivery of the co-loaded liposomes. Further, in vitro inhibiting hepatocellular carcinoma of the pH-sensitive sorafenib (Sf) and VEGF-siRNA co-loaded liposomes was discussed. The experimental results demonstrated co-delivery and penetration into 2-dimensional (2D) cultured HepG2 cells, 3-dimensional (3D) cultured HepG2 tumour spheroids and tumour regions of H22 tumour-bearing mice. Compared with free siRNA and single loaded carrier, co-delivery liposomes exhibited enhanced VEGF downregulating effect, inducing cell early apoptosis. Therefore, the CMCS-SiSf-CL delivery system can lay the foundation for the co-delivery systems development and provide new area for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Sorafenib/administration & dosage , Sorafenib/pharmacology , Vascular Endothelial Growth Factor A/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Silencing , Humans , Hydrogen-Ion Concentration , Liposomes , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice , Sorafenib/therapeutic use , Vascular Endothelial Growth Factor A/deficiency
4.
Mol Vis ; 25: 194-203, 2019.
Article in English | MEDLINE | ID: mdl-30996588

ABSTRACT

Purpose: To evaluate the reliability and reproducibility of a rodent choroidal neovascularization (CNV) model by subretinal injection of polyethylene glycol (PEG). Methods: C57BL/6 mice were injected subretinally with 2 µl PBS (Gibco, Invitrogen, Paisley, UK; n=14) or PEG (1 mg; n=18). Animals were sacrificed at either 0, 5, 14 or 21 days. Eyes were embedded in paraffin wax and serial sections were stained with haematoxylin and eosin or Fontana-Masson or immunostained for cytokeratin 8/18, isolectin B4 (IB4), vascular endothelial growth factor (VEGF) and von Willebrand factor (vWF). Results: Both the PBS and PEG groups had retinal degeneration and retinal pigment epithelium (RPE)/choroid modifications at 5 and 14 days. Pigment clumps and cell vacuolization at the RPE/choroid were identified as melanin-containing RPE cells. In PEG-injected eyes, CK8/18-positive cellular elements were present at the subretinal space, IB4 immunoreactivity was significantly increased and choroidal vessels appeared diffusely thickened. However, neither VEGF nor vWF (angiogenesis/neovascularization markers) were detected in either group. At 21 days, the retina/choroid of PBS-injected animals was normal in appearance, while retina/choroid changes remained in some PEG-injected mice. Conclusions: Subretinal injection of PEG induced retina/choroid degenerative modifications that mimic the initial steps of human CNV. However, ocular changes were heterogeneous among animals from PBS and PEG groups and did not follow a consistent pattern while most PBS-injected animals showed similar degenerative changes. Abnormal growth of new vessels originating from the choroidal vasculature was not observed. Therefore, we consider that this model does not consistently reproduce CNV and that researchers should choose other rodent models of CNV to avoid misinterpreting their results.


Subject(s)
Choroid/drug effects , Choroidal Neovascularization/pathology , Polyethylene Glycols/administration & dosage , Retinal Degeneration/pathology , Retinal Pigment Epithelium/drug effects , Analysis of Variance , Animals , Biomarkers/metabolism , Choroid/metabolism , Choroid/pathology , Choroidal Neovascularization/chemically induced , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Disease Models, Animal , Gene Expression , Humans , Injections, Intraocular , Keratin-18/genetics , Keratin-18/metabolism , Lectins/genetics , Lectins/metabolism , Male , Mice , Mice, Inbred C57BL , Retinal Degeneration/chemically induced , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
6.
Keio J Med ; 67(1): 1-9, 2018 Mar 23.
Article in English | MEDLINE | ID: mdl-28592747

