Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 430
Filter
1.
Bioorg Chem ; 136: 106529, 2023 07.
Article in English | MEDLINE | ID: mdl-37084585

ABSTRACT

The aberrant activation of the fibroblast growth factor 2 (FGF2)/fibroblast growth factor receptor (FGFR) signalling pathway drives severe pathologies, including cancer development and angiogenesis-driven pathologies. The perturbation of the FGF2/FGFR axis via extracellular allosteric small inhibitors is a promising strategy for developing FGFR inhibitors with improved safety and efficacy for cancer treatment. We have previously investigated the role of new extracellular inhibitors, such as rosmarinic acid (RA), which bind the FGFR-D2 domain and directly compete with FGF2 for the same binding site, enabling the disruption of the functional FGF2/FGFR interaction. To select ligands for the previously identified FGF2/FGFR RA binding site, NMR data-driven virtual screening has been performed on an in-house library of non-commercial small molecules and metabolites. A novel drug-like compound, a resorcinol derivative named RBA4 has been identified. NMR interaction studies demonstrate that RBA4 binds the FGF2/FGFR complex, in agreement with docking prediction. Residue-level NMR perturbations analysis highlights that the mode of action of RBA4 is similar to RA in terms of its ability to target the FGF2/FGFR-D2 complex, inducing perturbations on both proteins and triggering complex dissociation. Biological assays proved that RBA4 inhibited FGF2 proliferative activity at a level comparable to the previously reported natural product, RA. Identification of RBA4 chemical groups involved in direct interactions represents a starting point for further optimization of drug-like extracellular inhibitors with improved activity.


Subject(s)
Fibroblast Growth Factor 2 , Neoplasms , Humans , Fibroblast Growth Factor 2/antagonists & inhibitors , Magnetic Resonance Spectroscopy , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Resorcinols/chemistry , Resorcinols/pharmacology
2.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836603

ABSTRACT

Retinal neovascularization is a leading cause of severe visual loss in humans, and molecular mechanisms of microglial activation-driven angiogenesis remain unknown. Using single-cell RNA sequencing, we identified a subpopulation of microglia named sMG2, which highly expressed necroptosis-related genes Rip3 and Mlkl. Genetic and pharmacological loss of function demonstrated that hypoxia-induced microglial activation committed to necroptosis through the RIP1/RIP3-mediated pathway. Specific deletion of Rip3 gene in microglia markedly decreased retinal neovascularization. Furthermore, hypoxia induced explosive release of abundant FGF2 in microglia through RIP3-mediated necroptosis. Importantly, blocking signaling components of the microglia necropotosis-FGF2 axis largely ablated retinal angiogenesis and combination therapy with simultaneously blocking VEGF produced synergistic antiangiogenic effects. Together, our data demonstrate that targeting the microglia necroptosis axis is an antiangiogenesis therapy for retinal neovascular diseases.


Subject(s)
Microglia/pathology , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Retinal Diseases/pathology , Animals , Drug Therapy, Combination , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/metabolism , Hypoxia/pathology , Mice , Microglia/metabolism , Necroptosis/drug effects , Neovascularization, Pathologic , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
3.
Chembiochem ; 22(1): 160-169, 2021 01 05.
Article in English | MEDLINE | ID: mdl-32975328

ABSTRACT

Fibroblast growth factor (FGF2)/fibroblast growth factor receptor (FGFR) signalling plays a major role both in physiology and in several pathologies, including cancer development, metastasis formation and resistance to therapy. The development of small molecules, acting extracellularly to target FGF2/FGFR interactions, has the advantage of limiting the adverse effects associated with current intracellular FGFR inhibitors. Herein, we discuss the ability of the natural compound rosmarinic acid (RA) to induce FGF2/FGFR complex dissociation. The molecular-level description of the FGF2/FGFR/RA system, by NMR spectroscopy and docking, clearly demonstrates that RA binds to the FGFR-D2 domain and directly competes with FGF2 for the same binding site. Direct and allosteric perturbations combine to destabilise the complex. The proposed molecular mechanism is validated by cellular studies showing that RA inhibits FGF2-induced endothelial cell proliferation and FGFR activation. Our results can serve as the basis for the development of new extracellular inhibitors of the FGF/FGFR pathways.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Biological Products/pharmacology , Cinnamates/pharmacology , Depsides/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Angiogenesis Inhibitors/chemistry , Animals , Biological Products/chemistry , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Cinnamates/chemistry , Depsides/chemistry , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/metabolism , Molecular Docking Simulation , Phosphorylation/drug effects , Receptors, Fibroblast Growth Factor/chemistry , Rosmarinic Acid
4.
Sci Rep ; 10(1): 15968, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994514

ABSTRACT

Intraplaque angiogenesis increases the chance of unstable atherosclerotic plaque rupture and thrombus formation leading to myocardial infarction. Basic Fibroblast Growth Factor (bFGF) plays a key role in angiogenesis and inflammation and is involved in the pathogenesis of atherosclerosis. Therefore, we aim to test K5, a small molecule bFGF-inhibitor, on remodelling of accelerated atherosclerotic vein grafts lesions in ApoE3*Leiden mice. K5-mediated bFGF-signalling blockade strongly decreased intraplaque angiogenesis and intraplaque hemorrhage. Moreover, it reduced macrophage infiltration in the lesions by modulating CCL2 and VCAM1 expression. Therefore, K5 increases plaque stability. To study the isolated effect of K5 on angiogenesis and SMCs-mediated intimal hyperplasia formation, we used an in vivo Matrigel-plug mouse model that reveals the effects on in vivo angiogenesis and femoral artery cuff model to exclusively looks at SMCs. K5 drastically reduced in vivo angiogenesis in the matrigel plug model while no effect on SMCs migration nor proliferation could be seen in the femoral artery cuff model. Moreover, in vitro K5 impaired endothelial cells functions, decreasing migration, proliferation and tube formation. Our data show that K5-mediated bFGF signalling blockade in hypercholesterolemic ApoE3*Leiden mice reduces intraplaque angiogenesis, haemorrhage and inflammation. Therefore, K5 is a promising candidate to stabilize advanced atherosclerotic plaques.


Subject(s)
Apolipoproteins E/genetics , Fibroblast Growth Factor 2/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Plaque, Atherosclerotic/drug therapy , Small Molecule Libraries/administration & dosage , Animals , Cell Line , Cell Proliferation/drug effects , Chemokine CCL2/metabolism , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Molecular Structure , Neovascularization, Pathologic/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Small Molecule Libraries/pharmacology , Versicans/metabolism
5.
Sci Rep ; 10(1): 14756, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32901093

ABSTRACT

Liver fibrosis, a common outcome of chronic liver disease characterized by excessive accumulation of extracellular matrix (ECM), is a leading cause of mortality worldwide. The tyrosine kinase inhibitor neratinib is a human epidermal growth factor receptor 2 (HER2) inhibitor approved by the FDA for HER2-positive breast cancer treatment; however, it has not yet been evaluated for liver fibrosis treatment. We elucidated the anti-fibrotic effects of neratinib in hepatic stellate cells (HSCs) and in vivo models of CCl4-induced liver fibrosis. HSC activation is a key step in liver fibrogenesis and has a crucial role in collagen deposition, as it is primarily responsible for excessive ECM production. The effect of neratinib on HSC was evaluated in transforming growth factor (TGF-ß)-incubated LX-2 cells and culture-activated primary human HSCs. In vivo study results indicated that neratinib inhibited the inflammatory response, HSC differentiation, and collagen accumulation induced by CCl4. Moreover, the anti-fibrotic effects of neratinib were not associated with the HER2 signaling pathways. Neratinib inhibited FGF2 expression in activated HSCs and serum FGF2 level in the model, suggesting that neratinib possessed therapeutic potency against liver fibrosis and the potential for application against other fibrotic diseases.


Subject(s)
Fibroblast Growth Factor 2/antagonists & inhibitors , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/prevention & control , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Animals , Carbon Tetrachloride/toxicity , Cell Proliferation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
6.
Biomed Pharmacother ; 131: 110628, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32890968

ABSTRACT

BACKGROUND: Synovial fibroblasts (SFs) act as key effector cells mediating synovial inflammation and joint destruction in rheumatoid arthritis (RA). Fibroblast growth factor 2 (FGF2) and its receptors (FGFRs) play important roles in RASF-mediated osteoclastogenesis. Pentraxin 3 (PTX3) is a soluble pattern recognition receptor with nonredundant roles in inflammation and innate immunity. PTX3 is produced by various cell types, including SFs and is highly expressed in RA. However, the role of PTX3 in FGF2-induced osteoclastogenesis in RA and the underlying mechanism have been poorly elucidated. METHODS: We first determined the expression of FGF2 and RANKL in synovial tissue and synovial fluid of RA patients. We then examined the effect of PTX3 on RASF osteoclastogenesis induced by endogenous and exogenous FGF2 in isolated RASF cells treated with FGF2 and/or recombinant PTX3 (rPTX3). Thirdly, we analyzed the effect of PTX3 on FGF2 binding to FGFR-1 and HSPG receptors on RASFs. Lastly, we evaluated joint morphology after injection of rPTX3 into collagen-induced arthritis (CIA) mice. RESULTS: FGF2 was confirmed to be highly expressed in both synovial tissue and synovial fluid of RA patients. FGF2 promoted cell proliferation and increased the expressions of RANKL and ICAM-1 and RANKL/OPG to induce osteoclastogenesis in RASF, while anti-FGF2 neutralized this effect. PTX3 significantly inhibited FGF2-induced RASF cell growth and osteoclastogenesis by preventing the interaction of 125I-FGF2 and FGFRs on the same cells. In addition, administration of rPTX3 significantly ameliorated cartilage and bone destruction in mice with CIA. CONCLUSIONS: PTX3 exhibited an inhibitory effect on the autocrine and paracrine stimulation of FGF2 on SFs, and ameliorated bone destruction in CIA mice. PTX3 may be implicated in bone destruction in RA, which may provide theoretical evidence and potential therapeutic targets for RA treatment.


Subject(s)
Arthritis, Rheumatoid/metabolism , C-Reactive Protein/administration & dosage , Fibroblast Growth Factor 2/biosynthesis , Osteoclasts/metabolism , Serum Amyloid P-Component/administration & dosage , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/pathology , Cells, Cultured , Collagen/toxicity , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , Mice , Osteoclasts/drug effects , Osteoclasts/pathology , Random Allocation , Synovial Fluid/cytology , Synovial Fluid/drug effects , Synovial Fluid/metabolism
7.
Mol Biol Rep ; 47(10): 8301-8304, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32920756

ABSTRACT

The interest on the role of angiogenesis in the pathogenesis and progression of human interstitial lung diseases is growing, with conventional sprouting (SA) and non-sprouting intussusceptive angiogenesis (IA) being differently represented in specific pulmonary injury patterns. The role of viruses as key regulators of angiogenesis is known for several years. A significantly enhanced amount of new vessel growth, through a mechanism of IA, has been reported in lungs of patients who died from Covid-19; among the angiogenesis-related genes, fibroblast growth factor 2 (FGF2) was found to be upregulated. These findings are intriguing. FGF2 plays a role in some viral infections: the upregulation is involved in the MERS-CoV-induced strong apoptotic response crucial for its highly lytic replication cycle in lung cells, whereas FGF2 is protective against the acute lung injury induced by H1N1 influenza virus, improving the lung wet-to-dry weight ratio. FGF2 plays a role also in regulating IA, acting on pericytes (crucial for the formation of intraluminal pillars), and endothelium, and FGF2-induced angiogenesis may be promoted by inflammation and hypoxia. IA is a faster and probably more efficient process than SA, able to modulate vascular remodeling through pruning of redundant or inefficient blood vessels. We can speculate that IA might have the function of restoring a functional vascular plexus consequently to extensive endothelialitis and alveolar capillary micro-thrombosis observed in Covid-19. Anti-Vascular endothelial growth factor (anti-VEGF) strategies are currently investigated for treatment of severe and critically ill Covid-19 patients, but also FGF2, and its expression and/or signaling, might represent a promising target.


Subject(s)
Coronavirus Infections/pathology , Fibroblast Growth Factor 2/metabolism , Neovascularization, Pathologic/virology , Pneumonia, Viral/pathology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/etiology , Drug Delivery Systems , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , Intussusception/virology , Neovascularization, Pathologic/genetics , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/etiology
8.
Eur Rev Med Pharmacol Sci ; 24(12): 6707-6715, 2020 06.
Article in English | MEDLINE | ID: mdl-32633361

ABSTRACT

OBJECTIVE: This study was designed to investigate whether microRNA-936 can be involved in the development of gastric cancer (GCa) by down-regulating FGF2 expression and activating the phosphatidylinositol 3-kinase/protein kinase B (P13K/Akt) signaling pathway. PATIENTS AND METHODS: Quantitative polymerase chain reaction (qPCR) was carried out to examine microRNA-936 and FGF2 levels in GCa tissue samples and adjacent normal ones, and further in GCa cell lines. After transfection of microRNA-936 inhibitor in GCa cell lines BGC and SGC, cell invasion, and proliferation abilities were evaluated by transwell and cell counting kit-8 (CCK-8) assays, respectively. In addition, the Dual-Luciferase reporting assay was conducted to verify the binding relationship between microRNA-936 and FGF2. After simultaneous transfection of microRNA-936 inhibitor and si-FGF2 in GCa cells, we detected the expression of FGF2/P13K/Akt by performing qPCR and Western blot experiments to further verify the regulation of microRNA-936 on FGF2 and PI3K/AKT pathway. RESULTS: QPCR detection revealed that microRNA-936 was remarkably up-regulated while FGF2 was conversely down-regulated in GCa tissue samples, indicating a negative correlation between the two. In addition, compared with normal gastric mucosal cells GES, microRNA-936 showed a significant increased expression in GCa cell lines. Meanwhile, down-regulation of microRNA-936 caused a marked reduction in invasive and proliferation ability of GCa cells. Dual-Luciferase reporting assay demonstrated a direct binding of microRNA-936 to FGF2. QPCR and Western blot showed that microRNA-936 can inhibit FGF2 expression and activate the PI3K/AKT pathway at the same time. Further studies found that silencing FGF2 induced an enhancement in cell proliferation and invasiveness, which could be reversed by simultaneous downregulation of microRNA-936. The above observations suggested that microRNA-936 may accelerate the progression of GCa by inhibiting FGF2 expression and activating the PI3K/AKT pathway. CONCLUSIONS: Overexpression of microRNA-936 can be conducive to the development of GCa, mainly through the down-regulation of FGF2 and activation of the P13K/Akt signaling pathway.


