Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicon ; 165: 78-82, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31054289

ABSTRACT

The cutaneous wound healing property of a pro-angiogenic venom peptide (RVVAP) in a cream-based formulation was evaluated using the excision wound healing model on Wistar strain rats. The wound healing potency and modest antibacterial activity of RVVAP was enhanced significantly (p < 0.05) when combined with Aloe vera extract. RVVAP was also found to be non-toxic at the tested dose of 1.0 mg/kg. Nevertheless, the release of inflammatory cytokines such as IL-1, IL-6, IL-10, and TNF-α in RVVAP-treated mice was suppressed, compared to the untreated controls. This is the first report assessing the wound healing potential of a low-molecular mass, non-enzymatic, pro-angiogenic peptide purified from snake venom.


Subject(s)
Angiogenic Proteins/therapeutic use , Daboia , Viper Venoms/chemistry , Wound Healing/drug effects , Angiogenic Proteins/isolation & purification , Animals , Rats , Rats, Wistar
2.
Nat Commun ; 10(1): 51, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604775

ABSTRACT

The brain-specific angiogenesis inhibitor (BAI) subfamily of adhesion G protein-coupled receptors (aGPCRs) plays crucial roles in diverse cellular processes including phagocytosis, myoblast fusion, and synaptic development through the ELMO/DOCK/Rac signaling pathway, although the underlying molecular mechanism is not well understood. Here, we demonstrate that an evolutionarily conserved fragment located in the C-terminal cytoplasmic tail of BAI-aGPCRs is specifically recognized by the RBD-ARR-ELMO (RAE) supramodule of the ELMO family scaffolds. The crystal structures of ELMO2-RAE and its complex with BAI1 uncover the molecular basis of BAI/ELMO interactions. Based on the complex structure we identify aGPCR-GPR128 as another upstream receptor for the ELMO family scaffolds, most likely with a recognition mode similar to that of BAI/ELMO interactions. Finally, we map disease-causing mutations of BAI and ELMO and analyze their effects on complex formation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Angiogenic Proteins/genetics , Cytoskeletal Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Receptors, G-Protein-Coupled/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/isolation & purification , Adaptor Proteins, Signal Transducing/metabolism , Angiogenic Proteins/chemistry , Angiogenic Proteins/isolation & purification , Angiogenic Proteins/metabolism , Animals , Cell Line , Crystallography, X-Ray , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/isolation & purification , Cytoskeletal Proteins/metabolism , HEK293 Cells , Humans , Mice , Mutagenesis , Mutation , Neoplasms/genetics , Receptors, G-Protein-Coupled/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment
3.
Stem Cell Res Ther ; 8(1): 219, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28974256

ABSTRACT

BACKGROUND: The therapeutic potential of mesenchymal stem cells (MSCs) may be attributed partly to humoral factors such as growth factors, cytokines, and chemokines. Human term placental tissue-derived MSCs (PlaMSCs), or conditioned medium left over from cultures of these cells, have been reported to enhance angiogenesis. Recently, the exosome, which can transport a diverse suite of macromolecules, has gained attention as a novel intercellular communication tool. However, the potential role of the exosome in PlaMSC therapeutic action is not well understood. The purpose of this study was to evaluate PlaMSC-derived exosome angiogenesis promotion in vitro and in vivo. METHODS: MSCs were isolated from human term placental tissue by enzymatic digestion. Conditioned medium was collected after 48-h incubation in serum-free medium (PlaMSC-CM). Angiogenic factors present in PlaMSC-CM were screened by a growth factor array. Exosomes were prepared by ultracentrifugation of PlaMSC-CM, and confirmed by transmission electron microscopy, dynamic light scattering, and western blot analyses. The proangiogenic activity of PlaMSC-derived exosomes (PlaMSC-exo) was assessed using an endothelial tube formation assay, a cell migration assay, and reverse transcription-PCR analysis. The in-vivo angiogenic activity of PlaMSC-exo was evaluated using a murine auricle ischemic injury model. RESULTS: PlaMSC-CM contained both angiogenic and angiostatic factors, which enhanced endothelial tube formation. PlaMSC-exo were incorporated into endothelial cells; these exosomes stimulated both endothelial tube formation and migration, and enhanced angiogenesis-related gene expression. Laser Doppler blood flow analysis showed that PlaMSC-exo infusion also enhanced angiogenesis in an in-vivo murine auricle ischemic injury model. CONCLUSIONS: PlaMSC-exo enhanced angiogenesis in vitro and in vivo, suggesting that exosomes play a role in the proangiogenic activity of PlaMSCs. PlaMSC-exo may be a novel therapeutic approach for treating ischemic diseases.


Subject(s)
Angiogenic Proteins/pharmacology , Ear Auricle/drug effects , Exosomes/transplantation , Neovascularization, Physiologic/drug effects , Placenta/cytology , Reperfusion Injury/therapy , Angiogenic Proteins/isolation & purification , Animals , Biological Assay , Cell Movement , Culture Media, Conditioned/chemistry , Culture Media, Serum-Free , Ear Auricle/blood supply , Ear Auricle/injuries , Ear Auricle/pathology , Exosomes/chemistry , Female , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Placenta/metabolism , Pregnancy , Primary Cell Culture , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
4.
Stem Cell Res Ther ; 8(1): 212, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28969687

ABSTRACT

BACKGROUND: The main goal of bone tissue engineering has been the generation of healthy bone in order to replace affected tissue. Therefore, optimized biomaterials are needed which allow the survival and growth of mesenchymal stem cells. Until now the key challenge in the clinical application of cell-based tissue engineering bone implants was poor diffusion of oxygen into the tissue, making functional blood vessel networks a necessity. With their ability to evolve into different cell types, to expand extensively in vitro, and to release paracrine soluble factors, bone marrow stromal cells (BMSC) are highly attractive for tissue engineering. During the last years hypoxia became a proven method to control proliferation, differentiation, and pluripotency of BMSC. Here we applied different methods to characterize metabolically conditioned media (MCM) in comparison to hypoxia conditioned media (HCM) and evaluated their ability to attract BMSC in 2-D migration assays. METHODS: BMSC and fibroblasts of human origin were isolated and cultivated to obtain HCM and MCM. Both media were characterized by angiogenesis arrays, cytokine arrays, and ELISA for selected factors. 2-D migration tests were performed with Corning Transwell®-96 permeable support chambers with porous polyester membranes with a pore size of 8.0 µm. RESULTS: Characterization of HCM and MCM revealed that the concentration of angiogenic factors was higher in MCM than in HCM. However, the chemoattractive capacity of MCM for BMSC was equivalent to that of HCM. HCM and MCM produced by human skin fibroblasts attracted human BMSC as efficiently as HCM and MCM produced by human BMSC. CONCLUSIONS: HCM and MCM have a high chemoattractive capacity for BMSC. Both conditioned media harbor high concentrations of angiogenic factors which are important for angiogenesis and cell migration. Both chemoattracting conditioned media can also be derived from skin fibroblasts which can easily be obtained from patients in individualized therapy approaches.


Subject(s)
Angiogenic Proteins/pharmacology , Bone Marrow Cells/metabolism , Chemotactic Factors/pharmacology , Culture Media, Conditioned/chemistry , Fibroblasts/metabolism , Mesenchymal Stem Cells/drug effects , Angiogenic Proteins/biosynthesis , Angiogenic Proteins/isolation & purification , Angiogenic Proteins/metabolism , Biological Assay , Bone Marrow Cells/cytology , Cell Hypoxia , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemotactic Factors/biosynthesis , Chemotactic Factors/isolation & purification , Chemotactic Factors/metabolism , Chemotaxis/drug effects , Chemotaxis/physiology , Culture Media, Conditioned/pharmacology , Diffusion Chambers, Culture , Fibroblasts/cytology , Foreskin/cytology , Foreskin/metabolism , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic/drug effects , Primary Cell Culture
5.
J Infect ; 74(1): 10-21, 2017 01.
Article in English | MEDLINE | ID: mdl-27717783

ABSTRACT

BACKGROUND: Tuberculosis-diabetes co-morbidity (TB-DM) is characterized by increased inflammation with elevated circulating levels of inflammatory cytokines and other factors. Circulating angiogenic factors are intricately involved in the angiogenesis-inflammation nexus. METHODS: To study the association of angiogenic factors with TB-DM, we examined the systemic levels of VEGF-A, VEGF-C, VEGF-D, VEGF-R1, VEGF-R2, VEGF-R3 in individuals with either TB-DM (n = 44) or TB alone (n = 44). RESULTS: Circulating levels of VEGF-A, C, D, R1, R2 and R3 were significantly higher in TB-DM compared to TB individuals. Moreover, the levels of VEGF-A, C, R2 and/or R3 were significantly higher in TB-DM with bilateral or cavitary disease or with hemoptysis, suggesting an association with both disease severity and adverse clinical presentation. The levels of these factors also exhibited a significant positive relationship with bacterial burdens and HbA1c levels. In addition, VEGF-A, C and R2 levels were significantly higher (at 2 months of treatment) in culture positive compared to culture negative TB-DM individuals. Finally, the circulating levels of VEGF-A, C, D, R1, R2 and R3 were significantly reduced following successful chemotherapy at 6 months. CONCLUSION: Our data demonstrate that TB-DM is associated with heightened levels of circulating angiogenic factors, possibly reflecting both dysregulated angiogenesis and exaggerated inflammation.


Subject(s)
Angiogenic Proteins/blood , Diabetes Complications/blood , Diabetes Mellitus/blood , Tuberculosis/blood , Tuberculosis/complications , Adult , Aged , Angiogenic Proteins/isolation & purification , Bacterial Load , Biomarkers/blood , Comorbidity , Cytokines/blood , Diabetes Mellitus/microbiology , Female , Humans , Male , Middle Aged , Tuberculosis/microbiology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/isolation & purification , Vascular Endothelial Growth Factor C/blood , Vascular Endothelial Growth Factor C/isolation & purification , Vascular Endothelial Growth Factor D/blood , Vascular Endothelial Growth Factor D/isolation & purification
6.
J Immunol ; 196(12): 5089-100, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27194784

ABSTRACT

Milk fat globule epidermal growth factor-factor 8 (MFG-E8) is a peripheral glycoprotein that acts as a bridging molecule between the macrophage and apoptotic cells, thus executing a pivotal role in the scavenging of apoptotic cells from affected tissue. We have previously reported that apoptotic cell clearance activity or efferocytosis is compromised in diabetic wound macrophages. In this work, we test the hypothesis that MFG-E8 helps resolve inflammation, supports angiogenesis, and accelerates wound closure. MFG-E8(-/-) mice displayed impaired efferocytosis associated with exaggerated inflammatory response, poor angiogenesis, and wound closure. Wound macrophage-derived MFG-E8 was recognized as a critical driver of wound angiogenesis. Transplantation of MFG-E8(-/-) bone marrow to MFG-E8(+/+) mice resulted in impaired wound closure and compromised wound vascularization. In contrast, MFG-E8(-/-) mice that received wild-type bone marrow showed improved wound closure and improved wound vascularization. Hyperglycemia and exposure to advanced glycated end products inactivated MFG-E8, recognizing a key mechanism that complicates diabetic wound healing. Diabetic db/db mice suffered from impaired efferocytosis accompanied with persistent inflammation and slow wound closure. Topical recombinant MFG-E8 induced resolution of wound inflammation, improvements in angiogenesis, and acceleration of closure, upholding the potential of MFG-E8-directed therapeutics in diabetic wound care.


Subject(s)
Antigens, Surface/immunology , Antigens, Surface/metabolism , Diabetes Mellitus/physiopathology , Inflammation/drug therapy , Milk Proteins/immunology , Milk Proteins/metabolism , Wound Healing , Angiogenic Proteins/immunology , Angiogenic Proteins/isolation & purification , Angiogenic Proteins/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/pharmacology , Apoptosis , Diabetes Mellitus/immunology , Humans , Macrophages/immunology , Mice , Mice, Inbred C57BL , Milk Proteins/genetics , Milk Proteins/pharmacology , Phagocytosis
7.
J Biochem ; 153(1): 5-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23100270

ABSTRACT

Angiogenesis, a formation of neovessels, is regulated by the local balance between angiogenesis stimulators and inhibitors. A number of such endogenous regulators of angiogenesis have been found in the body. Recently, vasohibin-1 (VASH1) was isolated as a negative feedback regulator of angiogenesis produced by endothelial cells (ECs) and subsequently vasohibin-2 (VASH2) as a homologue of VASH1. It was then explored that VASH1 is expressed in ECs to terminate angiogenesis, whereas VASH2 is expressed in cells other than ECs to promote angiogenesis in the mouse model of angiogenesis. This review will focus on the vasohibin family members, which are novel regulators of angiogenesis.


Subject(s)
Angiogenic Proteins/metabolism , Cell Cycle Proteins/metabolism , Neovascularization, Physiologic , Angiogenic Proteins/chemistry , Angiogenic Proteins/genetics , Angiogenic Proteins/isolation & purification , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/isolation & purification , Endothelium, Vascular/metabolism , Feedback, Physiological , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism
8.
Thromb Haemost ; 99(3): 576-85, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18327407

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a plasma Kunitz-type serine protease inhibitor that is mainly known for its inhibition of tissue factor-mediated coagulation. In addition to its anticoagulant properties, emerging data show that TFPI may also regulate endothelial cell functions via a non-haemostatic pathway. In this work we demonstrate that at concentrations within the physiological range, TFPI inhibits both endothelial cell migration and their differentiation into capillary-like structures in vitro. These effects were specific to endothelial cells since no inhibitory effect was observed on the migration of tumor (glioblastoma) cells. Inhibition of endothelial cell migration was correlated with a concomitant loss in cell adhesion, suggesting an alteration of focal adhesion complex integrity. Accordingly, we observed that TFPI inhibited the phosphorylation of focal adhesion kinase and paxillin, two key proteins involved in the scaffolding of these complexes, and that this effect was specific to endothelial cells. These results suggest that TFPI influences the angiogenic process via a non-haemostatic pathway, by downregulating the migratory mechanisms of endothelial cells.


Subject(s)
Angiogenic Proteins/metabolism , Cell Movement , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Lipoproteins/metabolism , Neovascularization, Physiologic , Paxillin/metabolism , Angiogenic Proteins/isolation & purification , Angiogenic Proteins/pharmacology , Cell Adhesion , Cell Differentiation , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape , Cells, Cultured , Cloning, Molecular , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Humans , Lipoproteins/isolation & purification , Lipoproteins/pharmacology , Lysophospholipids/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic/drug effects , Phosphorylation , Recombinant Proteins/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...