ABSTRACT

The hypoxia response is a fundamental phenomenon mainly regulated by hypoxia-inducible factors (HIFs). For more than a decade, we have investigated and revealed the roles of the hypoxia response in the development, physiology, and pathophysiology of the retina by generating and utilizing cell-type-specific conditional knockout mice. To investigate the functions of genes related to the hypoxia response in cells composing the retina, we generated various mouse lines that lack HIFs and/or related genes specifically in retinal neurons, astrocytes, myeloid cells, or retinal pigment epithelium cells. We found that these genes in the different types of retinal cells contribute in various ways to the homeostasis of ocular vascular and visual function. We hypothesized that the activation of HIFs is likely involved in the development and progress of retinal diseases, and we subsequently confirmed the pathological roles of HIFs in animal models of neovascular and atrophic ocular diseases. Currently, anti-vascular endothelial growth factor (anti-VEGF) therapy is a first-line treatment widely used for neovascular retinal diseases. However, alternative or additional targets are now required because several recent large-scale clinical trials and animal studies, including our own research, have indicated that VEGF antagonism may induce retinal vascular and neuronal degeneration. We have identified and confirmed a microRNA as a candidate for an alternative target against neovascular retinal diseases, and we are now working to establish a novel HIF inhibitor for clinical use based on the disease mechanism that we identified.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/drug therapy , MicroRNAs/genetics , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Developmental , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Retinal Pigment Epithelium/blood supply , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/deficiency , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
7.
Nat Commun ; 8(1): 1597, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29150606

ABSTRACT

Productive angiogenesis, a prerequisite for tumour growth, depends on the balanced release of angiogenic and angiostatic factors by different cell types within hypoxic tumours. Natural killer (NK) cells kill cancer cells and infiltrate hypoxic tumour areas. Cellular adaptation to low oxygen is mediated by Hypoxia-inducible factors (HIFs). We found that deletion of HIF-1α in NK cells inhibited tumour growth despite impaired tumour cell killing. Tumours developing in these conditions were characterised by a high-density network of immature vessels, severe haemorrhage, increased hypoxia, and facilitated metastasis due to non-productive angiogenesis. Loss of HIF-1α in NK cells increased the bioavailability of the major angiogenic cytokine vascular endothelial growth factor (VEGF) by decreasing the infiltration of NK cells that express angiostatic soluble VEGFR-1. In summary, this identifies the hypoxic response in NK cells as an inhibitor of VEGF-driven angiogenesis, yet, this promotes tumour growth by allowing the formation of functionally improved vessels.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Killer Cells, Natural/metabolism , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics
8.
Sci Rep ; 7(1): 13027, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026147

ABSTRACT

Genome wide studies indicate that vascular endothelial growth factor A (VEGF) is associated with osteoarthritis (OA), and increased VEGF expression correlates with increased disease severity. VEGF is also a chondrocyte survival factor during development and essential for bone formation, skeletal growth and postnatal homeostasis. This raises questions of how the important embryonic and postnatal functions of VEGF can be reconciled with an apparently destructive role in OA. Addressing these questions, we find that VEGF acts as a survival factor in growth plate chondrocytes during development but only up until a few weeks after birth in mice. It is also required for postnatal differentiation of articular chondrocytes and the timely ossification of bones in joint regions. In surgically induced knee OA in mice, a model of post-traumatic OA in humans, increased expression of VEGF is associated with catabolic processes in chondrocytes and synovial cells. Conditional knock-down of Vegf attenuates induced OA. Intra-articular anti-VEGF antibodies suppress OA progression, reduce levels of phosphorylated VEGFR2 in articular chondrocytes and synovial cells and reduce levels of phosphorylated VEGFR1 in dorsal root ganglia. Finally, oral administration of the VEGFR2 kinase inhibitor Vandetanib attenuates OA progression.


Subject(s)
Cartilage, Articular/embryology , Cartilage, Articular/pathology , Osteoarthritis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Administration, Oral , Animals , Antibodies/pharmacology , Bone Development , Cell Differentiation , Cell Lineage , Chondrocytes/metabolism , Collagen Type II/metabolism , Disease Progression , Endothelium/metabolism , Female , Gene Deletion , Gene Expression Regulation , Growth Plate/metabolism , Growth Plate/pathology , Integrases/metabolism , Knee Joint/pathology , Mice, Inbred C57BL , Neovascularization, Physiologic , Osteoarthritis/pathology , Osteogenesis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Br J Cancer ; 117(6): 848-855, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28742793

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) and its receptors (VEGFRs) are key regulators of angiogenesis, affecting endothelial cell survival and function. However, the effect of VEGF-VEGFR signalling on tumour cell function is not well understood. Our previous studies in colorectal cancer (CRC) cells have demonstrated an intracrine VEGF/VEGFR1 signalling mechanism that mediates CRC cell survival and chemo-sensitivity. Since extracellular VEGF signalling regulates migration of endothelial cells and various tumour cells, we attempted to determine whether intracrine VEGF signalling affects CRC cell motility. METHODS: Migration and invasion of CRC cells, with and without VEGF or VEGFR1 depletion, were assayed using transwell migration chambers. Changes in cell morphology, epithelial-mesenchymal transition (EMT) markers, and markers of cell motility were assessed by immunostaining and western blot. RESULTS: Depletion of intracellular VEGF and VEGFR1 in multiple CRC cell lines led to strong inhibition of migration and invasion of CRC cells. Except for Twist, there were no significant differences in markers of EMT between control and VEGF/VEGFR1-depleted CRC cells. However, VEGF/VEGFR1-depleted CRC cells demonstrated a significant reduction in levels of phosphorylated focal adhesion kinase and its upstream regulators pcMET and pEGFR. CONCLUSIONS: Inhibition of intracrine VEGF signalling strongly inhibits CRC cell migration and invasion by regulating proteins involved in cell motility.


Subject(s)
Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Cell Adhesion , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , HCT116 Cells , HT29 Cells , Humans , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/deficiency , Vascular Endothelial Growth Factor Receptor-1/genetics
10.
Anat Rec (Hoboken) ; 300(12): 2239-2249, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28710834

ABSTRACT

Vascular endothelial growth factor-A (VEGF) influences several physiological processes including endothelial cell function, angiogenesis and maintenance of organ/tissue capillarity. While the functional aspects of VEGF were vigorously investigated, only little detail is known on structural integrity of skeletal muscle fibers and capillaries in mice lacking VEGF expression in their muscles. Therefore, we assessed systematically the architecture of the glycolytic plantaris and the oxidative soleus muscles obtained from muscle-specific VEGF knockout (mVEGF-KO, n = 7) mice and their wild-type (WT, n = 7) littermates by morphometry after transmission electron microscopy. The capillary/fiber ratio was lower (plantaris: -63.5%; soleus: -54.8%; P ≤ 0.05) in mVEGF-KO mice than in WT mice. In plantaris, quantification of volume density (Vv) of compartments revealed higher Vv of total mitochondria (+56.5%, P ≤ 0.05) as well as higher Vv-values for both intrafibrillar (+39%; P ≤ 0.05) and subsarcolemmal (+220%; P ≤ 0.05) mitochondrial pools in mVEGF-KO mice than WT mice. The capillary phenotype also differed (P ≤ 0.05) between the two mouse-strains: Vv (-17.4%), absolute area size (-19.1%) and thickness (-19.6%) of the endothelium layer were lower and Vv of capillary lumen (+15.1%) was higher in mVEGF-KO mice than in WT littermates. In soleus, mitochondrial Vv in fibers and the structural indicators specific to the capillary phenotype exhibited the same tendency in differences between the mouse strains without reaching statistical significance. Our morphometric analysis demonstrates that the lower capillary supply in plantaris of mVEGF-KO mice is accompanied by higher mitochondrial Vv in muscle fibers as well as lumen dilation and endothelium thinning of capillaries. These structural alterations were more pronounced in a glycolytic than an oxidative muscle. Anat Rec, 300:2239-2249, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Capillaries/metabolism , Capillaries/ultrastructure , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/blood supply , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics
11.
Int J Mol Sci ; 18(2)2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28230770

ABSTRACT

Vascular endothelial growth factor A (Vegfa) signaling regulates cardiovascular development. However, the cellular mechanisms of Vegfa signaling in early cardiogenesis remain poorly understood. The present study aimed to understand the differential functions and mechanisms of Vegfa signaling in cardiac development. A loss-of-function approach was utilized to study the effect of Vegfa signaling in cardiogenesis. Both morphants and mutants for vegfaa display defects in cardiac looping and chamber formation, especially the ventricle. Vegfa regulates the heart morphogenesis in a dose-dependent manner. Furthermore, the initial fusion of the bilateral myocardium population is delayed rather than endocardium. The results demonstrate that Vegfa signaling plays a direct impact on myocardium fusion, indicating that it is the initial cause of the heart defects. The heart morphogenesis is regulated by Vegfa in a dose-dependent manner, and later endocardium defects may be secondary to impaired myocardium-endocardium crosstalk.


Subject(s)
Heart/embryology , Myocardium/metabolism , Organogenesis/genetics , Vascular Endothelial Growth Factor A/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Cell Proliferation , Gene Expression Regulation, Developmental , Morphogenesis/genetics , Mutation , Myocytes, Cardiac/metabolism , Phenotype , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/metabolism
12.
J Nanosci Nanotechnol ; 16(6): 5464-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27427585

ABSTRACT

N-isopropylacrylamide modified PEI (PEN) was synthesized via Michael addition and was developed as an efficient siRNA delivery system both in vitro and in vivo. PEN showed significant enhanced cytocompatibility compared with commercial PEI-25k. The complexation of PEN with siRNA was studied by gel retardation, particle size and zeta potential measurement. The in vitro transfection ability of PEN was measured by qRT-PCR assay, and achieved obviously enhanced gene silencing efficiency compared with PEI-25k. The confocal imaging and flow cytometric analysis further validated its excellent intracellular trafficking ability. For antitumor treatment experiment, PEN mediated siVEGF showed obviously therapeutic effects for the treatment of CT26 tumor. Therefore, the present study demonstrated a useful strategy for constructing efficient siRNA delivery vehicles for antitumor therapy.


Subject(s)
Acrylamides/chemistry , Drug Carriers/chemistry , Genetic Therapy , Polyethyleneimine/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Drug Carriers/metabolism , Gene Silencing , Intracellular Space/metabolism , Mice , Polyethyleneimine/metabolism , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics
13.
PLoS One ; 11(3): e0151396, 2016.
Article in English | MEDLINE | ID: mdl-26978773

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) is a highly conserved, master regulatory molecule required for endothelial cell proliferation, organization, migration and branching morphogenesis. Podocoryne carnea and drosophila, which lack endothelial cells and a vascular system, express VEGF homologs, indicating potential roles beyond angiogenesis and vasculogenesis. The role of VEGF in the development and homeostasis of the postnatal small intestine is unknown. We hypothesized regulating VEGF bioavailability in the postnatal small intestine would exhibit effects beyond the vasculature and influence epithelial cell stem/progenitor populations. METHODS: VEGF mutant mice were created that overexpressed VEGF in the brush border of epithelium via the villin promotor following doxycycline treatment. To decrease VEGF bioavailability, sFlt-1 mutant mice were generated that overexpressed the soluble VEGF receptor sFlt-1 upon doxycycline administration in the intestinal epithelium. Mice were analyzed after 21 days of doxycycline administration. RESULTS: Increased VEGF expression was confirmed by RT-qPCR and ELISA in the intestine of the VEGF mutants compared to littermates. The VEGF mutant duodenum demonstrated increased angiogenesis and vascular leak as compared to littermate controls. The VEGF mutant duodenum revealed taller villi and increased Ki-67-positive cells in the transit-amplifying zone with reduced Lgr5 expression. The duodenum of sFlt-1 mutants revealed shorter villi and longer crypts with reduced proliferation in the transit-amplifying zone, reduced expression of Dll1, Bmp4 and VE-cadherin, and increased expression of Sox9 and EphB2. CONCLUSIONS: Manipulating VEGF bioavailability leads to profound effects on not only the intestinal vasculature, but epithelial stem and progenitor cells in the intestinal crypt. Elucidation of the crosstalk between VEGF signaling in the vasculature, mesenchyme and epithelial stem/progenitor cell populations may direct future cell therapies for intestinal dysfunction or disease.


Subject(s)
Duodenum/metabolism , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/physiology , Aberrant Crypt Foci/genetics , Animals , Animals, Suckling , Capillary Leak Syndrome/genetics , Cell Division , Cells, Cultured , Doxycycline/pharmacology , Duodenum/blood supply , Duodenum/cytology , Duodenum/growth & development , Gene Expression Regulation , Image Processing, Computer-Assisted , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Microvilli/ultrastructure , Neovascularization, Physiologic/genetics , Organoids , Recombinant Fusion Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Stem Cells/cytology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Vascular Endothelial Growth Factor Receptor-1/genetics
14.
J Cell Physiol ; 231(2): 505-11, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26201683

ABSTRACT

Exercise is dependent on adequate oxygen supply for mitochondrial respiration in both cardiac and locomotor muscle. To determine whether skeletal myofiber VEGF is critical for regulating exercise capacity, independent of VEGF function in the heart, ablation of the VEGF gene was targeted to skeletal myofibers (skmVEGF-/-) during embryogenesis (∼ E9.5), leaving intact VEGF expression by all other cells in muscle. In adult mice, VEGF levels were decreased in the soleus (by 65%), plantaris (94%), gastrocnemius (74%), EDL (99%) and diaphragm (64%) (P < 0.0001, each muscle). VEGF levels were unchanged in the heart. Treadmill speed (WT 86 ± 4 cm/sec, skmVEGF-/- 70 ± 5 cm/sec, P = 0.006) and endurance (WT 78 ± 24 min, skmVEGF-/- 18 ± 4 min, P = 0.0004) were severely limited in skmVEGF-/- mice in contrast to minor effect of conditional skmVEGF gene deletion in the adult. Body weight was also reduced (WT 22.8 ± 1.6 g, skmVEGF-/-, 21.1 ± 1.5, P = 0.02), but the muscle mass/body weight ratio was unchanged. The capillary/fiber ratio was lower in skmVEGF-/- plantaris (WT 1.51 ± 0.12, skmVEGF-/- 1.16 ± 0.20, P = 0.01), gastrocnemius (WT 1.61 ± 0.08, skmVEGF-/- 1.39 ± 0.08, P = 0.01), EDL (WT 1.36 ± 0.07, skmVEGF-/- 1.14 ± 0.13, P = 0.03) and diaphragm (WT 1.39 ± 0.18, skmVEGF-/- 0.79 ± 0.16, P = 0.0001) but, not in soleus. Cardiac function (heart rate, maximal pressure, maximal dP/dt, minimal dP/dt,) in response to dobutamine was not impaired in anesthetized skmVEGF-/- mice. Isolated soleus and EDL fatigue times were 16% and 20% (P < 0.02) longer, respectively, in skmVEGF-/- mice than the WT group. These data suggest that skeletal myofiber VEGF expressed during development is necessary to establish capillary networks that allow maximal exercise capacity.


Subject(s)
Muscle Fibers, Skeletal/physiology , Physical Exertion/physiology , Vascular Endothelial Growth Factor A/deficiency , Animals , Capillaries/growth & development , Capillaries/physiology , Exercise Test , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Muscle Fatigue/genetics , Muscle Fatigue/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/growth & development , Muscle, Skeletal/physiology , Physical Exertion/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology
15.
Arterioscler Thromb Vasc Biol ; 36(1): 19-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26603154

ABSTRACT

OBJECTIVE: Ocular neovascularization (ONV) is a pathological feature of sight-threatening human diseases, such as diabetic retinopathy and age-related macular degeneration. Macrophage depletion in mouse models of ONV reduces the formation of pathological blood vessels, and myeloid cells are widely considered an important source of the vascular endothelial growth factor A (VEGF). However, the importance of VEGF or its upstream regulators hypoxia-inducible factor-1α (HIF1α) and hypoxia-inducible factor-2α (HIF2α) as myeloid-derived regulators of ONV remains to be determined. APPROACH AND RESULTS: We used 2 mouse models of ONV, choroidal neovascularization and oxygen-induced retinopathy, to show that Vegfa is highly expressed by several cell types, but not myeloid cells during ONV. Moreover, myeloid-specific VEGF ablation did not reduce total ocular VEGF during choroidal neovascularization or oxygen-induced retinopathy. In agreement, the conditional inactivation of Vegfa, Hif1a, or Epas1 in recruited and resident myeloid cells that accumulated at sites of neovascularization did not significantly reduce choroidal neovascularization or oxygen-induced retinopathy. CONCLUSIONS: The finding that myeloid cells are not a significant local source of VEGF in these rodent models of ONV suggests that myeloid function in neovascular eye disease differs from skin wound healing and other neovascular pathologies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Choroidal Neovascularization/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloid Cells/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice, Knockout , Myeloid Cells/pathology , Oxygen , Retinal Neovascularization/chemically induced , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Retinal Vessels/pathology , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics
16.
Nutrition ; 31(9): 1083-95, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26233865

ABSTRACT

Preeclampsia is a low-grade systemic inflammatory condition in which oxidative stress and endothelial dysfunction occurs. Plasma levels of soluble receptor for vascular endothelial growth factor (VEGFR)-1, also known as sFlt1 (soluble fms-like tyrosine kinase 1), an antiangiogenic factor have been reported to be elevated in preeclampsia. It was reported that pregnant mice deficient in catechol-O-methyltransferase (COMT) activity show a preeclampsia-like phenotype due to a deficiency or absence of 2-methoxyoestradiol (2-ME), a natural metabolite of estradiol that is elevated during the third trimester of normal human pregnancy. Additionally, autoantibodies (AT1-AAs) that bind and activate the angiotensin II receptor type 1 a (AT1 receptor) also have a role in preeclampsia. None of these abnormalities are consistently seen in all the patients with preeclampsia and some of them are not specific to pregnancy. Preeclampsia could occur due to an imbalance between pro- and antiangiogenic factors. VEGF, an angiogenic factor, is necessary for the transport of polyunsaturated fatty acids (PUFAs) to endothelial cells. Hence reduced VEGF levels decrease the availability of PUFAs to endothelial cells. This leads to a decrease in the formation of anti-inflammatory and angiogenic factors: lipoxins, resolvins, protectins, and maresins from PUFAs. Lipoxins, resolvins, protectins, maresins, and PUFAs suppress insulin resistance; activation of leukocytes, platelets, and macrophages; production of interleukin-6 and tumor necrosis factor-α; and oxidative stress and endothelial dysfunction; and enhance production of prostacyclin and nitric oxide (NO). Estrogen enhances the formation of lipoxin A4 and NO. PUFAs also augment the production of NO and inhibit the activity of angiotensin-converting enzyme and antagonize the actions of angiotensin II. Thus, PUFAs can prevent activation of angiotensin II receptor type 1 a (AT1 receptor). Patients with preeclampsia have decreased plasma phospholipid concentrations of arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), the precursors of lipoxins (from AA), resolvins (from EPA and DHA), and protectins (from DHA) and prostaglandin E1 (PGE1 from DGLA: dihomo-γ-linolenic acid) and prostacyclin (PGI2 derived from AA). Based on these evidences, it is proposed that preeclampsia may occur due to deficiency of PUFAs and their anti-inflammatory products: lipoxins, resolvins, protectins, and maresins.


Subject(s)
Angiogenesis Inhibitors/blood , Cytokines/blood , Eicosanoids/blood , Fatty Acids, Unsaturated/blood , Inflammation/blood , Pre-Eclampsia/etiology , Vascular Endothelial Growth Factor A/blood , Animals , Endothelium, Vascular/metabolism , Fatty Acids, Unsaturated/deficiency , Female , Humans , Pre-Eclampsia/blood , Pregnancy , Receptor, Angiotensin, Type 1/blood , Vascular Endothelial Growth Factor A/deficiency
17.
J Clin Invest ; 125(7): 2661-76, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26053665

ABSTRACT

In mammals, the outflow tract (OFT) of the developing heart septates into the base of the pulmonary artery and aorta to guide deoxygenated right ventricular blood into the lungs and oxygenated left ventricular blood into the systemic circulation. Accordingly, defective OFT septation is a life-threatening condition that can occur in both syndromic and nonsyndromic congenital heart disease. Even though studies of genetic mouse models have previously revealed a requirement for VEGF-A, the class 3 semaphorin SEMA3C, and their shared receptor neuropilin 1 (NRP1) in OFT development, the precise mechanism by which these proteins orchestrate OFT septation is not yet understood. Here, we have analyzed a complementary set of ligand-specific and tissue-specific mouse mutants to show that neural crest-derived SEMA3C activates NRP1 in the OFT endothelium. Explant assays combined with gene-expression studies and lineage tracing further demonstrated that this signaling pathway promotes an endothelial-to-mesenchymal transition that supplies cells to the endocardial cushions and repositions cardiac neural crest cells (NCCs) within the OFT, 2 processes that are essential for septal bridge formation. These findings elucidate a mechanism by which NCCs cooperate with endothelial cells in the developing OFT to enable the postnatal separation of the pulmonary and systemic circulation.


Subject(s)
Heart Septum/embryology , Heart Ventricles/metabolism , Neural Crest/metabolism , Neuropilin-1/metabolism , Semaphorins/metabolism , Animals , Apoptosis , Cell Proliferation , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Female , Heart Septum/cytology , Heart Septum/metabolism , Heart Ventricles/embryology , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Neural Crest/embryology , Neuropilin-1/deficiency , Neuropilin-1/genetics , Pregnancy , Semaphorins/deficiency , Semaphorins/genetics , Signal Transduction , Tissue Distribution , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
Arch. Soc. Esp. Oftalmol ; 90(supl.1): 6-10, mar. 2015. ilus, tab
Article in Spanish | IBECS | ID: ibc-144565

ABSTRACT

Aflibercept es una proteina de fusion que combina en su estructura quimica, la fraccion constante de cualquier IgG con una fraccion variable construida con partes fundamentales de los receptores del factor de crecimiento del endotelio vascular, por ello es capaz de fijar a diversas isoformas del factor de crecimiento del endotelio vascular y tambien al factor de crecimiento placentario, lo que se ha puesto en relacion con un posible efecto sinergico en la eficacia. La afinidad es mayor que la que presentan ranibizumab y bevacizumab. Ademas produce un efecto antiinflamatorio intraocular. La administracion por via intravitrea cursa con la presencia de trazos del farmaco en el plasma del paciente; de hecho, las concentraciones son tan reducidas que la presencia de efectos adversos sistemicos, incluida la hipertension arterial, es practicamente nula. Una semivida de eliminacion intraocular prolongada unida a la afinidad elevada supone que sea posible la utilizacion en pautas posologicas comodas, ya que tras una inyeccion mensual para las 3 primeras dosis se aumenta el intervalo a una inyeccion cada 2 meses, que tras los primeros 12 meses puede vincularse a los resultados visuales y anatómicos (AU)


Aflibercept is a fusion protein whose chemical structure combines the constant fraction of any IgG with a variable fraction constructed with fundamental parts of VEGF receptors. Consequently, it is able to bind to various VEGF as well as to placental growth factor (PIGF), which has been related to a possible synergistic effect in efficacy. The affinity of this drug is higher than that of ranibizumab and bevacizumab. Moreover, it has an intraocular antiinflammatory effect. Intravitreal administration leads to the presence of traces of the drug in plasma but the concentrations are so reduced that the presence of systemic adverse effects, including arterial hypertension, is practically nil. Because of its prolonged intraocular elimination half-life and high affinity, the drug can be administered in convenient regimens, since, after an initial monthly injection for the first three doses, the interval between injections is increased to one every two months and, after the first 12 months, the dosing will depend on the visual and anatomical results (AU)


Subject(s)
Female , Humans , Male , Pharmacology/classification , Pharmacology/methods , Pharmacokinetics , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A , Dosage/analysis , Eye Abnormalities/complications , Eye Abnormalities/metabolism , Pharmacology/standards , Pharmacology/trends , Vascular Endothelial Growth Factor A/classification , Vascular Endothelial Growth Factor A/deficiency , Dosage/methods , Eye Abnormalities/genetics , Eye Abnormalities/pathology
19.
PLoS One ; 10(2): e0116332, 2015.
Article in English | MEDLINE | ID: mdl-25658474

ABSTRACT

Because VEGFA has been implicated in follicle development, the objective of this study was to determine the effects of granulosa- and germ cell-specific VEGFA loss on ovarian morphogenesis, function, and female fertility. pDmrt1-Cre mice were mated to floxed VEGFA mice to develop granulosa-/germ cell-specific knockouts (pDmrt1-Cre;Vegfa-/-). The time from mating to first parturition was increased when pDmrt1-Cre;Vegfa-/- females were mated to control males (P = 0.0008) and tended to be longer for heterozygous females (P < 0.07). Litter size was reduced for pDmrt1-Cre;Vegfa-/- females (P < 0.007). The time between the first and second parturitions was also increased for heterozygous females (P < 0.04) and tended to be increased for pDmrt1-Cre;Vegfa-/- females (P < 0.07). pDmrt1-Cre;Vegfa-/- females had smaller ovaries (P < 0.04), reduced plasma estradiol (P < 0.007), fewer developing follicles (P < 0.008) and tended to have fewer corpora lutea (P < 0.08). Expression of Igf1r was reduced (P < 0.05); expression of Foxo3a tended to be increased (P < 0.06); and both Fshr (P < 0.1) and Sirt6 tended to be reduced (P < 0.06) in pDmrt1-Cre;Vegfa-/- ovaries. To compare VEGFA knockouts, we generated Amhr2-Cre;Vegfa-/- mice that required more time from mating to first parturition (P < 0.003) with variable ovarian size. Both lines had more apoptotic granulosa cells, and vascular staining did not appear different. Taken together these data indicate that the loss of all VEGFA isoforms in granulosa/germ cells (proangiogenic and antiangiogenic) causes subfertility by arresting follicular development, resulting in reduced ovulation rate and fewer pups per litter.


Subject(s)
Fertility/physiology , Granulosa Cells/metabolism , Litter Size/physiology , Morphogenesis/physiology , Ovarian Follicle/growth & development , Vascular Endothelial Growth Factor A/deficiency , Analysis of Variance , Animals , DNA Primers/genetics , Estradiol/blood , Female , Fluorescent Antibody Technique , Genotype , Immunohistochemistry , Male , Mice , Mice, Knockout , Protein Isoforms/deficiency , Transcription Factors/metabolism
20.
FEBS Lett ; 588(12): 2078-86, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24815691

ABSTRACT

Oncogenic activation of VEGF is found in various malignancies, including ovarian cancer. In this study, we investigate the role of microRNA (miRNA) in the regulation of VEGF in ovarian cancer. We find that miR-718 is expressed at low levels and inversely correlates with VEGF expression in ovarian cancer specimens. MiR-718 also directly targets and represses VEGF expression. In addition, miR-718 restoration inhibits ovarian cancer proliferation both in vitro and in vivo. Moreover, VEGF expression could reverse the effect of miR-718 on ovarian cancer by increasing the levels of phosphorylated AKT. These results suggest a new therapeutic strategy in ovarian cancer by restoring miR-718 expression, which is involved in VEGF regulation.


Subject(s)
Disease Progression , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Vascular Endothelial Growth Factor A/deficiency , 3' Untranslated Regions/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...