Subject(s)
Cell Proliferation/physiology , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/metabolism , Cell Line, Tumor , Down-Regulation/physiology , Enzyme Activation/physiology , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Humans , MicroRNAs/biosynthesis , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
9.
Nanomedicine ; 29: 102254, 2020 10.
Article in English | MEDLINE | ID: mdl-32615335

ABSTRACT

FGF-2 accumulates in many tumor tissues and is closely related to the development of tumor angiogenesis and the immunosuppressive microenvironment. This study aimed to investigate whether active immunization against FGF-2 could modify antitumor immunity and enhance the efficacy of an HPV16 E7-specific therapeutic vaccine. Combined immunization targeting both FGF-2 and E7 significantly suppressed tumor growth, which was accompanied by significantly increased levels of IFN-γ-expressing splenocytes and effector CD8 T cells and decreased levels of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells(MDSCs) in both the spleen and tumor; in addition, the levels of FGF-2 and neovascularization in tumors were decreased in the mice receiving the combined immunization, and tumor cell apoptosis was promoted. The combination of an HPV16 E7-specific vaccine and active immunization against FGF-2 significantly enhances antitumor immune responses in mice with TC-1 tumors, indicating a promising strategy for tumor immunotherapy.


Subject(s)
Cancer Vaccines/pharmacology , Fibroblast Growth Factor 2/immunology , Neovascularization, Pathologic/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Humans , Immunotherapy , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Neovascularization, Pathologic/virology , Papillomavirus E7 Proteins/antagonists & inhibitors , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vaccination
10.
Future Oncol ; 16(22): 1619-1627, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32479124

ABSTRACT

Aim: We investigated the direct effects of sevoflurane on angiogenesis and a variety of tumor cells. Materials & methods: The antiangiogenic activity of sevoflurane was determined using angiogenesis and biochemical assays. Results: Sevoflurane at low doses inhibits capillary network formation. Sevoflurane inhibited VEGF- and bFGF-stimulated migration, adhesion and growth in endothelial cells and induced apoptosis. Sevoflurane only at high doses inhibited growth and migration of tumor cells, suggesting differential effects of sevoflurane between endothelial and tumor cells. Mechanistically, sevoflurane decreased growth factors-induced Ras and Rac1 activation, and suppressed Ras and Rac1 signaling. Conclusion: We demonstrate the antiangiogenic effects of sevoflurane and provide preclinical evidence into the potential mechanisms by which sevoflurane may negatively affect cancer growth and metastasis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Sevoflurane/pharmacology , Cell Line, Tumor , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Humans , Paxillin/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , ras Proteins/antagonists & inhibitors
11.
Cancer Med ; 9(12): 4360-4370, 2020 06.
Article in English | MEDLINE | ID: mdl-32347652

ABSTRACT

MicroRNA-646 (miR-646) has been implicated in several other cancers; however, its functional mechanism in non-small cell lung cancer (NSCLC) remains unclear. In this study, we observed the downregulation of miR-646 expression in NSCLC tissues and cell lines. Low-level expression of miR-646 was associated with metastasis and stage of NSCLCs. Functional assays showed that overexpression of miR-646 could suppress NSCLC cell proliferation, clonogenicity, invasion, and inhibit epithelial-mesenchymal transition (EMT), whereas decreased miR-646 expression showed the opposite effects. Importantly, miR-646 overexpression attenuated in vivo tumor growth and metastasis in nude mice models. Mechanically, miR-646 directly targeted and suppressed fibroblast growth factor 2 (FGF2) and cyclin D2 (CCND2) expression. Reintroduction of FGF2 and CCND2 attenuated miR-646-mediated suppression of proliferation and invasion in NSCLC. Collectively, these results demonstrate that miR-646 acts as a tumor suppressor in NSCLC by targeting FGF2 and CCND2, and may serve as a therapeutic target for patients with NSCLC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/secondary , Cyclin D2/antagonists & inhibitors , Fibroblast Growth Factor 2/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , MicroRNAs/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Cell Proliferation , Cyclin D2/genetics , Cyclin D2/metabolism , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
IUBMB Life ; 72(5): 825-841, 2020 05.
Article in English | MEDLINE | ID: mdl-32026601

ABSTRACT

Vasculogenic mimicry (VM) is regarded as a process where very aggressive cancer cells generate vascular-like patterns without the presence of endothelial cells. It is considered as the main mark of malignant cancer and has pivotal role in cancer metastasis and progression in various types of cancers. On the other hand, resistance to the antiangiogenesis therapies leads to the cancer recurrence. Therefore, development of novel chemotherapies and their combinations is urgently needed for abolition of VM structures and also for better tumor therapy. Hence, identifying compounds that target VM structures might be superior therapeutic factors for cancers treatment and controlling the recurrence and metastasis. In recent times, naturally occurring compounds, especially phytochemicals have obtained great attention due to their safe properties. Phytochemicals are also capable of targeting VM structure and also their main signaling pathways. Consequently, in this review article, we illustrated key signaling pathways in VM, and the phytochemicals that affect these structures including curcumin, genistein, lycorine, luteolin, columbamine, triptolide, Paris polyphylla, dehydroeffusol, jatrorrhizine hydrochloride, grape seed proanthocyanidins, resveratrol, isoxanthohumol, dehydrocurvularine, galiellalactone, oxacyclododecindione, brucine, honokiol, ginsenoside Rg3, and norcantharidin. The recognition of these phytochemicals and their safety profile may lead to new therapeutic agents' development for VM elimination in different types of tumors.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Phytochemicals/therapeutic use , Biological Mimicry/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Flavonoids/therapeutic use , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasm Metastasis , Neoplasm Recurrence, Local/blood supply , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Polyphenols/therapeutic use , Proanthocyanidins/therapeutic use , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Biomolecules ; 9(11)2019 10 24.
Article in English | MEDLINE | ID: mdl-31652997

ABSTRACT

BACKGROUND: Disease-related injury in any organ triggers a complex cascade of cellular and molecular responses that culminate in tissue fibrosis, inflammation, and angiogenesis simultaneously. Multiple cell angiogenesis is an essential part of the tissue damage response, which is involved in fibrosis development. RC28-E is a novel recombinant dual decoy receptor lgG1 Fc-fusion protein that can block vascular endothelial growth factor (VEGFA), platelet-derived growth factor (PDGF), and fibroblast growth factor-2 (FGF-2) simultaneously. This protein has stepped into clinical trials (NCT03777254) for the treatment of pathological neovascularization-related diseases. Here, we report on the role of RC28-E during anti-fibrosis and its potential multitarget function in regulating fibrosis. METHODS: A bleomycin-induced pulmonary fibrosis C57BL/6 mouse model was established. Hematoxylin and eosin staining (HE) and Masson staining (Masson's) were performed to evaluate the pulmonary fibrosis based on the scoring from, Ashcroft score. Fibrosis related factors and inflammatory cytokines including HYP, α-SMA, procollagen, ICAM, IL-6, IL-1, and TNF-α were also determined at the protein and mRNA levels to characterize the fibrosis. Both mRNA and protein levels of VEGF, FGF, and transforming growth factor (TGF)-ß were detected by quantitative real-time PCR (qRT-PCR) and immunohistochemical (IHC) analysis, respectively. Pulmonary fibrosis and related cytokines were re-evaluated in vivo after 3 doses of RC28-E (5 mg/kg, 15 mg/kg, and 50 mg/kg, ip. Tiw × 9) in comparison with a mono-target antagonist treatment (VEGF or FGF blocking). RC28-E attenuated the activation of TGF-ß induced fibroblasts in vitro. Expression levels of α-SMA and collagen I, as well as proliferation and migration, were determined with the human skin fibroblast cell line Detroit 551 and primary murine pulmonary fibroblast cells. The mechanism of RC28-E via the TGF-ß/Smad pathway was also investigated. RESULTS: RC28-E exhibits significant anti-fibrosis effects on Idiopathic pulmonary fibrosis (IPF) in vivo. Moreover, TGF-ß induced fibroblast activation in vitro via the inhibition of the TGF-ß downstream Smad pathway, thus providing potential therapeutics for clinical disease-related fibrosis-like IPF as well as chemotherapy-induced fibrosis in cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Lung Injury/drug therapy , Pulmonary Fibrosis/drug therapy , Recombinant Fusion Proteins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Bleomycin , Cell Line , Cell Survival/drug effects , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblasts/drug effects , Humans , Lung/drug effects , Lung/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Male , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
14.
Respir Med ; 156: 33-46, 2019 09.
Article in English | MEDLINE | ID: mdl-31421589

ABSTRACT

The endothelial cells play a crucial role in the progression of angiogenesis, which causes cell re-modulation, proliferation, adhesion, migration, invasion and survival. Angiogenic factors like cytokines, cell adhesion molecules, growth factors, vasoactive peptides, proteolytic enzymes (metalloproteinases) and plasminogen activators bind to their receptors on endothelial cells and activate the signal transduction pathways like epidermal growth factor receptor (EGFR phosphatidylinositol 3-kinase and (PI3K)/AKT/mammalian target of rapamycin (mTOR) which initiate the process of angiogenesis. Cytokines that stimulate angiogenesis include direct and indirect proangiogenic markers. The direct proangiogenic group of markers consists of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF-2) and hepatocyte growth factor (HGF) whereas the indirect proangiogenic markers include transforming growth factor-beta (TGF-ß), interleukin 6 (IL-6), interleukin 8 (IL-8) and platelet-derived growth factor (PDGF). VEGF and FGF-2 are the strongest activators of angiogenesis which stimulate migration and proliferation of endothelial cells in existing vessels to generate and stabilize new blood vessels. VEGF is released in hypoxic conditions as an effect of the hypoxia-inducible factor (HIF-1α) and causes re-modulation and inflammation of bronchi cell. Cell re-modulation and inflammation leads to the development of various lung disorders like pulmonary hypertension, chronic obstructive pulmonary disease, asthma, fibrosis and lung cancer. This indicates that there is a firm link between overexpression of VEGF and FGF-2 with lung disorders. Various natural and synthetic drugs are available for reducing the overexpression of VEGF and FGF-2 which can be helpful in treating lung disorders. Researchers are still searching for new angiogenic inhibitors which can be helpful in the treatment of lung disorders. The present review emphasizes on molecular mechanisms and new drug discovery focused on VEGF and FGF-2 inhibitors and their role as anti-angiogenetic agents in lung disorders.


Subject(s)
Fibroblast Growth Factor 2 , Lung Diseases/etiology , Vascular Endothelial Growth Factor A , Cytokines , Drug Discovery , Endothelial Cells , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , Lung Diseases/drug therapy , Molecular Targeted Therapy , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/antagonists & inhibitors
15.
J Pharm Pharmacol ; 71(9): 1412-1420, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31282010

ABSTRACT

OBJECTIVES: The basic fibroblast growth factor (bFGF)/fibroblast growth factor receptor (FGFR) signal transductional pathway plays an important role not only in tumour, but also in tumour stem cells. Thus, this study was designed to investigate the effects of bFGF signalling on cancer stem cells of lung cancer. METHODS: We blocked bFGF/FGFR signalling in cisplatin (DDP) selected A549 by knocking down bFGF via RNA interference, and subsequently, the stem cell marker of OCT-4 was determined, and cell proliferation, clone formation, invasiveness, apoptosis and drug resistance abilities of DDP selected A549 cells were investigated. KEY FINDINGS: The expressions of bFGF and OCT-4 in DDP selected A549 were higher than that of A549 cells. The findings suggested blocking of bFGF/FGFR signalling resulted in downregulation of bFGF, reduction in cell proliferation, clone formation, invasion and drug resistance abilities, and increase in cell apoptosis. Furthermore, our results also revealed OCT-4 was reduced after bFGF signalling blocking. CONCLUSIONS: In conclusion, our study suggested that bFGF/FGFR signalling plays an important role in maintaining lung cancer stem cell characteristics and regulating expression of cancer stem cell marker of OCT-4.


Subject(s)
Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/metabolism , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Receptors, Fibroblast Growth Factor/physiology , A549 Cells , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation/drug effects , Drug Resistance, Neoplasm/genetics , Fibroblast Growth Factor 2/genetics , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Paclitaxel/pharmacology , RNA, Small Interfering , Signal Transduction/drug effects
16.
Biochem Pharmacol ; 168: 91-99, 2019 10.
Article in English | MEDLINE | ID: mdl-31251937

ABSTRACT

Puerarin, a well-studied isoflavone isolated from Pueraria lobata, produces an antidepressant-like effect. Fibroblast growth factor-2 (FGF-2) is essentially required in the central nervous system as it acts as both a neurotrophic or anti-inflammatory regulator for the proliferation, differentiation and apoptosis of neurons. There is evidence that FGF-2 holds great promise for therapeutic intervention for depression. However, nothing was known about the involvement of FGF-2 in the antidepressant-like effect of puerarin. In the present study, the underlying mechanism of puerarin was evaluated in chronic stress induced depressive-like mice. The results indicated that puerarin treatment was effective to attenuate anhedonia and despair behaviors caused by chronic stress, as the sucrose preference and the immobility time were improved by puerarin. In addition, the results demonstrated that puerarin increased the expression of FGF-2 in the hippocampus. On the contrary, SU5402, an FGFR1 inhibitor, infusion into the brain could not only block the antidepressant-like effect of puerarin, but also abolish the effect of puerarin on hippocampal neurogenesis enhancement and neuroinflammation inhibition. Taken together, these findings provide new insights into the mechanism that the antidepressant-like actions of puerarin require FGF-2/FGFR signaling for the regulation of neurogenesis and neuroinflammation.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Depression/drug therapy , Fibroblast Growth Factor 2/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Isoflavones/pharmacology , Anhedonia/drug effects , Animals , Depression/etiology , Disease Models, Animal , Fibroblast Growth Factor 2/antagonists & inhibitors , Food Preferences/drug effects , Male , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Stress, Psychological/complications
17.
Mol Vis ; 25: 22-34, 2019.
Article in English | MEDLINE | ID: mdl-30804659

ABSTRACT

Purpose: To determine whether the mouse corneal endothelium enters endothelial to mesenchymal transition (EndoMT) following surgical injury in vivo. Methods: The corneal endothelium in anesthetized mice was surgically injured in vivo under direct visualization. The secretion of interleukin-1 beta (IL-1ß) and fibroblast growth factor 2 (FGF2) into the aqueous humor was analyzed with western blotting. The expression of FGF2, Snai1, Zeb1, Col1a1, Col1a2, Fn1, Vim, Cdk2, Ccne1, and Cdh1 was analyzed with semiquantitative RT-PCR in the mouse corneal endothelium ex vivo and in vivo. Knockdown of FGF2 was done using siRNA. Col8a2 was used as a corneal endothelial marker, and Keratocan (Ktcn) was used as a stromal marker. ß-actin was used as a loading control. Results: Sequential expression of IL-1ß and FGF2 was detected in the aqueous humor after surgical injury. FGF2 treatment induced expression of endothelial to mesenchymal transition-related genes including Snai1, and Zeb1 in the mouse ex vivo corneal endothelium. This led to increased expression of Col1a1, Col1a2, Fn1, and Vim and suppression of Cdh1 in a time-dependent manner. Expression of FGF2, Snai1, Zeb1, Col1a1, Col1a2, Fn1, Vim, Cdk2, and Ccne1 was completely abolished by FGF2 siRNA knockdown in the mouse corneal endothelium ex vivo. Surgical injury induced FGF2 expression in the in vivo mouse corneal endothelium. The injury-dependent expression of FGF2, Snai1, Zeb1, Col1a1, Col1a2, Fn1, Vim, Cdk2, and Ccne1 and the suppression of Cdh1 were inhibited by siRNA knockdown of FGF in the mouse corneal endothelium in vivo. Moreover, siRNA knockdown of FGF2 inhibited the formation of the injury-dependent retrocorneal membrane in the in vivo mouse corneal endothelium. Conclusions: These findings suggest that after surgical injury, FGF2 induces the expression of EndoMT-related genes Snai1, Zeb1, Col1a1, Col1a2, Fn1, Vim, Cdk2, and Ccne1 in the mouse corneal endothelium in vivo, similar to the human corneal endothelium ex vivo.


Subject(s)
Cornea/metabolism , Corneal Injuries/genetics , Endothelium/metabolism , Epithelial-Mesenchymal Transition/genetics , Fibroblast Growth Factor 2/genetics , Interleukin-1beta/genetics , Animals , Aqueous Humor/chemistry , Aqueous Humor/metabolism , Cdh1 Proteins/genetics , Cdh1 Proteins/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cornea/pathology , Corneal Injuries/metabolism , Corneal Injuries/pathology , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Endothelium/drug effects , Endothelium/injuries , Epithelial-Mesenchymal Transition/drug effects , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Tissue Culture Techniques , Vimentin/genetics , Vimentin/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
18.
J Biochem ; 165(6): 487-495, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30597085

ABSTRACT

The human fibroblast growth factor-2 (FGF-2) highly expressed in tumours is an important factor to promote tumour angiogenesis and lymphangiogenesis. A disulphide-stabilized diabody (ds-Diabody) could specifically target FGF-2 and show its advantages in inhibition of tumour angiogenesis and growth. It is very important for antibody drugs to confirm the fine epitope. Here, theoretical structure models of FGF-2 and antibody were built by homology modelling. The amino acid residues in the interaction interface of antigen and antibody were analysed by molecular docking. The potential epitope was predicted by homology modelling and molecular docking of antigen-antibody and site-directed mutation assays of alanine scanning. The predicted epitope was verified by antigen mutagenesis and enzyme-linked immunosorbent assay (ELISA). The epitope mapping assay showed that the epitope of ds-Diabody against FGF-2 was defined by the discontinuous sites including six amino acid residues (P23, Q65, R69, G70, Y82 and R118). The results showed that the epitope was localized in the interaction interface of FGF-2 and ds-Diabody. The fine epitope mapping provided the important information for understanding the inhibition activity of ds-Diabody against FGF-2 and helping in the further development of ds-Diabody against FGF-2 as a potentially promising antibody drug for future cancer therapy.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Disulfides/chemistry , Epitope Mapping , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/immunology , Neoplasms/drug therapy , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Antigen-Antibody Reactions , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/genetics , HEK293 Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Neoplasms/immunology , Neoplasms/pathology
19.
Eur J Neurosci ; 50(3): 2552-2561, 2019 08.
Article in English | MEDLINE | ID: mdl-30144335

ABSTRACT

Fibroblast growth factor 2 (FGF2) is a member of the FGF-family, which consists of 22 members, with four known FGF receptors (five in humans). Over the last 30 years, FGF2 has been extensively studied for its role in cell proliferation, differentiation, growth, survival and angiogenesis during development, as well as for its role in adult neurogenesis and regenerative plasticity. Over the past decade, FGF2 has been implicated in learning and memory, as well as in several neuropsychiatric disorders, including anxiety, stress, depression and drug addiction. In this review, we present accumulating evidence indicating the involvement of FGF2 in neuroadaptations caused by drugs of abuse, namely, amphetamine, cocaine, nicotine and alcohol. Moreover, evidence suggests that FGF2 is a positive regulator of alcohol and drug-related behaviors. Thus, although additional studies are yet required, we suggest that reducing FGF2 activity may provide a novel therapeutic approach for substance use disorders.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Illicit Drugs/metabolism , Substance-Related Disorders/metabolism , Animals , Dopamine Agents/administration & dosage , Dopamine Agents/metabolism , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , Illicit Drugs/adverse effects , Learning/drug effects , Learning/physiology , Memory/drug effects , Memory/physiology , Substance-Related Disorders/drug therapy
20.
Eur Rev Med Pharmacol Sci ; 22(24): 8866-8876, 2018 12.
Article in English | MEDLINE | ID: mdl-30575929

ABSTRACT

OBJECTIVE: The aim of this study was to investigate whether hypoxia in vivo can induce hypoxic pulmonary hypertension by inhibiting the activation of FGF2 by miR-203. MATERIALS AND METHODS: We established a rat model of hypoxic pulmonary hypertension (HPH), and measured the right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (right ventricular hypertrophy index). The ventricular hypertrophy index (RVHI) was calculated and HE staining of the lung tissue of HPH rats was performed. We extracted pulmonary arterial smooth muscle cells (PASMCs) from rats and identified them by immunofluorescence assay. The expression of miR-203 in hypoxic PASMCs was detected by quantitative Real time-polymerase chain reaction (qRT-PCR). The proliferation and migration of PASMCs were detected by EDU (5-Ethynyl-2'-deoxyuridine), cell counting kit-8 (CCK-8) and scratch assay, respectively. Dual Luciferase reporting assay and Western blot were used to detect the binding of miR-203 and FGF2. RESULTS: The results of qRT-PCR showed that miR-203 expression in rat PASMCs was significantly lower than that in normoxia control group at 24 h and 48 h after hypoxic treatment. EDU, CCK8 and scratch test results showed that proliferation and migration ability of PASMCs were weakened after overexpression of miR-203, and vice versa. Dual Luciferase reporter gene assays and Western blot experiments showed that miR-203 could target and combine with FGF2 to inhibit its expression. In vivo experiments showed that low expression of FGF2 could lead to decreased RVSP and RVHI, decreased FGF2 protein levels, and decreased WT% and (PM+FM)% in hypoxia-treated rats. CONCLUSIONS: Hypoxia in vivo is involved in the development of HPH by inhibiting the activation of FGF2 by miR-203. Meanwhile, specific inhibition of FGF2 can reduce hypoxia-induced pulmonary hypertension and improve pulmonary vascular remodeling.


Subject(s)
Fibroblast Growth Factor 2/antagonists & inhibitors , Hypertension, Pulmonary/etiology , Hypoxia/complications , MicroRNAs/physiology , Animals , Cells, Cultured , Fibroblast Growth Factor 2/physiology , Male , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...