Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hepatology ; 67(1): 247-259, 2018 01.
Article in English | MEDLINE | ID: mdl-28802066

ABSTRACT

Cholangiocyte senescence has been linked to primary sclerosing cholangitis (PSC). Persistent secretion of growth factors by senescent cholangiocytes leads to the activation of stromal fibroblasts (ASFs), which are drivers of fibrosis. The activated phenotype of ASFs is characterized by an increased sensitivity to apoptotic stimuli. Here, we examined the mechanisms of apoptotic priming in ASFs and explored a combined targeting strategy to deplete senescent cholangiocytes and ASFs from fibrotic tissue to ameliorate liver fibrosis. Using a coculture system, we determined that senescent cholangiocytes promoted quiescent mesenchymal cell activation in a platelet-derived growth factor (PDGF)-dependent manner. We also identified B-cell lymphoma-extra large (Bcl-xL) as a key survival factor in PDGF-activated human and mouse fibroblasts. Bcl-xL was also up-regulated in senescent cholangiocytes. In vitro, inhibition of Bcl-xL by the small molecule Bcl-2 homology domain 3 mimetic, A-1331852, or Bcl-xL-specific small interfering RNA induced apoptosis in PDGF-activated fibroblasts, but not in quiescent fibroblasts. Likewise, inhibition of Bcl-xL reduced the survival and increased apoptosis of senescent cholangiocytes, compared to nonsenescent cells. Treatment of multidrug resistance 2 gene knockout (Mdr2-/- ) mice with A-1331852 resulted in an 80% decrease in senescent cholangiocytes, a reduction of fibrosis-inducing growth factors and cytokines, decrease of α-smooth muscle actin-positive ASFs, and finally in a significant reduction of liver fibrosis. CONCLUSION: Bcl-xL is a key survival factor in ASFs as well as in senescent cholangiocytes. Treatment with the Bcl-xL-specific inhibitor, A-1331852, reduces liver fibrosis, possibly by a dual effect on activated fibroblasts and senescent cholangiocytes. This mechanism represents an attractive therapeutic strategy in biliary fibrosis. (Hepatology 2018;67:247-259).


Subject(s)
Benzothiazoles/pharmacology , Bile Ducts/cytology , Cholangitis, Sclerosing/pathology , Fibroblasts/drug effects , Isoquinolines/pharmacology , Platelet-Derived Growth Factor/drug effects , Animals , Biopsy, Needle , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured/drug effects , Cellular Senescence/drug effects , Cholangitis, Sclerosing/drug therapy , Disease Models, Animal , Drug Resistance, Multiple , Fibroblasts/metabolism , Fibroblasts/pathology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Targeted Therapy , Platelet-Derived Growth Factor/metabolism , Random Allocation , Reference Values
2.
Med Sci Monit ; 24: 4547-4554, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29961751

ABSTRACT

BACKGROUND The aim of this study was to evaluate the effects of 3-tetrazolyl methyl-3-hydroxy-oxindole hybrid (THOH) on cell proliferation, apoptosis, and the cell cycle in human lung cancer cell lines SK-LU-1, A549, and A-427, and the normal lung fibroblast cell line, MRC-5, in vitro. MATERIAL AND METHODS Human lung adenocarcinoma cells SK-LU-1, A549, and A-427, and the normal lung fibroblast cells, MRC-5 were cultured and treated with increasing concentrations of 10 mM of a stock solution of THOH in dimethyl sulfoxide (DMSO). An MTT cell proliferation assay was used. Cell apoptosis and the cell cycle were studied using fluorescence-activated cell sorting (FACs) with fluorescein isothiocyanate (FITC), Annexin-V, propidium iodide (PI), and nuclear staining with 4',6-diamidino-2-phenylindole (DAPI). DNA damage was measured using the comet (single-cell gel electrophoresis) assay. Cell migration was evaluated using a wound healing assay, and Western blotting was used to measure protein expression levels. RESULTS Treatment of SK-LU-1 cells with THOH inhibited cell migration. Treatment of lung cancer cells, SK-LU-1, A549, and A-427, with THOH inhibited cell proliferation, with the most marked inhibition found in the SK-LU-1 lung cancer cells (IC50, 12 µM). Treatment of lung cancer cells, SK-LU-1, A549, and A-427, with THOH increased cell apoptosis, resulted in G2/M cell cycle arrest, and inhibited both the platelet-derived growth factor D (PDGF-D) and MEK/ERK signaling pathways. CONCLUSIONS Treatment of adenocarcinoma cells, SK-LU-1, A549, and A-427, with THOH inhibited cell proliferation, apoptosis, and resulted in G2/M cell cycle arrest by targeting PDGF-D and the MEK/ERK signaling pathway.


Subject(s)
Indoles/pharmacology , Lymphokines/drug effects , MAP Kinase Signaling System/drug effects , Platelet-Derived Growth Factor/drug effects , Tetrazoles/pharmacology , A549 Cells/drug effects , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Indoles/metabolism , Lung Neoplasms/pathology , Oxindoles , Signal Transduction , Tetrazoles/metabolism
3.
J Nat Prod ; 80(7): 2146-2150, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28627872

ABSTRACT

Xanthohumol (1) is a principal prenylated chalcone found in hops. The aim of this study was to examine its influence on platelet-derived growth factor (PDGF)-BB-triggered vascular smooth muscle cell (VSMC) proliferation and migration in vitro and on experimentally induced neointima formation in vivo. Quantification of resazurin conversion indicated that 1 can inhibit PDGF-BB-induced VSMC proliferation concentration-dependently (IC50 = 3.49 µM). Furthermore, in a wound-healing assay 1 potently suppresses PDGF-BB-induced VSMC migration at 15 µM. Tested in a mouse femoral artery cuff model, 1 significantly reduces neointima formation. Taken together, we show that 1 represses PDGF-BB-induced VSMC proliferation and migration in vitro as well as neointima formation in vivo. This novel activity suggests 1 as an interesting candidate for further studies addressing a possible therapeutic application to counteract vascular proliferative disease.


Subject(s)
Flavonoids/pharmacology , Humulus/chemistry , Neointima/metabolism , Propiophenones/pharmacology , Animals , Becaplermin , Cell Movement , Cell Proliferation , Disease Models, Animal , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/isolation & purification , MAP Kinase Signaling System , Mice , Molecular Structure , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/chemically induced , Oxazines/metabolism , Phosphorylation , Platelet-Derived Growth Factor/drug effects , Propiophenones/chemistry , Propiophenones/isolation & purification , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Signal Transduction , Wound Healing/drug effects , Xanthenes/metabolism
4.
J Clin Periodontol ; 41(3): 295-302, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24433307

ABSTRACT

AIM: We hypothesized that platelet inactivation induced by drugs might interfere with periodontal repair in experimental periodontitis by suppressing the release of biological mediators from platelets at the site of injury. MATERIAL AND METHODS: Sixty rats were randomly assigned to six groups (n = 10) and ligatures were placed around lower first molars of three groups. The other three groups were used as negative controls. Ligatures were removed after 10 days of periodontitis induction and all groups were submitted to treatment with aspirin (Asp) (30 mg/kg), clopidogrel (Clop) (75 mg/kg) or NaCl 0.9% intra-gastrically once daily for 3 days. Periodontal tissue was assessed by the measurement of CXCL12, CXCL4, CCL5 and platelet-derived growth factor (PDGF) by enzyme-linked immunosorbent assay; histomorphometrical analysis of polymorphonuclear (PMN) infiltration, attachment loss, bone loss and osteoclast numbers and quantification of blood vessels by imunnohistochemistry. RESULTS: During periodontal repair and treatment with NaCl 0.9%, CCL5 was decreased and CXCL12 increased when compared with negative control groups. Asp and Clop did not affect CCL5 expression, decreased CXCL12 but only Clop decreased CXCL4 and PDGF content compared with saline-treated animals. Clop increased blood vessel number, reduced PMN count and decreased attachment and bone loss, also decreased osteoclast number in animals submitted or not to periodontal repair. CONCLUSION: Systemic administration of Clop for 3 days improved the repair process associated with experimental periodontal disease, suggesting that it may have therapeutic value under situations where tissues undergo a transition from inflammation to repair.


Subject(s)
Periodontitis/drug therapy , Periodontium/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Ticlopidine/analogs & derivatives , Alveolar Bone Loss/drug therapy , Animals , Aspirin/administration & dosage , Aspirin/therapeutic use , Cell Count , Chemokine CCL5/drug effects , Chemokine CXCL12/drug effects , Clopidogrel , Infusions, Parenteral , Male , Microvessels/drug effects , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Osteoclasts/drug effects , Periodontal Attachment Loss/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Platelet Factor 4/drug effects , Platelet-Derived Growth Factor/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Sodium Chloride , Ticlopidine/administration & dosage , Ticlopidine/therapeutic use
5.
BMC Pulm Med ; 14: 135, 2014 Aug 09.
Article in English | MEDLINE | ID: mdl-25108547

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is characterized by overwhelming inflammatory responses and lung remodeling. We hypothesized that leukocyte infiltration during the inflammatory response modulates epithelial remodeling through a mechanism of epithelial-mesenchymal transition (EMT). METHODS: Human lung epithelial cells were treated for 30 min with hydrochloric acid (HCl). Human monocytes were then cocultured with the epithelial cells for up to 48 h, in the presence or absence of blocking peptides against lymphocyte function-associated antigen-1 (LFA-1), or tyrphostin A9, a specific inhibitor for platelet-derived growth factor (PDGF) receptor tyrosine kinase. RESULTS: Exposure of lung epithelial cells to HCl resulted in increased expression of intercellular adhesion molecule-1 (ICAM-1) and production of interleukin (IL)-8 at 24 h. The expression of the epithelial markers E-cadherin decreased while the mesenchymal markers vimentin and α-smooth muscle actin (α-SMA) increased at 24 h and remained high at 48 h. The addition of monocytes augmented the profiles of lower expression of epithelial markers and higher mesenchymal markers accompanied by increased collagen deposition. This EMT profile was associated with an enhanced production of IL-8 and PDGF. Treatment of the lung epithelial cells with the LAF-1 blocking peptides CD11a237-246 or/and CD18112-122 suppressed monocyte adhesion, production of IL-8, PDGF and hydroxyproline as well as EMT markers. Treatment with tyrphostin A9 prevented the EMT profile shift induced by HCl stimulation. CONCLUSIONS: The interaction between epithelial cells and monocytes enhanced epithelial remodelling after initial injury through EMT signalling that is associated with the release of soluble mediators, including IL-8 and PDGF.


Subject(s)
Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Monocytes , Actins/metabolism , Alveolar Epithelial Cells/physiology , CD11a Antigen , CD18 Antigens , Cadherins/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Coculture Techniques , Humans , Hydrochloric Acid/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Lymphocyte Function-Associated Antigen-1/drug effects , Peptides/pharmacology , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/metabolism , Tyrphostins/pharmacology , Vimentin/metabolism
6.
J Hepatol ; 55(3): 612-625, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21251937

ABSTRACT

BACKGROUND & AIMS: Nilotinib is a novel tyrosine kinase inhibitor of Bcr-Abl and other kinases. In this study, we have examined its activity as an anti-fibrotic agent. METHODS: The in vitro effect of Nilotinib on rat and human HSCs was assessed using proliferation assays and Western blotting. The in vivo antifibrotic efficacy of Nilotinib was assessed in mice with liver fibrosis induced by CCl(4) and bile duct ligation (BDL). RESULTS: Nilotinib inhibited proliferation, migration, and actin filament formation, as well as the expression of α-SMA and collagen in activated HSCs. Nilotinib induced apoptosis of HSCs, which was correlated with reduced bcl-2 expression, increased p53 expression, cleavage of PARP, as well as increased expression of PPARγ and TRAIL-R. Nilotinib also induced cell cycle arrest, accompanied by increased expression of p27 and downregulation of cyclin D1. Interestingly, Nilotinib not only inhibited activation of PDGFR, but also TGFRII through Src. Nilotinib significantly inhibited PDGF and TGFß-simulated phosphorylation of ERK and Akt. Furthermore, PDGF- and TGFß-activated phosphorylated form(s) of Abl in human HSCs were inhibited by Nilotinib. In vivo, Nilotinib reduced collagen deposition and α-SMA expression in CCl(4) and BDL-induced fibrosis. These beneficial effects were associated with suppressed expression of procollagen-(I), TIMP-1, CD31, CD34, VEGF, and VEGFR. Nilotinib could induce HSC undergoing apoptosis in vivo, which was correlated with downregulation of bcl-2. We also observed reduced expression of phosphorylated ERK, Akt, and Abl in the Nilotinib-treated CCl(4) and BDL livers. In addition to its antifibrotic activity, the drug was hepatoprotective and reduced the elevations of ALT and AST after CCl(4) and BDL. CONCLUSIONS: These studies uncover a novel role of Bcr-Abl activity in treatment of liver fibrosis through multiple mechanisms and indicate that Nilotinib represents a potentially effective antifibrotic agent.


Subject(s)
Genes, abl/drug effects , Liver Cirrhosis/pathology , Platelet-Derived Growth Factor/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/drug effects , Actins/metabolism , Animals , Apoptosis/drug effects , Bile Ducts , Carbon Tetrachloride , Cell Proliferation/drug effects , Collagen/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Ligation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred BALB C , PPAR gamma/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
7.
J Pharmacol Sci ; 116(2): 204-13, 2011.
Article in English | MEDLINE | ID: mdl-21685708

ABSTRACT

The abnormal proliferation of vascular smooth muscle cells (VSMCs) in arterial wall is an important pathogenic factor for vascular disorders such as atherosclerosis and restenosis after angioplasty. The present study was designed to investigate the inhibitory effects of docetaxel on VSMC proliferation, as well as the molecular mechanism of this inhibition. Docetaxel at 10, 20 and 40 µM significantly inhibited both the proliferation and the DNA synthesis of fetal bovine serum (FBS)- and platelet-derived growth factor (PDGF)-BB-stimulated VSMCs in a concentration-dependent manner. In accordance with these findings, docetaxel blocked the FBS- and PDGF-BB-induced progression of synchronized cells through the G0/G1 phase of the cell cycle. Docetaxel also decreased the expressions of cell cycle-related proteins, including cyclin-dependent kinase (CDK) 2, cyclin E, CDK4, cyclin D1, retinoblastoma protein, and proliferative cell nuclear antigen in PDGF-BB-stimulated VSMCs. Docetaxel significantly inhibited the phosphorylation of extracellular signal-regulated kinase 1/2, Akt, and phospholipase C-γ1, downstream molecule in the PDGF-BB signaling pathway. Docetaxel suppressed the phosphorylation of PDGF receptor (PDGF-R) ß, the upstream molecule in PDGF-BB signaling cascade, suggesting that the inhibitory effect of docetaxel on the proliferation of VSMCs may occur by blocking PDGF-Rß phosphorylation. Thus, docetaxel may be a potential antiproliferative agent for the treatment of atherosclerosis and angioplasty restenosis.[Supplementary Figures: available only at http://dx.doi.org/10.1254/jphs.10276FP].


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/physiology , Taxoids/pharmacology , Animals , Becaplermin , Blotting, Western , Cell Cycle/drug effects , Cells, Cultured , DNA Replication/drug effects , Docetaxel , Muscle, Smooth, Vascular/cytology , Phosphorylation , Platelet-Derived Growth Factor/drug effects , Proto-Oncogene Proteins c-sis , Rats
8.
Clin Oral Implants Res ; 22(3): 338-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21561475

ABSTRACT

OBJECTIVES: To assess the influence of nicotine on the proliferation and gene expression of osteogenic and angiogenic mediators of osteoblasts. MATERIAL AND METHODS: Rabbit primary osteoblasts were exposed to various concentrations of nicotine (0.001, 0.1 and 10 µmol/l). The cell proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. The gene expression of transforming growth factor (TGF)-ß(1), bone morphogenetic protein (BMP)-2, platelet-derived growth factor (PDGF)-AA and vascular endothelial growth factor (VEGF) was evaluated using real-time reverse transcription - polymerase chain reaction. RESULTS: The osteoblast proliferation was inhibited by nicotine at the concentration of 0.001-10 µM at 48 and 72 h of culture, but with no significant effect at 24 h. The expression of TGF-ß(1), BMP-2, PDGF-AA and VEGF was inhibited by nicotine at the concentrations of 0.1 and 10 µM, but with no significant difference at the low concentration of 0.001 µM. CONCLUSIONS: Nicotine suppresses osteoblast proliferation and inhibits the expression of some key osteogenic and angiogenic mediators in the in vitro experimental model. These inhibitory effects of nicotine on the osteoblast activity may reflect, to a certain degree, the overall detrimental effects of tobacco use on the survival rate of dental implants.


Subject(s)
Nicotine/pharmacology , Osteoblasts/drug effects , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/drug effects , Cell Culture Techniques , Cell Proliferation/drug effects , Coloring Agents , Neovascularization, Physiologic/drug effects , Nicotine/administration & dosage , Osteogenesis/drug effects , Phenotype , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/drug effects , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Thiazoles , Time Factors , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/drug effects
9.
Respiration ; 81(5): 420-30, 2011.
Article in English | MEDLINE | ID: mdl-21502778

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is characterized by progressive fibrosis and a poor prognosis. Alveolar epithelial cells (AECs) are considered to play important roles by releasing growth factors and matrix metalloproteinases (MMPs) and by being involved in epithelial mesenchymal transition in IPF. Doxycycline hydrochloride (DOXY), an inhibitor of MMPs, attenuates pulmonary fibrosis in models and in patients with IPF; however, the mechanism of this action remains obscure. OBJECTIVES: The present study investigated the effect of DOXY on growth factors and MMP production in AECs. METHODS: Bleomycin (BL)-induced murine pulmonary fibrosis was treated with DOXY and examined by pathological and immunohistochemical staining. The human alveolar epithelial cell line A549 was stimulated with transforming growth factor (TGF)-ß1 and incubated with DOXY, and then the expression of growth factors, MMPs, and collagen type I was evaluated at the mRNA and protein levels. We also evaluated the effects of DOXY on the TGF-ß1-induced Smad signaling pathway. RESULTS: DOXY reduced fibrosis scores and the production of collagen type I, connective tissue growth factor (CTGF), and TGF-ß1 in BL models. DOXY inhibited the mRNA expression of MMP-2, MPP-9, CTGF, and collagen type I as well as the production of MMP-2 and platelet-derived growth factor-AA protein induced in A549 cells by TGF-ß1 but not by Smad2 and Smad3 phosphorylation. We did not find a similar effect of DOXY in normal lung fibroblasts. CONCLUSIONS: Our results suggest that DOXY could be useful for attenuating pulmonary fibrosis through the inhibition of growth factors and MMP production in AECs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Epithelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinases/drug effects , Matrix Metalloproteinases/metabolism , Pulmonary Alveoli/cytology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Animals , Blotting, Western , Cell Line , Collagen Type I/drug effects , Collagen Type I/metabolism , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lung/pathology , Male , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred ICR , Phosphorylation , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Smad Proteins/physiology , Transforming Growth Factor beta1/pharmacology
10.
Drug Deliv ; 28(1): 2051-2061, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34595978

ABSTRACT

Fibrotic diseases pose significant clinical challenges due to their broadness and complexity. Thus, a better understanding of fibrogenesis and the development of more effective treatments is imperative. Recent evidence suggests a significant antifibrotic potential of an endogenous glycoprotein, endostatin. While endostatin has been widely studied for its role as an anticancer adjuvant by inhibiting tumor angiogenesis, its possible implication in fibrosis remains largely unclear. Here, we review the role of endostatin in various cellular processes and highlight its antifibrotic activity. We hypothesize that endostatin conveys a homeostatic function in the process of fibrosis by regulating (a) TGF-ß1 and its downstream signaling; (b) RhoA/ROCK pathway; (c) NF-κB signaling pathway; (d) expression of EGR-1; (e) PDGF/PDGFR pathway; (f) autophagy-related pathways; (g) pathways associated with cell proliferation and apoptosis. Finally, we propose a schematic model of the antifibrotic roles and mechanisms of endostatin; also, we outline future research directions of endostatin and aim to present a potential therapeutic approach for fibrosis.


Subject(s)
Endostatins/pharmacology , Fibrosis/drug therapy , Transforming Growth Factor beta1/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Cell Proliferation/drug effects , NF-kappa B/drug effects , Platelet-Derived Growth Factor/drug effects , Signal Transduction/drug effects , rhoA GTP-Binding Protein/drug effects
11.
J Cell Biochem ; 111(2): 433-41, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20518073

ABSTRACT

In previous in vitro studies, we showed that imatinib abrogated platelet-derived growth factor receptor α (PDGFRα) signaling, disrupting both breast cancer and smooth muscle cells (SMC). PDGF is also a powerful mitogen for neural crest origin cells like melanocytes. The purpose of the present study was to evaluate the effect of imatinib on melanoma growth and in angiogenesis, with emphasis to the involvement in PDGF signaling. B16 melanoma cells incubation with 5 µM (IC50) imatinib resulted in a significant reduction in cell proliferation and migration. Apoptosis, however, was not significantly affected. Phosphorylated-PDGFRα expression was decreased in B16 lysates. In a mouse model of B16 melanoma, intraperitoneal administration of imatinib at early day light significantly decreased tumor growth. These findings were corroborated by a highly significant reduction in cell proliferation and increase in apoptosis in melanoma tumors. This was accompanied by a decrease in microvessel density and in the number of SMC-presenting vessels. Imatinib further inhibited PDGFRα expression and activity, as confirmed by the down-regulation of downstream Erk signaling pathway. Altogether, this study demonstrates that besides targeting tumor cells, imatinib also prevents vascular integrity. The current study provides evidence that the paracrine crosstalk between tumor cells and host neighboring cells is crucial for the elucidation of imatinib effects. In addition, the fact that this molecule targets vascular support cells further enlarges its therapeutic purpose to a wide range of vasculoproliferative pathologies.


Subject(s)
Melanoma, Experimental/drug therapy , Myocytes, Smooth Muscle/drug effects , Piperazines/pharmacology , Platelet-Derived Growth Factor/drug effects , Pyrimidines/pharmacology , Signal Transduction/drug effects , Animals , Benzamides , Imatinib Mesylate , Melanoma, Experimental/metabolism , Mice , Muscle, Smooth, Vascular/pathology , Neovascularization, Pathologic , Paracrine Communication , Piperazines/therapeutic use , Platelet-Derived Growth Factor/metabolism , Pyrimidines/therapeutic use
12.
J Cardiovasc Pharmacol ; 55(1): 39-48, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786891

ABSTRACT

Idiopathic pulmonary arterial hypertension (IPAH) is a progressive disease characterized by inappropriate increase of pulmonary artery smooth muscle cells (PASMCs) leading to occlusion of pulmonary arterioles. Inhibition of platelet-derived growth factor (PDGF) signaling is starting to garner attention as a targeted therapy for IPAH. We assessed the inhibitory effects of simvastatin, a 3-hydroxy-3-methylglutanyl coenzyme A reductase inhibitor, on PDGF-induced proliferation and migration of PASMCs obtained from 6 patients with IPAH who underwent lung transplantation. PDGF stimulation caused a significantly higher growth rate of PASMCs from patients with IPAH than that of normal control PASMCs as assessed by (3)H-thymidine incorporation. Simvastatin (0.1 micromol/L) significantly inhibited PDGF-induced cell proliferation of PASMCs from patients with IPAH but did not inhibit proliferation of normal control cells at the same concentration. Western blot analysis revealed that simvastatin significantly increased the expression of cell cycle inhibitor p27. PDGF significantly increased the migration distance of IPAH-PASMCs compared with that of normal PASMCs, and simvastatin (1 micromol/L) significantly inhibited PDGF-induced migration. Immunofluorescence staining revealed that simvastatin (1 micromol/L) inhibited translocation of Rho A from the cytoplasm to membrane and disorganized actin fibers in PASMCs from patients with IPAH. In conclusion, simvastatin had inhibitory effects on inappropriate PDGF signaling in PASMCs from patients with IPAH.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypertension, Pulmonary/drug therapy , Platelet-Derived Growth Factor/drug effects , Simvastatin/pharmacology , Adolescent , Adult , Aged , Cell Movement/drug effects , Cell Proliferation/drug effects , Child , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Platelet-Derived Growth Factor/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Signal Transduction/drug effects , Young Adult
13.
Br J Nutr ; 103(3): 378-85, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19818196

ABSTRACT

We investigated the effects of a high-cholesterol (HC) diet administered long term (25 or 55 weeks) on metabolic disorders including hepatic damage in mice. The mice were fed the HC diet (15 % milk fat, 1.5 % cholesterol and 0.1 % cholic acid, w/w) for 25 or 55 weeks. Body and adipose tissue weights were similar to those of mice fed a control diet. Consumption of the HC diet long term resulted in hypercholesterolaemia, hepatic steatosis and gallstones. In addition, focal nodular hyperplasia (FNH) and mild fibrosis of the liver developed in all mice fed the HC diet for 55 weeks. Plasma levels of monocyte chemoattractant protein (MCP)-1 were elevated, and the level of hepatic platelet-derived growth factor (PDGF)-B protein was increased in mice fed the HC diet compared with those fed the control diet. Thus, it seems likely that the liver fibrosis and FNH caused by the long-term consumption of a HC diet may be partly due to an elevation of plasma MCP-1 and hepatic PDGF expression.


Subject(s)
Cholesterol, Dietary/pharmacology , Fatty Liver/chemically induced , Gallstones/chemically induced , Liver Cirrhosis/chemically induced , Liver/pathology , Animals , Becaplermin , Body Weight/drug effects , Chemokine CCL2/blood , Chemokine CCL2/drug effects , Cholesterol/metabolism , Cytokines/metabolism , Gallstones/metabolism , Glucose Tolerance Test , Lipids/physiology , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Reference Values
14.
Przegl Lek ; 67(6): 413-6, 2010.
Article in Polish | MEDLINE | ID: mdl-21344772

ABSTRACT

Because of unsatisfactory treatment results in high risk neuroblastoma, it is necessary to find new, effective and safe, treatment options. Tyrosine kinase inhibitors, including imatinib, are one of the most profoundly examinated drugs. Its employment in neuroblastoma treatment is potentially possible because of expression of c-kit and PDGFR, which are cellular targets of imatinib. It was shown that this drug inhibits growth of neuroblastoma cell lines both in vivo and in vitro. The prognostic meaning of receptors expression is still not clear and depends on the method used for evaluation. The only found phase II study did not reveal the satisfactory response for imanitinib in treatment of resistant neuroblastoma, but the presence of receptors expression was not inclusion criteria. However, it seems that assuming the correct patient's eligibility criteria and proper dosage schedule, possible in combination with chemotherapy, imatinib may become safe and effective treatment modality.


Subject(s)
Biomarkers, Tumor/analysis , Neuroblastoma/diagnosis , Neuroblastoma/drug therapy , Piperazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/analysis , Pyrimidines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Imatinib Mesylate , Neuroblastoma/chemistry , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/drug effects , Prognosis , Proto-Oncogene Proteins c-kit/drug effects
15.
Inflamm Bowel Dis ; 26(5): 678-686, 2020 04 11.
Article in English | MEDLINE | ID: mdl-31943022

ABSTRACT

BACKGROUND: Intestinal fibrosis is a hallmark of Crohn's disease. Here, we investigated the impact of several putative antifibrotic compounds on the expression of fibrosis markers using murine precision-cut intestinal slices. METHODS: Murine precision-cut intestinal slices were cultured for 48 hours in the presence of profibrotic and/or antifibrotic compounds. The fibrotic process was studied on gene and protein level using procollagen 1a1 (Col1α1), heat shock protein 47 (Hsp47), fibronectin (Fn2), and plasminogen activator inhibitor-1 (Pai-1). The effects of potential antifibrotic drugs mainly inhibiting the transforming growth factor ß (TGF-ß) pathway (eg, valproic acid, tetrandrine, pirfenidone, SB203580, and LY2109761) and compounds mainly acting on the platelet-derived growth factor (PDGF) pathway (eg, imatinib, sorafenib, and sunitinib) were assessed in the model at nontoxic concentrations. RESULTS: Murine precision-cut intestinal slices remained viable for 48 hours, and an increased expression of fibrosis markers was observed during culture, including Hsp47, Fn2, and Pai-1. Furthermore, TGF-ß1 stimulated fibrogenesis, whereas PDGF did not have an effect. Regarding the tested antifibrotics, pirfenidone, LY2109761, and sunitinib had the most pronounced impact on the expression of fibrosis markers, both in the absence and presence of profibrotic factors, as illustrated by reduced levels of Col1α1, Hsp47, Fn2, and Pai-1 after treatment. Moreover, sunitinib significantly reduced Hsp47 and Fn2 protein expression and the excretion of procollagen 1. CONCLUSIONS: Precision-cut intestinal slices can successfully be used as a potential preclinical screening tool for antifibrotic drugs. We demonstrated that sunitinib reduced the expression of several fibrosis markers, warranting further evaluation of this compound for the treatment of intestinal fibrosis.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Gastrointestinal Agents/pharmacology , Intestines/drug effects , Signal Transduction/drug effects , Animals , Collagen Type I/drug effects , Collagen Type I, alpha 1 Chain , Crohn Disease/drug therapy , Crohn Disease/pathology , Fibronectins/drug effects , Fibrosis/drug therapy , Fibrosis/pathology , HSP47 Heat-Shock Proteins/drug effects , Intestines/pathology , Mice , Platelet-Derived Growth Factor/drug effects , Serpin E2/drug effects , Sunitinib/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors
16.
Nat Commun ; 11(1): 3704, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709869

ABSTRACT

FGF-2 displays multifarious functions in regulation of angiogenesis and vascular remodeling. However, effective drugs for treating FGF-2+ tumors are unavailable. Here we show that FGF-2 modulates tumor vessels by recruiting NG2+ pricytes onto tumor microvessels through a PDGFRß-dependent mechanism. FGF-2+ tumors are intrinsically resistant to clinically available drugs targeting VEGF and PDGF. Surprisingly, dual targeting the VEGF and PDGF signaling produces a superior antitumor effect in FGF-2+ breast cancer and fibrosarcoma models. Mechanistically, inhibition of PDGFRß ablates FGF-2-recruited perivascular coverage, exposing anti-VEGF agents to inhibit vascular sprouting. These findings show that the off-target FGF-2 is a resistant biomarker for anti-VEGF and anti-PDGF monotherapy, but a highly beneficial marker for combination therapy. Our data shed light on mechanistic interactions between various angiogenic and remodeling factors in tumor neovascularization. Optimization of antiangiogenic drugs with different principles could produce therapeutic benefits for treating their resistant off-target cancers.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibroblast Growth Factor 2/drug effects , Fibroblast Growth Factor 2/metabolism , Neoplasms/drug therapy , Platelet-Derived Growth Factor/drug effects , Vascular Endothelial Growth Factor A/drug effects , Animals , Biomarkers, Tumor , Blood Pressure , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Capillary Permeability , Cell Proliferation , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Therapy, Combination , Humans , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta , Signal Transduction/drug effects , Tumor Hypoxia , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor A/metabolism
17.
J Appl Oral Sci ; 28: e20190215, 2020.
Article in English | MEDLINE | ID: mdl-31939521

ABSTRACT

OBJECTIVE: This study evaluated the angiogenesis-enhancing potential of a tricalcium silicate-based mineral trioxide aggregate (ProRoot MTA), Biodentine, and a novel bioceramic root canal sealer (Well-Root ST) in human dental pulp stem cells (hDPSCs), human periodontal ligament stem cells (hPLSCs), and human tooth germ stem cells (hTGSCs). METHODOLOGY: Dulbecco's modified Eagle's medium was conditioned for 24 h by exposure to ProRoot MTA, Biodentine, or Well-Root ST specimens (prepared according to the manufacturers' instructions). The cells were cultured in these conditioned media and their viability was assessed with 3-(4,5-dimethyl-thiazol-2-yl)-5-(3-carboxy-methoxy-phenyl)-2-(4-sulfo-phenyl)-2H tetrazolium (MTS) on days 1, 3, 7, 10, and 14. Angiogenic growth factors [platelet-derived growth factor (PDGF), basic fibroblast growth factor (FGF-2), and vascular endothelial growth factor (VEGF)] were assayed by sandwich enzyme-linked immunosorbent assay (ELISA) on days 1, 7, and 14. Human umbilical vein endothelial cell (HUVEC) migration assays were used to evaluate the vascular effects of the tested materials at 6-8 h. Statistical analyses included Kruskal-Wallis, Mann-Whitney U, and Friedman and Wilcoxon signed rank tests. RESULTS: None of tricalcium silicate-based materials were cytotoxic and all induced a similar release of angiogenic growth factors (PDGF, FGF-2, and VEGF) (p>0.05). The best cell viability was observed for hDPSCs (p<0.05) with all tricalcium silicate-based materials at day 14. Tube formation by HUVECs showed a significant increase with all tested materials (p<0.05). CONCLUSION: The tricalcium silicate-based materials showed potential for angiogenic stimulation of all stem cell types and significantly enhanced tube formation by HUVECs.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Calcium Compounds/pharmacology , Ceramics/pharmacology , Root Canal Filling Materials/pharmacology , Silicates/pharmacology , Stem Cells/drug effects , Biocompatible Materials/pharmacology , Cell Survival/drug effects , Dental Pulp/cytology , Dental Pulp/drug effects , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/drug effects , Flow Cytometry , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Materials Testing , Neovascularization, Physiologic/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/drug effects , Reproducibility of Results , Statistics, Nonparametric , Tooth Germ/cytology , Tooth Germ/drug effects , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/drug effects
18.
FASEB J ; 22(7): 2151-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18272655

ABSTRACT

Understanding growth factor function during wound repair is necessary for the development of therapeutic interventions to improve healing outcomes. In the current study, we compare the effects of serum and purified growth factors on human fibroblast function in three different collagen matrix models: cell migration in nested matrices, floating matrix contraction, and stressed-released matrix contraction. The results of these studies indicate that platelet-derived growth factor (PDGF) is unique in its capacity to promote cell migration. Serum, lysophosphatidic acid, sphingosine-1-phophate (S1P), and endothelin-1 promote stressed-released matrix contraction but not cell migration. In addition, we found that S1P inhibits fibroblast migration and treatment of serum to remove lipid growth factors or treatment of cells to interfere with S1P(2) receptor function increases serum promigratory activity. Our findings suggest that different sets of growth factors generate promigratory and procontractile tissue environments for fibroblasts and that the balance between PDGF and S1P is a key determinant of fibroblast promigratory activity.


Subject(s)
Collagen/chemistry , Collagen/physiology , Fibroblasts/physiology , Cell Culture Techniques , Cell Movement/physiology , Collagen/drug effects , Fibroblasts/cytology , Foreskin , Gene Silencing , Growth Substances/pharmacology , Humans , Lysophospholipids/pharmacology , Lysophospholipids/physiology , Male , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/physiology , Receptors, Lysosphingolipid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine/physiology , Wound Healing/physiology
19.
Acta Odontol Scand ; 67(2): 65-73, 2009.
Article in English | MEDLINE | ID: mdl-19031159

ABSTRACT

OBJECTIVE: Periodontal regeneration is histologically defined as regeneration of the tooth supporting structures, including alveolar bone, periodontal ligament, and cementum. Cells in the remaining periodontal tissues need optimal conditions if they are to perform their functions in the regeneration process. The present study is an investigation of the molecular effects of ABM/P-15 on human periodontal ligament cells (PDL) in vitro. MATERIAL AND METHODS: PDL cells obtained from healthy subjects were used for in vitro experiments. Cell proliferation, morphology, and mineralization using Von kossa staining were evaluated. mRNA expressions for transforming growth factor-beta (TGF-beta), insulin-like growth factor-I (IGF-I), basic fibroblast growth factor (b-FGF), vascular endothelial growth factor (VEGF), bone morphogenic protein-2 (BMP-2), platelet-derived growth factor (PDGF), and type 1 collagen (COL1) were assessed on days 3 and 7 using RT-PCR. RESULTS: ABM/P-15 enhanced proliferation of cultured PDL cells. It increased the mRNA expression of TGF-beta and BMP-2 in cultured PDL cells on days 3 and 7. IGF-I and b-FGF mRNA expressions showed a slight decrease, while PDGF expression was observed to have increased on day 3. VEGF and COL1 mRNA expressions were found not to be different on days 3 and 7. No differences were observed in the mineralization properties of cultured PDL cells treated with or without ABM/P-15. CONCLUSIONS: Based on the results of this in vitro study, it may be concluded that ABM/P-15 enhanced the regenerative capacity of PDL by regulating specific gene expressions of cells during early wound healing.


Subject(s)
Bone Substitutes/pharmacology , Collagen/pharmacology , Guided Tissue Regeneration, Periodontal/methods , Hydroxyapatites/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Peptide Fragments/pharmacology , Periodontal Ligament/metabolism , Animals , Bone Matrix , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Drug Combinations , Fibroblast Growth Factor 2/drug effects , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/analysis , Reference Values , Tissue Scaffolds , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factors/drug effects , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
20.
Clin Pharmacokinet ; 58(9): 1131-1147, 2019 09.
Article in English | MEDLINE | ID: mdl-31016670

ABSTRACT

Nintedanib is an oral, small-molecule tyrosine kinase inhibitor approved for the treatment of idiopathic pulmonary fibrosis and patients with advanced non-small cell cancer of adenocarcinoma tumour histology. Nintedanib competitively binds to the kinase domains of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF). Studies in healthy volunteers and in patients with advanced cancer have shown that nintedanib has time-independent pharmacokinetic characteristics. Maximum plasma concentrations of nintedanib are reached approximately 2-4 h after oral administration and thereafter decline at least bi-exponentially. Over the investigated dose range of 50-450 mg once daily and 150-300 mg twice daily, nintedanib exposure increases are dose proportional. Nintedanib is metabolised via hydrolytic ester cleavage, resulting in the formation of the free acid moiety that is subsequently glucuronidated and excreted in the faeces. Less than 1% of drug-related radioactivity is eliminated in urine. The terminal elimination half-life of nintedanib is about 10-15 h. Accumulation after repeated twice-daily dosing is negligible. Sex and renal function have no influence on nintedanib pharmacokinetics, while effects of ethnicity, low body weight, older age and smoking are within the inter-patient variability range of nintedanib exposure and no dose adjustments are required. Administration of nintedanib in patients with moderate or severe hepatic impairment is not recommended, and patients with mild hepatic impairment should be monitored closely and the dose adjusted accordingly. Nintedanib has a low potential for drug-drug interactions, especially with drugs metabolised by cytochrome P450 enzymes. Concomitant treatment with potent inhibitors or inducers of the P-glycoprotein transporter can affect the pharmacokinetics of nintedanib. At an investigated dose of 200 mg twice daily, nintedanib does not have proarrhythmic potential.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Aged , Animals , Carcinoma, Non-Small-Cell Lung/complications , Case-Control Studies , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Fibroblast Growth Factors/drug effects , Humans , Indoles/administration & dosage , Indoles/blood , Lung Neoplasms/pathology , Male , Middle Aged , Models, Animal , Platelet-Derived Growth Factor/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Rats , Vascular Endothelial Growth Factor A/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL