Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Protein J ; 43(4): 697-710, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39014261

ABSTRACT

The interaction between vascular endothelial growth factor A (VEGFA) and VEGF receptor 1(VEGFR1) is a central focus for drug development in pathological angiogenesis, where aberrant angiogenesis underlies various anomalies necessitating therapeutic intervention. Identifying hotspots of these proteins is crucial for developing new therapeutics. Although machine learning techniques have succeeded significantly in prediction tasks, they struggle to pinpoint hotspots linked to angiogenic activity accurately. This study involves the collection of diverse VEGFA and VEGFR1 protein sequences from various species via the UniProt database. Electron-ion interaction Potential (EIIP) values were assigned to individual amino acids and transformed into frequency-domain representations using discrete Fast Fourier Transform (FFT). A consensus spectrum emerged by consolidating FFT data from multiple sequences, unveiling specific characteristic frequencies. Subsequently, the Stockwell Transform (ST) was employed to yield the hotspots. The Resonant Recognition Model (RRM) identified a characteristic frequency of 0.128007 with an associated wavelength of 1570 nm and RRM-ST identified hotspots for VEGFA (Human 36, 46, 48, 67, 71, 74, 82, 86, 89, 93) and VEGFR1 (Human 224, 259, 263, 290, 807, 841, 877, 881, 885, 892, 894, 909, 913, 1018, 1022, 1026, 1043). These findings were cross-validated by Hotspots Wizard 3.0 webserver and Protein Data Bank (PDB). The study proposes using a 1570 nm wavelength for photo bio modulation to boost VEGFA/VEGFR1 interaction in the condition that is needed. It also aims to reduce VEGFA/VEGFR2 interaction, limiting harmful angiogenesis in conditions like diabetic retinopathy. Also, the identified hotspots assist in designing agonistic or antagonistic peptides tailored to specific medical requirements with abnormal angiogenesis.


Subject(s)
Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/genetics , Humans , Neovascularization, Pathologic/metabolism , Protein Binding , Angiogenesis
2.
J Coll Physicians Surg Pak ; 33(12): 1395-1399, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38062595

ABSTRACT

OBJECTIVE: To determine the reference interval of soluble FMS-like tyrosine kinase-1 (sFIt-1) in healthy, non-pregnant and pregnant females. STUDY DESIGN: Observational study. Place and Duration of the Study: Department of Chemical Pathology, Chughtai Institute of Pathology, Lahore, from January to May 2023. METHODOLOGY: Blood samples were collected from 120 disease-free non-pregnant females of reproductive age group and 120 disease-free pregnant females with singleton fetuses from 15 to 28 weeks of gestational age. Healthy reference individuals were selected by correlating history with medical disorders like diabetes mellitus, hypertension, autoimmune diseases, inherited disorders, and by excluding any other drug history. All findings were recorded on health screening questionnaire. Levels of sFlt-1 were measured by a fully automated immunoassay analyser Cobas e601. Kolmogorov-Smirnov test was applied. The value of p <0.05 was considered significant. The 2.5th and 97.5th percentiles were computed at 90% CI by using the formula 0.025x (n+1) and 0.975x (n+1) which corresponded to rank number 1 and 7, respectively. The reference interval was calculated by the Rank-based method. RESULTS: Reference interval of sFlt-1 in non-pregnant and pregnant females were determined on the basis of 2.5th and 97.5th percentiles which were 57.7 to 118.5 pg/mL and 563.5 to 3288.0 pg/mL, respectively. CONCLUSION: The present study determined reference interval of sFlt-1 in healthy, non-pregnant and pregnant females in Lahore. KEY WORDS: Reference interval, Soluble FMS-like tyrosine kinase-1, Pre-eclampsia, Rank-based method.


Subject(s)
Hypertension , Pre-Eclampsia , Vascular Endothelial Growth Factor Receptor-1 , Child, Preschool , Female , Humans , Pregnancy , Biomarkers , Placenta Growth Factor , Pre-Eclampsia/diagnosis , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/chemistry
3.
J Nanobiotechnology ; 20(1): 7, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34983556

ABSTRACT

BACKGROUND: Inhibition of tumor angiogenesis through simultaneous targeting of vascular endothelial growth factor receptor (VEGFR)-1 and -2 is highly efficacious. An antagonist peptide of VEGFA/VEGFB, referred to as VGB3, can recognize and neutralize both VEGFR1 and VEGFR2 on the endothelial and tumoral cells, thereby inhibits angiogenesis and tumor growth. However, improved efficacy and extending injection intervals is required for its clinical translation. Given that gold nanoparticles (GNPs) can enhance the efficacy of biotherapeutics, we conjugated VGB3 to GNPs to enhance its efficacy and extends the intervals between treatments without adverse effects. RESULTS: GNP-VGB3 bound to VEGFR1 and VEGFR2 in human umbilical vein endothelial (HUVE) and 4T1 mammary carcinoma cells. GNP-VGB3 induced cell cycle arrest, ROS overproduction and apoptosis and inhibited proliferation and migration of endothelial and tumor cells more effectively than unconjugated VGB3 or GNP. In a murine 4T1 mammary carcinoma tumor model, GNP-VGB3 more strongly than VGB3 and GNP inhibited tumor growth and metastasis, and increased animal survival without causing weight loss. The superior antitumor effects were associated with durable targeting of VEGFR1 and VEGFR2, thereby inhibiting signaling pathways of proliferation, migration, differentiation, epithelial-to-mesenchymal transition, and survival in tumor tissues. MicroCT imaging and inductively coupled plasma mass spectrometry showed that GNP-VGB3 specifically target tumors and exhibit greater accumulation within tumors than the free GNPs. CONCLUSION: Conjugation to GNPs not only improved the efficacy of VGB3 peptide but also extended the intervals between treatments without adverse effects. These results suggest that GNP-VGB3 is a promising candidate for clinical translation.


Subject(s)
Angiogenesis Inhibitors , Gold/chemistry , Metal Nanoparticles/chemistry , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factor Receptor-2 , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Circ Genom Precis Med ; 14(2): e003196, 2021 04.
Article in English | MEDLINE | ID: mdl-33625251

ABSTRACT

BACKGROUND: Randomized clinical trials indicate that the immune response plays a significant role in coronary artery disease (CAD), a disorder impacting the lifespan potential. However, the identification of targets critical to the immune response in atheroma is still hampered by a lack of solid inference. METHODS: Herein, we implemented a system genetics approach to identify causally associated immune targets implicated in atheroma. We leveraged genome-wide association studies to perform mapping and Mendelian randomization to assess causal associations between gene expression in blood cells with CAD and the lifespan. Expressed genes (eGenes) were prioritized in network and in single-cell expression derived from plaque immune cells. RESULTS: Among 840 CAD-associated blood eGenes, 37 were predicted causally associated with CAD and 6 were also associated with the parental lifespan in Mendelian randomization. In multivariable Mendelian randomization, the impact of eGenes on the lifespan potential was mediated by the CAD risk. Predicted causal eGenes were central in network. FLT1 and CCR5 were identified as targets of approved drugs, whereas 22 eGenes were deemed tractable for the development of small molecules and antibodies. Analyses of plaque immune single-cell expression identified predicted causal eGenes enriched in macrophages (GPX1, C4orf3) and involved in ligand-receptor interactions (CCR5). CONCLUSIONS: We identified 37 blood eGenes predicted causally associated with CAD. The predicted expression for 6 eGenes impacted the lifespan potential through the risk of CAD. Prioritization based on network, annotations, and single-cell expression identified targets deemed tractable for the development of drugs and for drug repurposing.


Subject(s)
Coronary Artery Disease/genetics , Longevity , Antibodies/immunology , Coronary Artery Disease/pathology , Genome-Wide Association Study , Glutathione Peroxidase/genetics , Humans , Ligands , Macrophages/cytology , Macrophages/metabolism , Mendelian Randomization Analysis , Odds Ratio , Protein Interaction Maps/genetics , Quantitative Trait Loci , Receptors, CCR5/chemistry , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Risk Factors , Single-Cell Analysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Glutathione Peroxidase GPX1
5.
Curr Med Sci ; 40(4): 671-676, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32862377

ABSTRACT

Acute respiratory distress syndrome (ARDS) is associated with a mortality of 45%. Our previous research indicated that anti-vascular endothelial growth factor (VEGF) could maintain the normal structure and function of the respiratory barrier. However, systemic application of VEGF antagonists would lead to animal death. This study attempts to study the targeted drug delivery for ARDS. In this study, we used soluble fms-like tyrosine kinase-1 (sFlt)-targeted ultrasound microbubbles to antagonize the effect of VEGF on lung tissue. Ninety male BALB/c mice were randomly assigned to 6 groups: phosphate buffer saline (PBS) group (PBS+PBS); blank group (PBS+empty microbubbles); lipopolysaccharide (LPS) group (LPS+PBS); ARDS group (LPS+empty microbubbles); control group (PBS+sFlt microbubbles); and treatment group (LPS+sFlt microbubbles). After administration of LPS or PBS in the corresponding groups, the sFlt-targeted microbubbles or empty microbubbles were injected into the blood circulation. Then the lungs were irradiated with ultrasound, which ruptured the drug-loaded microbubbles and helped release drugs to the lung tissues targeted. The lung injury score, lung wet/dry ratio (W/D), liver and kidney functions, and the mortality of the mice in all groups were investigated at the predetermined time point. The difference in mortality between groups was examined by Fisher test. Other data were analyzed by one-way analysis of variance (ANOVA). A value of P<0.05 indicates that the difference was significant. The results showed that the PaO2 levels were normal in the PBS group, the blank group, and the control group. The LPS group and ARDS group showed significant hypoxia. PaO2 was improved significantly in the treatment group. The lung injury score and W/D were normal in the PBS group, the blank group, and the control group. The lung injury score and W/D increased significantly in the LPS group and ARDS group and decreased in the treatment group (P<0.05). The mortality rate of the ARDS model was 60% (95% confidence interval 47.5%-72.5%), and that with sFlt-targeted microbubbles was significantly lower at only 40% (95% confidence interval 27.5%-52.5%, P<0.05). It was concluded that anti-VEGF with sFlt targeted ultrasound microbubbles attenuated the lung injury and ultimately reduced the 7-day mortality effectively. It might be a suitable therapeutic tool for the treatment of ARDS.


Subject(s)
Lipopolysaccharides/adverse effects , Respiratory Distress Syndrome/drug therapy , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/administration & dosage , Animals , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred BALB C , Microbubbles , Random Allocation , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/mortality , Treatment Outcome , Ultrasonic Waves , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/pharmacology
6.
Sci Rep ; 10(1): 7308, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355171

ABSTRACT

Recent treatment developments for metastatic renal cell carcinoma offer combinations of immunotherapies or immunotherapy associated with tyrosine kinase inhibitors (TKI). There is currently no argument to choose one solution or another. Easy-to-use markers to assess longitudinal responses to TKI are necessary to determine when to switch to immunotherapies. These new markers will enable an earlier adaptation of therapeutic strategy in order to prevent tumor development, unnecessary toxicity and financial costs. This study evaluates the potential of ultrasound molecular imaging to track the response to sunitinib in a clear cell renal carcinoma model (ccRCC). We used a patient-derived xenograft model for this imaging study. Mice harboring human ccRCC were randomized for sunitinib treatment vs. control. The tumors were imaged at days 0, 7, 14 and 28 with ultrasound molecular imaging. Signal enhancement was quantified and compared between the two groups after injections of non-targeted microbubbles and microbubbles targeting VEGFR1 and FSHR. The tumor growth of the sunitinib group was significantly slower. There was a significantly lower expression of both VEGFR-1 and FSHR molecular ultrasound imaging signals in the sunitinib group at all times of treatment (Days 7, 14 and 28). These results confirm the study hypothesis. There was no significant difference between the 2 groups for the non-targeted microbubble ultrasound signal. This study demonstrated for the first time the potential of VEGFR1 and FSHR, by ultrasound-based molecular imaging, to follow-up the longitudinal response to sunitinib in ccRCC. These results should trigger developments for clinical applications.


Subject(s)
Carcinoma, Renal Cell/diagnostic imaging , Kidney Neoplasms/diagnostic imaging , Microbubbles , Receptors, FSH/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Animals , Carcinoma, Renal Cell/drug therapy , Female , Humans , Immunotherapy , Kidney Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Molecular Imaging , Neoplasm Transplantation , Perfusion , Protein Kinase Inhibitors/therapeutic use , Sunitinib/therapeutic use , Ultrasonography
7.
BMC Cancer ; 20(1): 196, 2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32164575

ABSTRACT

BACKGROUND: The expression of vascular endothelial growth factor (VEGF)-A/ VAGF receptors (VEGFRs) signaling plays a pivotal role in the tumor angiogenesis and the development of the immunosuppressive tumor microenvironment in glioblastomas. We have previously conducted exploratory clinical studies investigating VEGFRs peptide vaccination with and without multiple glioma oncoantigens in patients with recurrent high-grade gliomas. Recently, an exploratory clinical investigation of VEGFRs peptide vaccination was conducted in patients with progressive neurofibromatosis type 2. Those studies suggested that cytotoxic T lymphocytes (CTLs) induced by the vaccination can directly kill a wide variety of cells associated with tumor growth, including tumor vessels, tumor cells, and immunosuppressive cells expressing VEGFR1 and/or 2. In the present study, synergistic activity of the combination of VEGFRs peptide vaccination with chemotherapy was evaluated. METHODS: We performed the first clinical trial to assess VEGFR1 and 2 vaccination along with temozolomide (TMZ) -based chemoradiotherapy for the patients with primary glioblastomas. Furthermore, histopathological changes after the vaccination were evaluated using paired pre- and post- vaccination specimens. RESULTS: The disappearance of radiographically enhanced lesion was observed in 2 patients after the vaccination, including one in which the methylation of the O6-methylguanine-DNA methyltransferase (MGMT) promoter was not observed. The histopathological findings of pre- and post-vaccination specimens demonstrated that tumor vessels showed negative or slight VEGFRs expressions after the vaccination and most endothelial cells were covered with PDGFR-ß-positive pericytes. Notably, CTLs induced by VEGFRs peptide vaccination attacked not only tumor vessels but also tumor cells and regulatory T cells expressing VEGFRs even in recurrent tumors. CONCLUSIONS: VEGFR1 and 2 vaccination may have a preliminary synergistic effect when administered with TMZ. The limitation of the present study was the paucity of the number of the samples. Further studies involving more patients are warranted to confirm the findings of this study. TRIAL REGISTRATION: This study was registered as UMIN000013381 (University Hospital Medical Information Network-Clinical Trial Registry: UMIN-CTR) on 5 March, 2014 and with the Japan Registry of Clinical Trials (jRCT) as jRCTs031180170 on 1 March, 2019.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Glioblastoma/drug therapy , Peptide Fragments/administration & dosage , Temozolomide/administration & dosage , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Cancer Vaccines/pharmacology , Drug Synergism , Feasibility Studies , Female , Humans , Male , Middle Aged , Peptide Fragments/therapeutic use , Survival Analysis , Temozolomide/therapeutic use , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry
8.
Mol Biol Rep ; 47(3): 2061-2071, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32072404

ABSTRACT

Tumor angiogenesis allows tumor cells to grow and migrate toward the bloodstream and initiate metastasis. The interactions of vascular endothelial growth factors (VEGF) A and B, as the important regulating factors for blood vessel growth, with VEGFR1 and VEGFR2 trigger angiogenesis process. Thus, preventing these interactions led to the effective blockade of VEGF/VEGFRs signaling pathways. In this study, the inhibitory effect of a 23-mer linear peptide (VGB4), which binds to both VEGFR1 and VEGFR2, on VEGF-stimulated Human Umbilical Vein Endothelial Cells (HUVECs) and highly metastatic human breast cancer cell MDA-MB-231 proliferation was examined using MTT assay. To assess the anti-migratory potential of VGB4, HUVECs and also MDA-MB-231 cells wound healing assay was carried out at 48 and 72 h. In addition, downstream signaling pathways of VEGF associated with cell migration and invasion were investigated by quantification of mRNA and protein expression using real-time quantitative PCR and western blot in 4T1 tumor tissues and MDA-MB-231 cells. The results revealed that VGB4 significantly impeded proliferation of HUVECs and MDA-MB-231 cells, in a dose- and time-dependent manner, and migration of HUVECs and MDA-MB-231 cells for a prolonged time. We also observed statistically significant reduction of the transcripts and protein levels of focal adhesion kinase (FAK), Paxillin, matrix metalloproteinase-2 (MMP-2), RAS-related C3 botulinum substrate 1 (Rac1), P21-activated kinase-2 (PAK-2) and Cofilin-1 in VGB4-treated 4T1 tumor tissues compared to controls. The protein levels of phospho-VEGFR1, phospho-VEGFR2, Vimentin, ß-catenin and Snail were markedly decreased in both VGB4-treated MDA-MB-231 cells and VGB4-treated 4T1 tumor tissues compared to controls as evidenced by western blotting. These results, in addition to our previous studies, confirm that dual blockage of VEGFR1 and VEGFR2, due to the inactivation of diverse signaling mediators, effectively suppresses tumor growth and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Biomarkers, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Gene Expression , Humans , Mice , Molecular Targeted Therapy , Peptides , RNA, Messenger/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/chemistry , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
9.
Sci Rep ; 9(1): 15571, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666542

ABSTRACT

Soluble Flt-1 (sFlt-1), an endogenous antagonist of the proatherogenic cytokine placental growth factor, is decreased in chronic kidney disease (CKD), leading to atherosclerotic progression. In this study, we investigated the effect of AST-120, an oral carbon adsorbent which can remove uremic toxins, on sFlt-1 expression levels and atherosclerosis progression. Atherosclerotic apolipoprotein E-deficient mice underwent a 5/6 nephrectomy (5/6 NR) or a sham operation (sham) at 8 weeks of age and were then treated or not with oral AST-120 for 12 weeks. sFlt-1 expression levels and the degree of atherosclerosis were assessed at 22 weeks of age in each of the four groups (sham; n = 7, 5/6 NR; n = 10, sham + AST-120: n = 8, 5/6 NR + AST-120; n = 8). The expression levels of sFlt-1 mRNA in the kidney were significantly lower in the 5/6 NR group than in the sham group, but AST-120 treatment prevented this decrease in sFlt-1 levels. Similarly, the atherosclerotic plaque area of the thoracoabdominal aorta was significantly larger in the 5/6 NR group than in the sham group, and AST-120 treatment prevented this increase in atherosclerosis. AST-120 could, therefore, be used as a therapeutic to treat atherosclerosis in patients with CKD.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/pathology , Carbon/pharmacology , Gene Expression Regulation/drug effects , Kidney Failure, Chronic/complications , Oxides/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics , Administration, Oral , Animals , Atherosclerosis/complications , Carbon/administration & dosage , Disease Models, Animal , Disease Progression , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Oxides/administration & dosage , RNA, Messenger/genetics , Solubility , Vascular Endothelial Growth Factor Receptor-1/chemistry
10.
Molecules ; 24(21)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671706

ABSTRACT

A biosimilar fusion protein VEGFR-IgG consisting of vascular endothelial growth factor receptors 1 and 2 (VEGFR-1, VEGFR-2) and the Fc portion of human IgG1 was prepared for this study. The prepared fusion protein was expected to possess a total of five N-linked glycosylation sites: two sites in the VEGFR-1 region, two sites in the VEGFR-2 region, and one site in the human IgG Fc region. For site-specific glycan analysis, the fusion protein was hydrolyzed with trypsin, and the resulting tryptic digests were analyzed by liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI MS/MS). The expected N-linked glycosylation sites were successfully identified and site-specific glycopeptide mapping was completed by Integrated GlycoProteome Analyzer (I-GPA) for the resulting raw tandem mass data. Finally, it was clearly confirmed that N-linked glycans for each glycosylation site showed significantly different patterns in microheterogeneity, which may indicate certain functions for each glycosylation site in the protein. Based on the mapping results, the unique features in glycan microheterogeneity for the five glycosylation sites of VEGFR-IgG fusion protein were compared site-specifically and further discussed to understand the functional meaning of each glycosylation pattern.


Subject(s)
Glycopeptides/chemistry , Immunoglobulin G/chemistry , Polysaccharides/chemistry , Recombinant Fusion Proteins/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Amino Acid Sequence , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Sci Rep ; 9(1): 6810, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31048752

ABSTRACT

The toxic manifestations of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an environmental contaminant, primarily depend on its ability to activate aryl hydrocarbon receptor (AhR), which is a ligand-dependent transcription factor belonging to the superfamily of basic-helix-loop-helix DNA-binding proteins. In the present study, we aimed to identify novel protein receptor targets for TCDD using computational and in vitro validation experiments. Interestingly, results from computational methods predicted that Vascular Endothelial Growth Factor Receptor 1 (VEGFR1) could be one of the potential targets for TCDD in both mouse and humans. Results from molecular docking studies showed that human VEGFR1 (hVEGFR1) has less affinity towards TCDD compared to the mouse VEGFR1 (mVEGFR1). In vitro validation results showed that TCDD can bind and phosphorylate hVEGFR1. Further, results from molecular dynamic simulation studies showed that hVEGFR1 interaction with TCDD is stable throughout the simulation time. Overall, the present study has identified VEGFR1 as a novel target for TCDD, which provides the basis for further elucidating the role of TCDD in angiogenesis.


Subject(s)
Computational Biology , Models, Molecular , Polychlorinated Dibenzodioxins/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Amino Acids , Animals , Binding Sites , Humans , Ligands , Mice , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Phosphorylation , Polychlorinated Dibenzodioxins/pharmacology , Protein Binding , Quantitative Structure-Activity Relationship , Reproducibility of Results , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
12.
J Pept Sci ; 25(5): e3146, 2019 May.
Article in English | MEDLINE | ID: mdl-30652389

ABSTRACT

The placental growth factor (PlGF), a member of VEGF family, plays a crucial role in pathological angiogenesis, especially ischemia, inflammation, and cancer. This activity is mediated by its selective binding to VEGF receptor 1 (VEGFR-1), which occurs predominantly through receptor domains 2 and 3. The PlGF ß-hairpin region spanning residues Q87 to V100 is one of the key binding elements on the protein side. We have undertaken a study on the design, preparation, and functional characterization of the peptide reproducing this region and of a set of analogues where glycine 94, occurring at the corner of the hairpin in the native protein, is replaced by charged as well as hydrophobic residues. Also, some peptides with arginine 96 replaced by other residues have been studied. We find that the parent peptide weakly binds VEGFR-1, but replacement of G94 with residues bearing H-bond donating residues significantly improves the affinity. Replacement of R96 instead blocks the interaction between the peptide and the domain. The strongest affinity is observed with the G94H (peptide PlGF-2) and G94W (peptide PlGF-10) mutants, while the peptide PlGF-8, bearing the R96G mutation, is totally inactive. The PlGF-1 and PlGF-2 peptides also bind the VEGFR-2 receptors, though with a reduced affinity, and are able to interfere with the VEGF-induced receptor signaling on endothelial cells. The peptides also bind VEGFR-2 on the surface of cells, while PlGF-8 is inactive. Data suggest that these peptides have potential applications as PlGF/VEGF mimic in various experimental settings.


Subject(s)
Human Umbilical Vein Endothelial Cells/chemistry , Membrane Proteins/chemistry , Peptides/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry , Binding Sites , Cell Proliferation , Endothelial Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Membrane Proteins/metabolism , Peptides/metabolism , Surface Properties , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Pak J Biol Sci ; 22(10): 502-509, 2019 Jan.
Article in English | MEDLINE | ID: mdl-31930840

ABSTRACT

BACKGROUND AND OBJECTIVE: The waste products of the tamarind canning industry have been discarded; however, it has potential health benefits. Herein, the study was carried out HPLC profiling of phenolic constituents of Tamarindis indica pericarp and seeds. Furthermore, the cytotoxic activity against HUH-7 cells was evaluated and assessed with molecular docking study on angiogenesis-related VEGF kinase proteins in addition to evaluating the level of released VEGF in treated HUH-7 cells by ELISA. MATERIALS AND METHODS: Folin-ciocalteu and AlCl3 assays were used for quantification of total phenolics (TPC) and total flavonoids (TFC) contents, respectively. Molecular docking study was done on VEGF kinase proteins. RESULTS: TPC and TFC of pericarp and seeds were 0.35±0.02 g GAE g-1 DE and 0.12±0.009 g CE g-1 DE, 0.39±0.01 g GAE g-1 DE and 0.03±0.006 g CE g-1 DE, respectively. In pericarp, 8 phenolics were tentatively identified, where (+)-catechin was the major (27,386.04 µg g-1 DE) followed by gallic acid and naringenin (931.47, 500.42 µg g-1 DE) respectively. While in seeds, 11 phenolics were tentatively identified, where naringenin was the major (95,305.47 µg g-1 DE) followed by (+)-catechin and rutin (54,930.29, 15,361.66 µg g-1 DE) respectively. Aqueous and methanol seeds extracts exhibit cytotoxic effect with IC50 27.4±1.81 and 13.4±0.94 µg mL-1, respectively, it was more potent than aqueous and methanol pericarp extracts which had IC50 132±5.82 and 61.6±3.16 µg mL-1. The tested phenolics were fit on the active sites of VEGF kinase targets with varied degree of interactions. The cytotoxic and anti-angiogenic activities were confirmed in light of phenolics docking interactions. CONCLUSION: Results demonstrate for the first time that phenolics could inhibit angiogenesis via inhibiting kinase proteins, which could therefore be developed as antiangiogenic drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Phenols/pharmacology , Tamarindus/chemistry , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Catechin/chemistry , Cell Line, Tumor , Cell Survival , Enzyme-Linked Immunosorbent Assay , Flavanones/chemistry , Flavonoids/chemistry , Gallic Acid/chemistry , Humans , Methanol/chemistry , Molecular Docking Simulation , Plant Extracts/chemistry , Protein Conformation , Rutin/chemistry , Seeds
14.
Acta Histochem ; 121(1): 94-100, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30442382

ABSTRACT

The vomeronasal system (VNS) is an accessory olfactory structure present in most mammals adhibited to the detection of specific chemosignals implied in social and reproductive behavior. The VNS comprises the vomeronasal organ (VNO), vomeronasal nerve and accessory olfactory bulb. VNO is characterized by a neuroepithelium constituted by bipolar neurons and supporting and stem/progenitor cells. In humans, VNO is present during fetal life and is supposed to possess chemoreceptor activity and participate in gonadotropin-releasing hormone neuronal precursor migration toward the hypothalamus. Instead, the existence and functions of VNO in postnatal life is debated. Vascular endothelial growth factor (VEGF) and its receptors (VEGFRs) have been demonstrated to play fundamental roles in various neurogenic events. However, there are no data regarding the localization and possible function of VEGF/VEGFRs in human fetal VNO. Therefore, this study was conceived to investigate the expression of VEGF/VEGFRs in human VNO in an early developmental period (9-12 weeks of gestation), when this organ appears well structured. Coronal sections of maxillofacial specimens were subjected to peroxidase-based immunohistochemistry for VEGF, VEGFR-1 and VEGFR-2. Double immunofluorescence for VEGF, VEGFR-1 or VEGFR-2 and the neuronal marker protein gene product 9.5 (PGP 9.5) was also performed. VEGF expression was evident in the entire VNO epithelium, with particularly strong reactivity in the middle layer. Strongly VEGF-immunostained cells with aspect similar to bipolar neurons and/or their presumable precursors were detected in the middle and basal layers. Cells detaching from the basal epithelial layer and detached cell groups in the surrounding lamina propria showed moderate/strong VEGF expression. The strongest VEGFR-1 and VEGFR-2 expression was detected in the apical epithelial layer. Cells with aspect similar to bipolar neurons and/or their presumable precursors located in the middle and basal layers and the detaching/detached cells displayed a VEGFR-1 and VEGFR-2 reactivity similar to that of VEGF. The basal epithelial layer exhibited stronger staining for VEGFRs than for VEGF. Cells with morphology and VEGF/VEGFR expression similar to those of the detaching/detached cells were also detected in the middle and basal VNO epithelial layers. Double immunofluorescence using anti-PGP 9.5 antibodies demonstrated that most of the VEGF/VEGFR-immunoreactive cells were neuronal cells. Collectively, our findings suggest that during early fetal development the VEGF/VEGFR system might be involved in the presumptive VNO chemoreceptor activity and neuronal precursor migration.


Subject(s)
Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vomeronasal Organ , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Vomeronasal Organ/chemistry , Vomeronasal Organ/embryology
15.
Int J Mol Sci ; 19(5)2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29702562

ABSTRACT

Vascular endothelial growth factor receptor (VEGFR)-1 exists in different forms, derived from alternative splicing of the same gene. In addition to the transmembrane form, endothelial cells produce a soluble VEGFR-1 (sVEGFR-1) isoform, whereas non-endothelial cells produce both sVEGFR-1 and a different soluble molecule, known as soluble fms-like tyrosine kinase (sFlt)1-14. By binding members of the vascular endothelial growth factor (VEGF) family, the soluble forms reduce the amounts of VEGFs available for the interaction with their transmembrane receptors, thereby negatively regulating VEGFR-mediated signaling. In agreement with this activity, high levels of circulating sVEGFR-1 or sFlt1-14 are associated with different pathological conditions involving vascular dysfunction. Moreover, sVEGFR-1 and sFlt1-14 have an additional role in angiogenesis: they are deposited in the endothelial cell and pericyte extracellular matrix, and interact with cell membrane components. Interaction of sVEGFR-1 with α5β1 integrin on endothelial cell membranes regulates vessel growth, triggering a dynamic, pro-angiogenic phenotype. Interaction of sVEGFR-1/sFlt1-14 with cell membrane glycosphingolipids in lipid rafts controls kidney cell morphology and glomerular barrier functions. These cell⁻matrix contacts represent attractive novel targets for pharmacological intervention in addition to those addressing interactions between VEGFs and their receptors.


Subject(s)
Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Mice , Models, Animal , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/chemistry
16.
PLoS One ; 13(4): e0187082, 2018.
Article in English | MEDLINE | ID: mdl-29641523

ABSTRACT

Preeclampsia is one of the most serious complications of pregnancy. Currently there are no medical treatments. Given placental oxidative stress may be an early trigger in the pathogenesis of preeclampsia, therapies that enhance antioxidant pathways have been proposed as treatments. Melatonin is a direct free-radical scavenger and indirect antioxidant. We performed in vitro assays to assess whether melatonin 1) enhances the antioxidant response element genes (heme-oxygenase 1, (HO-1), glutamate-cysteine ligase (GCLC), NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1), thioredoxin (TXN)) or 2) alters secretion of the anti-angiogenic factors soluble fms-like tyrosine kinase-1 (sFLT) or soluble endoglin (sENG) from human primary trophoblasts, placental explants and human umbilical vein endothelial cells (HUVECs) and 3) can rescue TNF-α induced endothelial dysfunction. In primary trophoblast melatonin treatment increased expression of the antioxidant enzyme TXN. Expression of TXN, GCLC and NQO1 was upregulated in placental tissue with melatonin treatment. HUVECs treated with melatonin showed an increase in both TXN and GCLC. Melatonin did not increase HO-1 expression in any of the tissues examined. Melatonin reduced sFLT secretion from primary trophoblasts, but had no effect on sFLT or sENG secretion from placental explants or HUVECs. Melatonin did not rescue TNF-α induced VCAM-1 and ET-1 expression in endothelial cells. Our findings suggest that melatonin induces antioxidant pathways in placenta and endothelial cells. Furthermore, it may have effects in reducing sFLT secretion from trophoblast, but does not reduce endothelial dysfunction. Given it is likely to be safe in pregnancy, it may have potential as a therapeutic agent to treat or prevent preeclampsia.


Subject(s)
Antioxidants/metabolism , Melatonin/pharmacology , Placenta/metabolism , Pre-Eclampsia/drug therapy , Trophoblasts/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Melatonin/therapeutic use , Placenta/drug effects , Pre-Eclampsia/pathology , Pregnancy , Solubility , Trophoblasts/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor Receptor-1/chemistry
17.
Oncol Rep ; 39(5): 2385-2392, 2018 May.
Article in English | MEDLINE | ID: mdl-29498403

ABSTRACT

Cancer vaccines have been developed as a new therapeutic approach, however, their clinical benefit remains limited. We previously performed a phase II study for advanced colorectal cancer (CRC) using five human leukocyte antigen (HLA-A*24:02)-restricted peptides derived from kinase of the outer chloroplast membrane 1, translocase of outer mitochondrial membrane 34 (TOMM34), ring finger protein 43 (RNF43), vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2. In the present study the relationship between overall survival (OS) and several biomarkers, including cytotoxic T lymphocyte (CTL) and immunoglobulin G (IgG) responses to these five peptides, was investigated. In 89 advanced CRC patients treated with a combination therapy consisting of these five peptides and oxaliplatin-based chemotherapy, plasma was collected before and after 3 months of vaccine administration. IgGs reactive to each of the five peptides were assessed using the multiplex bead suspension Luminex system. Antigen-specific T-cell responses were estimated by enzyme-linked immunoSpot assay. Plasma levels of TOMM34 IgG (P<0.001), RNF43 IgG (P<0.001) and VEGFR2 IgG (P<0.001) were significantly increased after vaccination and stronger VEGFR2 IgG responses correlated significantly with OS in HLA-matched patients (P=0.034). CTL responses to VEGFR1 and VEGFR2 were also significantly increased in the HLA-matched group (P=0.049 and P<0.001, respectively). However, increased CTL response did not correlate with OS. Multivariate analysis indicated that IgG responses to VEGFR2 were the most significant predictor for OS in the HLA-A*24:02-matched group (P=0.04). Our findings indicated that VEGFR2 IgG responses may be an important immunological biomarker in the early course of treatment for CRC patients treated with therapeutic epitope peptides.


