Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Peptides ; 154: 170823, 2022 08.
Article in English | MEDLINE | ID: mdl-35660637

ABSTRACT

Skeletal muscle is the major effector organ for exercise. It has been proposed that VEGFB is significantly related to apoptosis in various cell types but not yet in skeletal muscle. We hypothesize that the decrease of VEGFB in skeletal muscle participates in the occurrence of skeletal muscle apoptosis and that exercise inhibits apoptosis by elevating the expression of VEGFB in skeletal muscle cells. Based on this hypothesis, we developed in vitro experiments to mimic the effect of exercise through electrical pulse stimulation (EPS) to observe the effect of EPS on apoptosis and the change in VEGFB expression in differentiated myotubes. In addition, we employed RNA interference to explore whether VEGFB is directly involved in the regulation of myotube apoptosis during EPS. Our results showed that exogenous VEGFB167 significantly inhibited C2C12 myotube apoptosis induced by TNF-α treatment and that endogenous VEGFB in differentiated C2C12 myotubes was significantly upregulated by EPS. In addition, EPS significantly changed the expression of the apoptotic indicators Bax and Bcl-2 at the mRNA level and downregulated the protein expression of cleaved caspase-3. The antiapoptotic effect of EPS weakened substantially as VEGFB in C2C12 myotubes was inhibited. Taken together, these results indicate that exercise-like EPS inhibits apoptosis by increasing the expression of C2C12 myotube-derived VEGFB.


Subject(s)
Muscle Fibers, Skeletal , Vascular Endothelial Growth Factor B , Apoptosis , Electric Stimulation/methods , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/pharmacology
2.
Int J Mol Sci ; 22(24)2021 Dec 12.
Article in English | MEDLINE | ID: mdl-34948148

ABSTRACT

It has been demonstrated that vascular endothelial growth factor B (VEGFB) plays a vital role in regulating vascular biological function. However, the role of VEGFB in regulating skeletal muscle cell proliferation and differentiation remains unclear. Thus, this study aimed to investigate the effects of VEGFB on C2C12 myoblast proliferation and differentiation and to explore the underlying mechanism. For proliferation, VEGFB significantly promoted the proliferation of C2C12 myoblasts with the upregulating expression of cyclin D1 and PCNA. Meanwhile, VEGFB enhanced vascular endothelial growth factor receptor 1 (VEGFR1) expression and activated the PI3K/Akt signaling pathway in a VEGFR1-dependent manner. In addition, the knockdown of VEGFR1 and inhibition of PI3K/Akt totally abolished the promotion of C2C12 proliferation induced by VEGFB, suggesting that VEGFB promoted C2C12 myoblast proliferation through the VEGFR1-PI3K/Akt signaling pathway. Regarding differentiation, VEGFB significantly stimulated the differentiation of C2C12 myoblasts via VEGFR, with elevated expressions of MyoG and MyHC. Furthermore, the knockdown of VEGFR1 rather than NRP1 eliminated the VEGFB-stimulated C2C12 differentiation. Moreover, VEGFB activated the PI3K/Akt/mTOR signaling pathway in a VEGFR1-dependent manner. However, the inhibition of PI3K/Akt/mTOR blocked the promotion of C2C12 myoblasts differentiation induced by VEGFB, indicating the involvement of the PI3K/Akt pathway. To conclude, these findings showed that VEGFB promoted C2C12 myoblast proliferation and differentiation via the VEGFR1-PI3K/Akt signaling pathway, providing new insights into the regulation of skeletal muscle development.


Subject(s)
Cell Differentiation , Cell Proliferation , Myoblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Cell Line , Mice , Vascular Endothelial Growth Factor B/pharmacology
3.
Life Sci ; 253: 117677, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32305525

ABSTRACT

Impaired vascularization of adipose tissue leads to local hypoxia and results in chronic inflammation and obesity-related metabolic disorders. We have recently constructed an engineered protein named tPep-VEGF-B by bridging vascular endothelial growth factor (VEGF-B) with an adipose-targeted peptide. Here, we reported tPep-VEGF-B diminishes obesity and alleviates metabolic syndrome. High fat diet (HFD) treated mice had reduced adipose vascular density and showed adipose hypoxia and metabolic complications. In contrast, the treatment of tPep-VEGF-B repressed HFD-induced body weight gain, which led to increased adipose vasculature and reduced hypoxia. This treatment also alleviated obesity associated hyperlipidemia and fatty liver disease. This study provided a leading molecule for the treatment of type 2 diabetes and other metabolic diseases. It also provided experimental support for the theory that modulation of angiogenesis plays a key role in the treatment of metabolic diseases.


Subject(s)
Adipose Tissue/blood supply , Drug Delivery Systems , Fatty Liver/prevention & control , Obesity/prevention & control , Vascular Endothelial Growth Factor B/administration & dosage , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Hyperlipidemias/prevention & control , Male , Mice , Mice, Inbred C57BL , Peptides/chemistry , Vascular Endothelial Growth Factor B/pharmacology
4.
Growth Factors ; 37(1-2): 76-84, 2019 04.
Article in English | MEDLINE | ID: mdl-31215273

ABSTRACT

To investigate (1) the effect of vascular endothelial growth factor B (VEGFB) on lipid accumulation and the alteration of fatty acids and fatty acid-related enzymes in C2C12 myotubes incubated with fatty acids and (2) the regulatory effect of VEGFB on skeletal muscle lipid metabolism. Mouse C2C12 myotubes were incubated with oleic acid (OA) and palmitic acid (PA), and differentiated mature C2C12 myotubes were treated with VEGFB. Oil-red O staining, BODIPY staining and cell triglycerides (TG) content were examined. Total RNA was isolated, and real-time PCR analysis was performed. Treatment with 100 µM OA and 50 µM PA induced lipid droplet accumulation and increased TG content (p < .01), and 100 ng/mL VEGFB reduced lipid droplet accumulation and decreased TG content (p < .01). Treatment with 100 ng/mL VEGFB significantly induced the mRNA expression of fatty acid transport protein 1 (FATP1) (p < .01) and FATP4 (p < .01). Treatment with 100 ng/mL VEGFB significantly induced the mRNA expression of adipose TG lipase and hormone-sensitive lipase (p < .01) as well as carnitine palmitoyltransferase I (p < .01), peroxisome proliferator-activated receptor-γ coactivator-1α (p < .01), acyl-coa dehydrogenase very long chain (p < .05), acyl-coa synthetase long-chain family member 1 (p < .01), peroxisomal acyl-coenzyme A oxidase 1 (p < .05), and mitochondrial uncoupling protein 3 (p < .01). VEGFB enhanced FATP1and FATP4 expression, promoted C2C12 myotube fatty acid oxidation and TG decomposition, and inhibited C2C12 myotube fatty acid re-esterification, thus inhibiting lipid accumulation in C2C12 myotubes incubated with fatty acids.


Subject(s)
Lipid Metabolism , Muscle Fibers, Skeletal/metabolism , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , Vascular Endothelial Growth Factor B/pharmacology , Animals , Cell Line , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Lipid Droplets/metabolism , Mice , Muscle Fibers, Skeletal/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
5.
Pharmacol Res ; 143: 33-39, 2019 05.
Article in English | MEDLINE | ID: mdl-30851357

ABSTRACT

Oxidative stress, due to insufficiency of antioxidants or over-production of oxidants, can lead to severe cell and tissue damage. Oxidative stress occurs constantly and has been shown to be involved in innumerable diseases, such as degenerative, cardiovascular, neurological, and metabolic disorders, cancer, and aging, thus highlighting the vital need of antioxidant defense mechanisms. Vascular endothelial growth factor B (VEGF-B) was discovered a long time ago, and is abundantly expressed in most types of cells and tissues. VEGF-B remained functionally mysterious for many years and later on has been shown to be minimally angiogenic. Recently, VEGF-B is reported to be a potent antioxidant by boosting the expression of key antioxidant enzymes. Thus, one major role of VEGF-B lies in safeguarding tissues and cells from oxidative stress-induced damage. VEGF-B may therefore have promising therapeutic utilities in treating oxidative stress-related diseases. In this review, we discuss the current knowledge on the newly discovered antioxidant function of VEGF-B and the related molecular mechanisms, particularly, in relationship to some oxidative stress-related diseases, such as retinitis pigmentosa, age-related macular degeneration, diabetic retinopathy, glaucoma, amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease.


Subject(s)
Antioxidants/therapeutic use , Eye Diseases/drug therapy , Neurodegenerative Diseases/drug therapy , Vascular Endothelial Growth Factor B/therapeutic use , Animals , Antioxidants/pharmacology , Eye Diseases/metabolism , Humans , Neurodegenerative Diseases/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/pharmacology
6.
Biochem J ; 476(4): 645-663, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30700502

ABSTRACT

Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are pivotal regulators of angiogenesis. The VEGF-VEGFR system is therefore an important target of anti-angiogenesis therapy. Based on the X-ray structure of VEGF-B/VEGFR-1 D2, we designed a cyclic peptide (known as VGB1) reproducing the α1 helix and its adjacent region to interfere with signaling through VEGFR-1. Unexpectedly, VGB1 bound VEGFR-2 in addition to VEGFR-1, leading to inhibition of VEGF-stimulated proliferation of human umbilical vein endothelial cells and 4T1 murine mammary carcinoma cells, which express VGEFR-1 and VEGFR-2, and U87 glioblastoma cells that mostly express VEGFR-2. VGB1 inhibited different aspects of angiogenesis, including proliferation, migration and tube formation of endothelial cells stimulated by VEGF-A through suppression of extracellular signal-regulated kinase 1/2 and AKT (Protein Kinase B) phosphorylation. In a murine 4T1 mammary carcinoma model, VGB1 caused regression of tumors without causing weight loss in association with impaired cell proliferation (decreased Ki67 expression) and angiogenesis (decreased CD31 and CD34 expression), and apoptosis induction (increased TUNEL staining and p53 expression, and decreased Bcl-2 expression). According to far-UV circular dichroism (CD) and molecular dynamic simulation data, VGB1 can adopt a helical structure. These results, for the first time, demonstrate that α1 helix region of VEGF-B recognizes both VEGFR-1 and VEGFR-2.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Peptides, Cyclic , Vascular Endothelial Growth Factor B , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factor Receptor-2 , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Protein Structure, Secondary , Vascular Endothelial Growth Factor B/chemistry , Vascular Endothelial Growth Factor B/pharmacology , Vascular Endothelial Growth Factor Receptor-1/agonists , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/agonists , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Proc Natl Acad Sci U S A ; 115(41): 10351-10356, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30249667

ABSTRACT

VEGF-B was discovered a long time ago. However, unlike VEGF-A, whose function has been extensively studied, the function of VEGF-B and the mechanisms involved still remain poorly understood. Notwithstanding, drugs that inhibit VEGF-B and other VEGF family members have been used to treat patients with neovascular diseases. It is therefore critical to have a better understanding of VEGF-B function and the underlying mechanisms. Here, using comprehensive methods and models, we have identified VEGF-B as a potent antioxidant. Loss of Vegf-b by gene deletion leads to retinal degeneration in mice, and treatment with VEGF-B rescues retinal cells from death in a retinitis pigmentosa model. Mechanistically, we demonstrate that VEGF-B up-regulates numerous key antioxidative genes, particularly, Gpx1 Loss of Gpx1 activity largely diminished the antioxidative effect of VEGF-B, demonstrating that Gpx1 is at least one of the critical downstream effectors of VEGF-B. In addition, we found that the antioxidant function of VEGF-B is mediated mainly by VEGFR1. Given that oxidative stress is a crucial factor in numerous human diseases, VEGF-B may have therapeutic value for the treatment of such diseases.


Subject(s)
Antioxidants/metabolism , Retinal Degeneration/genetics , Vascular Endothelial Growth Factor B/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Disease Models, Animal , Gene Expression Regulation , Glutathione Peroxidase/genetics , Mice, Inbred C57BL , Mice, Mutant Strains , Oxidative Stress , Retina/drug effects , Retina/pathology , Retinal Degeneration/drug therapy , Retinitis Pigmentosa/genetics , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Glutathione Peroxidase GPX1
8.
Mol Neurobiol ; 55(5): 3611-3626, 2018 May.
Article in English | MEDLINE | ID: mdl-28290152

ABSTRACT

Ischemic stroke triggers endogenous angiogenic mechanisms, which correlates with longer survival in patients. As such, promoting angiogenesis appears to be a promising approach. Experimental studies investigated mostly the potent angiogenic factor vascular endothelial growth factor isoform-A (VEGF-A). However, VEGF-A increases the risk of destabilizing the brain microvasculature, thus hindering the translation of its usage in clinics. An attractive alternative VEGF isoform-B (VEGF-B) was recently reported to act as a survival factor rather than a potent angiogenic factor. In this study, we investigated the therapeutic potential of VEGF-B in ischemic stroke using different in vivo and in vitro approaches. We showed that the delayed intranasal administration of VEGF-B reduced neuronal damage and inflammation. Unexpectedly, VEGF-B stimulated the formation of stable brain microvasculature within the injured region by promoting the interaction between endothelial cells and pericytes. Our data indicate that the effects of VEGF-B were mediated via its specific receptor VEGF receptor-1 (VEGFR-1) that is predominately expressed in brain pericytes. Importantly, VEGF-B promoted the survival of pericytes, and not brain endothelial cells, by inducing expression of the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) and the main protein involved in energy homeostasis AMP-activated protein kinase α (AMPKα). Moreover, we showed that VEGF-B stimulated the pericytic release of factors stimulating a "reparative angiogenesis" that does not compromise microvasculature stability. Our study unraveled hitherto unknown role of VEGF-B/VEGFR-1 signaling in regulating the function of pericytes. Furthermore, our findings suggest that brain microvasculature stabilization via VEGF-B constitutes a safe therapeutic approach for ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Pericytes/drug effects , Stroke/drug therapy , Vascular Endothelial Growth Factor B/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/metabolism , Administration, Intranasal , Animals , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Male , Mice , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Signal Transduction/drug effects , Stroke/metabolism , Vascular Endothelial Growth Factor B/pharmacology
9.
J Transl Med ; 14(1): 116, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27146579

ABSTRACT

AIMS: To investigate whether vascular endothelial growth factor B (VEGF-B) improves myocardial survival and cardiac stem cell (CSC) function in the ischemia-reperfusion (I/R) heart and promotes CSC mobilization and angiogenesis. METHODS AND RESULTS: One hour after myocardial ischemia and infarction, rats were treated with recombinant human VEGF-B protein following 24 h or 7 days of myocardial reperfusion. Twenty-four hours after myocardial I/R, VEGF-B increased pAkt and Bcl-2 levels, reduced p-p38MAPK, LC3-II/I, beclin-1, CK, CK-MB and cTnt levels, triggered cardiomyocyte protection against I/R-induced autophagy and apoptosis, and contributed to the decrease of infarction size and the improvement of heart function during I/R. Simultaneously, an in vitro hypoxia-reoxygenation (H/R)-induced H9c2 cardiomyocyte injury model was used to mimic I/R injury model in vivo; in this model, VEGF-B decreased LDH release, blocked H/R-induced apoptosis by inhibiting cell autophagy, and these special effects could be abolished by the autophagy inducer, rapamycin. Mechanistically, VEGF-B markedly activated the Akt signaling pathway while slightly inhibiting p38MAPK, leading to the blockade of cell autophagy and thus protecting cardiomyocyte from H/R-induced activation of the intrinsic apoptotic pathway. Seven days after I/R, VEGF-B induced the expression of SDF-1α and HGF, resulting in the massive mobilization and homing of c-Kit positive cells, triggering further angiogenesis and vasculogenesis in the infracted heart and contributing to the improvement of I/R heart function. CONCLUSION: VEGF-B could contribute to a favorable short- and long-term prognosis for I/R via the dual manipulation of cardiomyocytes and CSCs.


Subject(s)
Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Vascular Endothelial Growth Factor B/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Shape/drug effects , Creatine Kinase/metabolism , Disease Models, Animal , Heart Function Tests/drug effects , Male , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stem Cells/drug effects , Troponin T/metabolism , bcl-2-Associated X Protein/metabolism
10.
Int J Oral Maxillofac Surg ; 44(12): 1506-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26338076

ABSTRACT

Angiogenesis is critical for peri-implant bone regeneration and osseointegration. Endothelial cells (ECs) play an important role in angiogenesis during the early stage of bone formation. Nerve growth factor (NGF) is also reported to function as an angiogenic growth factor. The effects of NGF on ECs seeded on titanium surfaces are unclear. This study was done to investigate the influence of NGF on peri-implant angiogenesis in vitro and in vivo. We used two different titanium surfaces. ECs seeded on these surfaces were treated with indicated concentrations of NGF or vascular endothelial growth factor (VEGF). Proliferation, differentiation, morphological features, and amounts attached were assessed. Chicken embryo chorioallantoic membrane (CAM) was adopted to evaluate the effect of NGF in vivo. The results showed that NGF could promote EC proliferation on both titanium surfaces (F1d=2.083, P=0.156; F3d=30.857, P=0.0002; F5d=4.440, P=0.041; F7d=11.065, P=0.001). NGF and the SLA surface upregulated mRNA of NGF, TrkA, and p75 expression (FNGF=11.941, P=0.003; FTrkA=28.514, P=0.004; Fp75=7.725, P=0.01). In vivo, the supernatants of the NGF-treated group could promote neovascularization in CAM (F=17.662, P=0.009). This study demonstrated that NGF could enhance EC proliferation, gene expression on different titanium surfaces, and neovascularization in CAM. This provides novel information in relation to the promotion of early dental implant osseointegration.


Subject(s)
Endothelial Cells/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor B/pharmacology , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Chick Embryo , Flow Cytometry , Humans , In Vitro Techniques , Neovascularization, Physiologic , Rats , Real-Time Polymerase Chain Reaction , Surface Properties , Titanium , Umbilical Veins/cytology
11.
Proc Natl Acad Sci U S A ; 112(22): E2900-9, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25991856

ABSTRACT

The biological functions of VEGF-B in cancer progression remain poorly understood. Here, we report that VEGF-B promotes cancer metastasis through the remodeling of tumor microvasculature. Knockdown of VEGF-B in tumors resulted in increased perivascular cell coverage and impaired pulmonary metastasis of human melanomas. In contrast, the gain of VEGF-B function in tumors led to pseudonormalized tumor vasculatures that were highly leaky and poorly perfused. Tumors expressing high levels of VEGF-B were more metastatic, although primary tumor growth was largely impaired. Similarly, VEGF-B in a VEGF-A-null tumor resulted in attenuated primary tumor growth but substantial pulmonary metastases. VEGF-B also led to highly metastatic phenotypes in Vegfr1 tk(-/-) mice and mice treated with anti-VEGF-A. These data indicate that VEGF-B promotes cancer metastasis through a VEGF-A-independent mechanism. High expression levels of VEGF-B in two large-cohort studies of human patients with lung squamous cell carcinoma and melanoma correlated with poor survival. Taken together, our findings demonstrate that VEGF-B is a vascular remodeling factor promoting cancer metastasis and that targeting VEGF-B may be an important therapeutic approach for cancer metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Microvessels/drug effects , Neoplasm Metastasis/physiopathology , Neoplasms/blood supply , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/pharmacology , Animals , Blotting, Western , Cell Hypoxia , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Immunohistochemistry , Injections, Subcutaneous , Kaplan-Meier Estimate , Mice , Polymerase Chain Reaction , Vascular Endothelial Growth Factor B/administration & dosage , Zebrafish
12.
Graefes Arch Clin Exp Ophthalmol ; 253(6): 885-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25663437

ABSTRACT

PURPOSE: Inhibition of vascular endothelial growth factor (VEGF) is a promising strategy to treat retinal complications of diabetes. In contrast to VEGF-A binding ranibizumab, aflibercept also binds to other members of the VEGF family including VEGF-B, but potential effects of this factor on permeability and angiogenic processes are unclear. Therefore, we studied how VEGF-B variants as single agents or together with VEGF-A165 might affect proliferation, migration, or barrier function of retinal endothelial cells (REC). Also investigated was the normalization of REC properties with both VEGF-inhibitors to explore if additional targeting of VEGF-B is relevant. METHODS: Stimulation of proliferation or migration of immortalized bovine REC (iBREC) and disturbance of their barrier by exposure to VEGF-B variants (as single factors or together with VEGF-A165) was determined with or without VEGF-binding proteins being added. Permeability of iBREC was assessed by measuring their transendothelial resistance (TER) and expression of the tight junction protein claudin-1. RESULTS: VEGF-B167 and VEGF-B186 enhanced proliferation of iBREC but these isoforms did not affect cell migration. Interestingly, ranibizumab completely blocked both migration and proliferation induced by VEGF-A plus VEGF-B. Both VEGF-B variants did also not affect barrier function or claudin-1 expression in a normal or high-glucose environment. Accordingly, binding VEGF-A was enough to normalize a reduced TER and reinstate claudin-1 lost during treatment with this factor in combination with VEGF-B. CONCLUSIONS: Important properties and functions of REC seem not to be affected by any VEGF-B variant and targeting the key factor VEGF-A is sufficient to normalize growth factor-disturbed cells of this type.


Subject(s)
Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor B/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Blotting, Western , Cattle , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Claudin-1/metabolism , Electric Impedance , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fluorescent Antibody Technique, Indirect , Ranibizumab , Receptors, Vascular Endothelial Growth Factor/pharmacology , Recombinant Fusion Proteins/pharmacology , Retinal Vessels/cytology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
13.
J Biol Regul Homeost Agents ; 29(4): 835-40, 2015.
Article in English | MEDLINE | ID: mdl-26753644

ABSTRACT

Using a microelectrode technique we studied the effects of vascular endothelial growth factor-B on the activity of rat atrial myocardium under normal conditions and after gradual stretching of the tissue. It was shown that vascular endothelial growth factor-B increased duration of the action potential only at the level of 90% re-polarization. Effects on the frequency and force of contraction were absent. The repetition frequency of the action potentials did not change. Close observation of the vascular endothelial growth factor-B-induced mechanisms and stretch-induced alteration in action potential durations to 90% of repolarization, confirmed the existence of a link between the examining growth factor-B and stretch induced mechanisms.


Subject(s)
Heart Atria/drug effects , Vascular Endothelial Growth Factor B/pharmacology , Action Potentials/drug effects , Animals , Male , Microelectrodes , Rats , Rats, Wistar , Stress, Mechanical
14.
Proc Natl Acad Sci U S A ; 111(48): 17272-7, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404333

ABSTRACT

VEGF-B primarily provides neuroprotection and improves survival in CNS-derived neurons. However, its actions on the peripheral nervous system have been less characterized. We examined whether VEGF-B mediates peripheral nerve repair. We found that VEGF-B induced extensive neurite growth and branching in trigeminal ganglia neurons in a manner that required selective activation of transmembrane receptors and was distinct from VEGF-A-induced neuronal growth. VEGF-B-induced neurite elongation required PI3K and Notch signaling. In vivo, VEGF-B is required for normal nerve regeneration: mice lacking VEGF-B showed impaired nerve repair with concomitant impaired trophic function. VEGF-B treatment increased nerve regeneration, sensation recovery, and trophic functions of injured corneal peripheral nerves in VEGF-B-deficient and wild-type animals, without affecting uninjured nerves. These selective effects of VEGF-B on injured nerves and its lack of angiogenic activity makes VEGF-B a suitable therapeutic target to treat nerve injury.


Subject(s)
Cornea/drug effects , Nerve Regeneration/drug effects , Neurites/drug effects , Neurons/drug effects , Vascular Endothelial Growth Factor B/pharmacology , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cornea/innervation , Cornea/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Neurites/physiology , Neurons/metabolism , Neurons/physiology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/physiopathology , Peripheral Nerves/drug effects , Peripheral Nerves/pathology , Peripheral Nerves/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Trigeminal Ganglion/cytology , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
15.
Neuroscience ; 258: 385-400, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24291725

ABSTRACT

Vascular endothelial growth factor B (VEGF-B) has recently been shown to be a promising novel neuroprotective agent for several neurodegenerative conditions. In the current study we extended previous work on neuroprotective potential for Parkinson's disease (PD) by testing an expanded dose range of VEGF-B (1 and 10 µg) and directly comparing both neuroprotective and neurorestorative effects of VEGF-B in progressive unilateral 6-hydroxydopamine (6-OHDA) PD models to a single dose of glial cell line-derived neurotrophic factor (GDNF, 10 µg), that has been established by several groups as a standard in both preclinical PD models. In the amphetamine-induced rotational tests the treatment with 1 and 10 µg VEGF-B resulted in significantly improved motor function of 6-OHDA-lesioned rats compared to vehicle-treated 6-OHDA-lesioned rats in the neuroprotection paradigm. Both doses of VEGF-B caused an increase in tyrosine hydroxylase (TH)-positive cell and fiber count in the substantia nigra (SN) and striatum in the neuroprotective experiment. The effect size was comparable to the effects seen with GDNF. In the neurorestoration paradigm, VEGF-B injection had no significant effect in either the behavioral or the immunohistochemical analyses, whereas GDNF injection significantly improved the amphetamine-induced rotational behavior and reduced TH-positive neuronal cell loss in the SN. We also present a strong positive correlation (p=1.9e-50) of the expression of VEGF-B with nuclear-encoded mitochondrial genes involved in fatty acid metabolism in rat midbrain, pointing to the mitochondria as a site of action of VEGF-B. GDNF showed a positive correlation with nuclear-encoded mitochondrial genes that was not nearly as strong (p=0.018). VEGF-B counteracted rotenone-induced reduction of (a) fatty acid transport protein 1 and 4 levels and (b) both Akt protein and phosphorylation levels in SH-SY5Y cells. We further verified VEGF-B expression in the human SN pars compacta of healthy controls and PD patients, in neuronal cells that show co-expression with neuromelanin. These results have demonstrated that VEGF-B has potential as a neuroprotective agent for PD therapy and should be further investigated.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Nootropic Agents/pharmacology , Parkinson Disease/drug therapy , Vascular Endothelial Growth Factor B/pharmacology , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Disease Models, Animal , Humans , Male , Motor Activity/drug effects , Neurons/drug effects , Neurons/physiology , Oxidopamine , Parkinson Disease/physiopathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/physiopathology
16.
Exp Eye Res ; 109: 17-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23370270

ABSTRACT

A key model for examining the activity-dependent development of primary visual cortex (V1) involves the imbalance in activity between the two eyes induced by monocular deprivation (MD). MD early in life causes dramatic changes in the functional and structural organization of mammalian visual cortex. The molecular signals that mediate the effects of MD on the development of visual cortex are not well defined. Neurotrophic factors are important in regulating the plasticity of visual cortex, but the choice of an appropriate growth factor as well as its delivery has proven difficult. Although vascular endothelial growth factor-B (VEGF-B) is a homolog of the angiogenic factor VEGF-A, it has only minimal angiogenic activity, raising the question of whether this factor has other (more relevant) biological properties. Intrigued by the possibility that VEGF family members affect neuronal cells, we explored whether VEGF-B has a role in the nervous system. In rats, VEGF-B infusion during monocular deprivation (MD) counteracted the normally occurring ocular dominance (OD) shift toward the non-deprived eye so that the deprived eye dominated the VEGF-B-treated cortex after MD. In particular, VEGF-B counteracted the effects of MD without causing detectable alterations in spontaneous discharge or behavior. In conclusion, the simultaneous analysis of visual cortical cell discharge and ocular dominance plasticity suggests that VEGF-B has important effects on the functional architecture of the visual cortex. Therefore, VEGF-B is a new candidate trophic challenge molecule for the visual cortex.


Subject(s)
Dominance, Ocular/physiology , Vascular Endothelial Growth Factor B/physiology , Vision, Binocular/physiology , Vision, Monocular/physiology , Visual Cortex/growth & development , Visual Cortex/physiology , Action Potentials/physiology , Animals , Critical Period, Psychological , Dominance, Ocular/drug effects , Electrodes, Implanted , Models, Neurological , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Rats , Rats, Long-Evans , Sensory Deprivation/physiology , Vascular Endothelial Growth Factor B/pharmacology , Vision, Binocular/drug effects , Vision, Monocular/drug effects , Visual Cortex/drug effects , Visual Pathways/drug effects , Visual Pathways/growth & development , Visual Pathways/physiology
17.
Circ Res ; 109(3): e14-26, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21636803

ABSTRACT

RATIONALE: Systemic sclerosis (SSc) is characterized by widespread microangiopathy, fibrosis, and autoimmunity. Despite the lack of angiogenesis, the expression of vascular endothelial growth factor A (VEGF) was shown to be upregulated in SSc skin and circulation; however, previous studies did not distinguish between proangiogenic VEGF(165) and antiangiogenic VEGF(165)b isoforms, which are generated by alternative splicing in the terminal exon of VEGF pre-RNA. OBJECTIVE: We investigated whether VEGF isoform expression could be altered in skin and circulation of patients with SSc. METHODS AND RESULTS: Here, we show that the endogenous antiangiogenic VEGF(165)b splice variant is selectively overexpressed at both the mRNA and protein levels in SSc skin. Elevated VEGF(165)b expression correlated with increased expression of profibrotic transforming growth factor-ß1 and serine/arginine protein 55 splicing factor in keratinocytes, fibroblasts, endothelial cells, and perivascular inflammatory cells. Circulating levels of VEGF(165)b were significantly higher in patients with SSc than in control subjects. Microvascular endothelial cells (MVECs) isolated from SSc skin expressed and released higher levels of VEGF(165)b than healthy MVECs. Transforming growth factor-ß1 upregulated the expression of VEGF(165)b and serine/arginine protein 55 in both SSc and healthy MVECs. In SSc MVECs, VEGF receptor-2 was overexpressed, but its phosphorylation was impaired. Recombinant VEGF(165)b and SSc-MVEC-conditioned medium inhibited VEGF(165)-mediated VEGF receptor-2 phosphorylation and capillary morphogenesis in healthy MVECs. The addition of anti-VEGF(165)b blocking antibodies abrogated the antiangiogenic effect of SSc-MVEC-conditioned medium. Capillary morphogenesis was severely impaired in SSc MVECs and could be ameliorated by treatment with recombinant VEGF(165) and anti-VEGF(165)b blocking antibodies. CONCLUSIONS: In SSc, a switch from proangiogenic to antiangiogenic VEGF isoforms may have a crucial role in the insufficient angiogenic response to chronic ischemia.


Subject(s)
Alternative Splicing/physiology , Endothelial Cells/physiology , Neovascularization, Pathologic/physiopathology , Scleroderma, Systemic/physiopathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor B/genetics , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dermis/blood supply , Endothelial Cells/cytology , Gene Expression/drug effects , Gene Expression/physiology , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , RNA-Binding Proteins , Scleroderma, Systemic/genetics , Scleroderma, Systemic/metabolism , Serine-Arginine Splicing Factors , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
J Invest Surg ; 24(2): 67-76, 2011.
Article in English | MEDLINE | ID: mdl-21345006

ABSTRACT

PURPOSE: To explore the molecular mechanism of Vascular endothelial growth factor receptor-1 (VEGFR-1) in invasion and metastasis of hepatocellular carcinoma. METHODS: Reverse transcription polymerase chain reaction was performed to test expression of VEGFR-1 and its ligand VEGF-B19 in four hepatoma carcinoma cell. Fluorescent immunohistochemistry and western blotting were used to test the change of expression of E-cadherin or α-catenin. RESULTS: VEGF-B-treated cells exhibited a change in E-cadherin from an organized, membrane-bound structure to a disorganized state that was dispersed throughout the cytoplasm. The maximal changes in E-cadherin were observed 24 hr after treatment of cells with VEGF-B. α-catenin was observed to translocate to the nucleus from its usual membrane-bound location 24 hr after treatment with either VEGF-B. Expression of the epithelial adhesion molecules E-cadherin was observed to decrease 48 hours after VEGF-B treatment. The nuclear expression of α-catenin was observed to increase 24 hr after treatment with VEGF-B. CONCLUSIONS: VEGFR-1 on tumor cells may contribute to the aggressive behavior of hepatocellular carcinoma cells by inducing epithelial to mesenchymal transition (EMT). Targeting VEGFR-1 and downstream mediators of EMT may provide the foundation for the development of novel therapeutic approaches for this morbid and lethal disease.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-1/physiology , Cadherins/metabolism , Cell Line, Tumor , Humans , Vascular Endothelial Growth Factor B/pharmacology , alpha Catenin/metabolism
19.
Cardiovasc Res ; 86(1): 122-30, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19955220

ABSTRACT

AIMS: Vascular endothelial growth factors (VEGFs) and their receptors (VEGF-Rs) are among the most powerful factors regulating vascular growth. However, it has remained unknown whether stimulation of VEGF-R1, VEGF-R2 or both of the receptors produces the best angiogenic responses in myocardium. The aim of this study was to compare the VEGF-R1-specific ligand VEGF-B(186), VEGF-R2-specific ligand VEGF-E and VEGF-A(165,) which stimulates both receptors, regarding their effects on angiogenesis and left ventricular function in mice. METHODS AND RESULTS: High-resolution echocardiography was used to guide the closed-chest injections of adenoviral (Ad) vectors expressing VEGF-B(186,) VEGF-E, and VEGF-A(165) into the anterior wall of the left ventricle in C57Bl/6J mice. Angiogenic and functional effects were analysed using histology, ultrasound and perfusion analyses 6 (D6) and 14 (D14) days after the Ad injection. AdVEGF-A(165) induced a strong angiogenic response seen as an enlargement of myocardial capillaries whereas angiogenesis induced by AdVEGF-B(186) and AdVEGF-E seemed more physiological. The increase in the capillary area was accompanied with an increase in myocardial perfusion at D6 after the gene injection. AdVEGF-A(165) and AdVEGF-E induced endothelial-specific proliferation whereas AdVEGF-B(186) mostly induced proliferation of cardiomyocytes. AdVEGF-A(165) induced more pronounced tissue damage than AdVEGF-B(186) and AdVEGF-E. Left ventricular function measured as ejection fraction did not change during the follow-up. AdVEGF-A(165) increased both VEGF-R1 and VEGF-R2 protein expression whereas AdVEGF-B(186) and AdVEGF-E did not affect endogenous receptor expression levels. CONCLUSION: AdVEGF-B(186) and AdVEGF-E are equally potent in inducing therapeutic angiogenesis in mouse myocardium and produce less side effects than AdVEGF-A(165).


Subject(s)
Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Function, Left/physiology , Adenoviridae/genetics , Animals , Blotting, Western , Cell Division/drug effects , Cell Division/physiology , Coronary Circulation/drug effects , Coronary Circulation/physiology , Echocardiography , Enzyme-Linked Immunosorbent Assay , Ligands , Male , Mice , Mice, Inbred C57BL , Models, Cardiovascular , Myocarditis/chemically induced , Myocarditis/metabolism , Myocarditis/physiopathology , Neovascularization, Physiologic/drug effects , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Stroke Volume/drug effects , Stroke Volume/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/pharmacology , Ventricular Function, Left/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/pharmacology
20.
Am J Physiol Renal Physiol ; 297(6): F1656-67, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19828679

ABSTRACT

Podocytes are an important component of the glomerular filtration barrier and are the major source of vascular endothelial growth factor (VEGF) in the glomerulus. The role of VEGF for the phenotype of the glomerular endothelium has been intensely studied; however, the direct effects of autocrine VEGF on the podocyte are largely unknown. In this study we characterized the expression of VEGF isoforms and VEGF receptors in cultured human podocytes and examined direct effects on cell signaling and apoptosis after stimulation with exogenous VEGF or ablation of autocrine VEGF. We identified VEGF-A and VEGF-C as the dominant isoforms in human podocytes and showed that autocrine levels of both are important for the intracellular activation of antiapoptotic phosphoinositol 3-kinase/AKT and suppression of the proapoptotic p38MAPK via VEGFR-2. We demonstrated that ablation of VEGF-A or VEGF-C as well as treatment with bevacizumab or a VEGFR-2/-3 tyrosine kinase inhibitor led to reduced podocyte survival. In contrast, ablation of VEGF-B had no effect on podocyte survival. Treatment with exogenous VEGF-C reversed the effect of VEGF-A neutralization, and exogenous VEGF-A abrogated the effect of VEGF-C ablation in human podocytes. Our results underline the importance of autocrine VEGF for podocyte survival and indicate the delicate balance of VEGF-A and VEGF-C to influence progression of glomerular diseases.


Subject(s)
Autocrine Communication/physiology , Podocytes/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Apoptosis/physiology , Bevacizumab , Cell Differentiation , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , Enzyme Activation/drug effects , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Podocytes/cytology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor B/pharmacology , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...