Subject(s)
Cancer Vaccines/administration & dosage , Colorectal Neoplasms/drug therapy , HLA-A24 Antigen/immunology , Immunoglobulin G/metabolism , Aged , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/immunology , Double-Blind Method , Epitopes/immunology , Female , Humans , Male , Middle Aged , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/immunology , Mitochondrial Precursor Protein Import Complex Proteins , Oncogene Proteins/chemistry , Oncogene Proteins/immunology , Survival Analysis , Treatment Outcome , Ubiquitin-Protein Ligases , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/immunology
18.
Integr Biol (Camb) ; 10(1): 18-33, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29327758

ABSTRACT

Inducing therapeutic angiogenesis to effectively form hierarchical, non-leaky networks of perfused vessels in tissue engineering applications and ischemic disease remains an unmet challenge, despite extensive research and multiple clinical trials. Here, we use a previously-developed, multi-scale, computational systems pharmacology model of human peripheral artery disease to screen a diverse array of promising pro-angiogenic strategies, including gene therapy, biomaterials, and antibodies. Our previously-validated model explicitly accounts for VEGF immobilization, Neuropilin-1 binding, and weak activation of VEGF receptor 2 (VEGFR2) by the "VEGFxxxb" isoforms. First, we examine biomaterial-based delivery of VEGF engineered for increased affinity to the extracellular matrix. We show that these constructs maintain VEGF close to physiological levels and extend the duration of VEGFR2 activation. We demonstrate the importance of sub-saturating VEGF dosing to prevent angioma formation. Second, we examine the potential of ligand- or receptor-based gene therapy to normalize VEGF receptor signaling. Third, we explore the potential for antibody-based pro-angiogenic therapy. Our model supports recent observations that improvement in perfusion following treatment with anti-VEGF165b in mice is mediated by VEGF-receptor 1, not VEGFR2. Surprisingly, the model predicts that the approved anti-VEGF cancer drug, bevacizumab, may actually improve signaling of both VEGFR1 and VEGFR2 via a novel 'antibody swapping' effect that we demonstrate here. Altogether, this model provides insight into the mechanisms of action of several classes of pro-angiogenic strategies within the context of the complex molecular and physiological processes occurring in vivo. We identify molecular signaling similarities between promising approaches and key differences between promising and ineffective strategies.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Medical Informatics , Peripheral Arterial Disease/drug therapy , Animals , Bevacizumab/therapeutic use , Biocompatible Materials/therapeutic use , Drug Delivery Systems , Genetic Therapy , Humans , Ischemia/pathology , Mice , Muscle, Skeletal/pathology , Neovascularization, Pathologic/metabolism , Neuropilin-1/chemistry , Protein Binding , Signal Transduction , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry
19.
J Med Chem ; 60(15): 6598-6606, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28686443

ABSTRACT

Cancer angiogenesis is mainly initiated by vascular endothelial growth factors (VEGFs). On the basis of the reported crystal structures of three natural ligands (VEGF-A, -B, and PlGF) with the major receptors VEGFR-1 and VEGFR-2, we scanned receptor-binding epitopes of these ligands by designing linear and cyclic peptides with the aim to disrupt the VEGF-A/VEGFR-1 interaction, which is implicated in cancer development. The ability of peptides to inhibit this interaction was evaluated by an ELISA-based assay. Several peptides, especially those mimicking loop 1 (L1) of these ligands that binds primarily to domain D3 of VEGFRs, have demonstrated higher inhibition for VEGF-A/VEGFR-1 binding. They have also shown inhibitory effects on VEGF-induced tube formation in HUVECs (human umbilical vein endothelial cells). These results validate the domain D3 of VEGFRs as an efficient target for the design of VEGFR antagonists.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Peptide Fragments/pharmacology , Peptides/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Drug Design , Epitopes/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Membrane Proteins/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Protein Domains , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor B/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry
20.
Anal Biochem ; 530: 107-112, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28477965

ABSTRACT

The VEGFR1 has been shown to play a role in the regulation of angiogenesis, and has therefore been associated to several pathologies. In order to extend our toolbox of screening methods for the identification of compounds disrupting the VEGF receptor 1/VEGF interaction, we developed a fast and accurate displacement assay, in which VEGF receptor 1 domain 2 is directly labeled with an enzyme, bypassing the classical streptavidin-biotin interaction system. A description of this straightforward strategy is provided here, including its advantages and disadvantages. Optimization of the reagents preparation, purification and conservation, and displacement assay with known molecular entities are presented.


Subject(s)
Biological Assay/methods , Horseradish Peroxidase/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Binding, Competitive , Biotin/chemistry , Biotin/metabolism , Enzyme-Linked Immunosorbent Assay , Horseradish Peroxidase/chemistry , Humans , Ligands , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Streptavidin/chemistry , Streptavidin/metabolism , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL