Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Inflammation ; 46(6): 2270-2275, 2023 Dec.
Article En | MEDLINE | ID: mdl-37486528

The osteogenic potential of mesenchymal stem cells (MSc) in axial spondyloarthritis (AxSpA) depends on the interplay of inflammation and multiple hormonal and local mechanical factors. In this study, MCs, derived from the adipose tissue of a healthy donor, were cultured under or without continuous mechanical load in the osteogenic differentiation medium with or without the addition of testosterone, cocktail of INF-γ/TNF-α/IL-22, or both. Real-time PCR for osteogenic transcription factors demonstrated that in the absence of INF-γ/TNF-α/IL-22, mechanical load causes significant upregulation of SPP1 (osteopontin), while the presence of the inflammatory cytokines almost completely abolishes this effect. In addition, exposure to INF-γ/TNF-α/IL-22 slightly upregulated BMP2, but suppressed the expression of ALPL, Col1A1, and SPP1, reinforcing the hypothesis that the inflammatory environment allows MSc to commit toward the IL-22-driven osteogenic differentiation but can restrict the later stages of osteogenesis. In summary, osteopontin can play a role in the pathogenesis of AxSpA, linking between mechanical load and pathological bone formation.


Axial Spondyloarthritis , Mesenchymal Stem Cells , Humans , Tumor Necrosis Factor-alpha/metabolism , Osteogenesis , Osteopontin/genetics , Osteopontin/metabolism , Osteopontin/pharmacology , Up-Regulation , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Cells, Cultured , Interleukin-22
2.
Cancers (Basel) ; 15(6)2023 Mar 17.
Article En | MEDLINE | ID: mdl-36980704

BACKGROUND: Osteosarcoma (OS) mortality is attributed to lung metastases. Endothelial progenitor cells (EPCs) mediate the angiogenic switch in several cancers. The spatial proximity between EPCs and OS in the bone led to the hypothesis that EPCs-osteosarcoma interactions may possibly promote OS progression and aggressiveness. METHODS: A PI3K inhibitor, Bevacizumab (an anti-VEGF-A antibody), and an anti-FGF2 antibody were added to the EPCs' conditioned medium (EPC-CM), and their impacts on OS cell (U2-OS and 143B) proliferation, migration, invasion, MMP9 expression, and AKT phosphorylation were determined. The autocrine role of VEGF-A was assessed using Bevacizumab treatment and VEGF-A silencing in OS cells. Toward this end, an orthotopic mouse OS model was established. Mouse and human tumors were immunolabeled with antibodies to the abovementioned factors. RESULTS: EPC-CM enhanced osteosarcoma MMP9 expression, invasiveness, and migration via the PI3K/AKT pathway. The addition of Bevacizumab and an anti-FGF2 antibody to the EPC-CM diminished OS cell migration. The autocrine role of VEGF-A was assessed using Bevacizumab and VEGF-A silencing in OS cells, resulting in decreased AKT phosphorylation and, consequently, diminished invasiveness and migration. Consistently, OS xenografts in mice displayed high VEGF-A and FGF2 levels. Remarkably, lung metastasis specimens derived from OS patients exhibited marked immunolabeling of CD31, VEGF-A, and FGF2. Conclusions: EPCs promote OS progression not only by physically incorporating into blood vessels, but also by secreting cytokines, which act via paracrine signaling. EPCs induced in vitro MMP9 overexpression, invasion, and migration. Additional animal studies are warranted to further expand these results. These findings may pave the way toward the development of novel EPCs-targeted therapeutics aimed at blocking OS metastasis.

3.
Front Pharmacol ; 13: 1002550, 2022.
Article En | MEDLINE | ID: mdl-36386129

Zoledronic acid (Zol) is a potent bisphosphonate that inhibits the differentiation of monocytes into osteoclasts. It is often used in combination with dexamethasone (Dex), a glucocorticoid that promotes the resolution of inflammation, to treat malignant diseases, such as multiple myeloma. This treatment can result in bone pathologies, namely medication related osteonecrosis of the jaw, with a poor understanding of the molecular mechanism on monocyte differentiation. IFN-ß is a pro-resolving cytokine well-known as an osteoclast differentiation inhibitor. Here, we explored whether Zol and/or Dex regulate macrophage osteoclastic differentiation via IFN-ß. RAW 264.7 and peritoneal macrophages were treated with Zol and/or Dex for 4-24 h, and IFN-ß secretion was examined by ELISA, while the IFN stimulated gene (ISG) 15 expression was evaluated by Western blotting. RANKL-induced osteoclastogenesis of RAW 264.7 cells was determined by TRAP staining following treatment with Zol+Dex or IFN-ß and anti-IFN-ß antibodies. We found only the combination of Zol and Dex increased IFN-ß secretion by RAW 264.7 macrophages at 4 h and, correspondingly, ISG15 expression in these cells at 24 h. Moreover, Zol+Dex blocked osteoclast differentiation to a similar extent as recombinant IFN-ß. Neutralizing anti-IFN-ß antibodies reversed the effect of Zol+Dex on ISG15 expression and partially recovered osteoclastic differentiation induced by each drug alone or in combination. Finally, we found Zol+Dex also induced IFN-ß expression in peritoneal resolution phase macrophages, suggesting these drugs might be used to enhance the resolution of acute inflammation. Altogether, our findings suggest Zol+Dex block the differentiation of osteoclasts through the expression of IFN-ß. Revealing the molecular pathway behind this regulation may lead to the development of IFN-ß-based therapy to inhibit osteoclastogenesis in multiple myeloma patients.

4.
Oral Dis ; 26(7): 1523-1531, 2020 Oct.
Article En | MEDLINE | ID: mdl-32400918

OBJECTIVES: In a previous rat model, MRONJ occurrence was 50%. Our aim was to investigate the potential of endothelial progenitor cells (EPCs) to improve fibroblasts function and prevent MRONJ. METHODS: Human gingival fibroblasts were cultured with EPC-conditioned media (EPC-CM); endothelial growth media (EGM-2) or DMEM followed by incubation with 10 µM zoledronic (ZOL) and dexamethasone (DEX). Cell proliferation and migration were assessed by XTT and scratch wound healing assays. In vivo, ten Lewis rats were treated weekly with ZOL and DEX for 11 weeks. After a week, EPCs or EGM-2 were injected to the gingiva around the molars. At 3 weeks, bilateral molars were extracted. After 8 weeks, wound healing was assessed, and serum VEGF and blood vessels were quantified. RESULTS: ZOL/DEX significantly reduced fibroblasts proliferation and wound healing. Treatment with EPC-CM before ZOL/DEX improved cell proliferation, and scratch healing (p = .007, p = .023). In vivo, local EPC injection before tooth extraction increased serum VEGF (p = .01) and soft tissue vascularization (p = .05). Normal healing was similar (80%) in EPCs and EGM-2 groups. CONCLUSION: EPC rescued fibroblasts from the cytotoxic effect of ZOL/DEX and elevated serum VEGF and vessel density that might reduce MRONJ occurrence to 20% compared to 50% in a similar model.


Bisphosphonate-Associated Osteonecrosis of the Jaw , Osteonecrosis , Animals , Bisphosphonate-Associated Osteonecrosis of the Jaw/prevention & control , Diphosphonates , Fibroblasts , Rats , Rats, Inbred Lew , Zoledronic Acid
5.
Cells ; 9(6)2020 05 26.
Article En | MEDLINE | ID: mdl-32466427

Vascularization is a prerequisite for bone formation. Endothelial progenitor cells (EPCs) stimulate bone formation by creating a vascular network. Moreover, EPCs secrete various bioactive molecules that may regulate bone formation. The aim of this research was to shed light on the pathways of EPCs in bone formation. In a subcutaneous nude mouse ectopic bone model, the transplantation of human EPCs onto ß-TCP scaffold increased angiogenesis (p < 0.001) and mineralization (p < 0.01), compared to human neonatal dermal fibroblasts (HNDF group) and a-cellular scaffold transplantation (ß-TCP group). Human EPCs were lining blood vessels lumen; however, the majority of the vessels originated from endogenous mouse endothelial cells at a higher level in the EPC group (p < 01). Ectopic mineralization was mostly found in the EPCs group, and can be attributed to the recruitment of endogenous mesenchymal cells ten days after transplantation (p < 0.0001). Stromal derived factor-1 gene was expressed at high levels in EPCs and controlled the migration of mesenchymal and endothelial cells towards EPC conditioned medium in vitro. Blocking SDF-1 receptors on both cells abolished cell migration. In conclusion, EPCs contribute to osteogenesis mainly by the secretion of SDF-1, that stimulates homing of endothelial and mesenchymal cells. This data may be used to accelerate bone formation in the future.


Chemokine CXCL12/metabolism , Endothelial Progenitor Cells/metabolism , Osteogenesis , Paracrine Communication , Receptors, CXCR4/metabolism , Signal Transduction , 5'-Nucleotidase/metabolism , Adult , Animals , Calcification, Physiologic/drug effects , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Endothelial Progenitor Cells/drug effects , Female , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Nude , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Paracrine Communication/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction/drug effects , Young Adult
6.
Sci Rep ; 9(1): 18896, 2019 12 11.
Article En | MEDLINE | ID: mdl-31827217

Medication-related osteonecrosis of the jaw (MRONJ) is a serious adverse effect of antiresorptive and antiangiogenic therapies. MRONJ is identified by chronic wounds in the oral mucosa associated with exposed necrotic bone. We hypothesized that zoledronic acid (ZOL) impairs keratinocyte and fibroblast function and reduces soft tissue vascularization; therefore, treating MRONJ with proangiogenic cells may benefit MRONJ patients. The effect of ZOL and dexamethasone (DEX) on gingival fibroblasts and keratinocytes was investigated. In-vitro, ZOL inhibited fibroblast and keratinocyte proliferation, delaying scratch healing. In-vivo, exposed bone was detected at tooth extraction sites, mainly in ZOL(+)/DEX(+) rats; and was associated with significantly decreased soft tissue vascularization, serum-VEGF, and tissue-VEGF. Local injection of early and late endothelial progenitor cells (EPCs) healed 13 of 14 MRONJ lesions compared with 2/7 lesions in the mesenchymal stem cells, and 2/6, in culture-medium group. The EPCs reduced necrotic bone area, increased serum and tissue VEGF levels. EPCs engraftment was minimal, suggesting their paracrine role in MRONJ healing. The EPC-conditioned medium improved scratch healing of keratinocytes and fibroblasts via VEGF pathway and elevated mRNA of VEGFA and collagen1A1. In conclusion, a novel MRONJ treatment with EPCs, increased vascularization and improved epithelial and fibroblast functions as well as cured the lesion.


Bisphosphonate-Associated Osteonecrosis of the Jaw/therapy , Cell Proliferation/drug effects , Cell Transplantation , Dexamethasone/pharmacology , Endothelial Progenitor Cells/transplantation , Zoledronic Acid/pharmacology , Animals , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Bone Density Conservation Agents/pharmacology , Diphosphonates/pharmacology , Disease Models, Animal , Fibroblasts/drug effects , Humans , Keratinocytes/drug effects , Rats , Wound Healing/drug effects
7.
Biomolecules ; 9(11)2019 11 08.
Article En | MEDLINE | ID: mdl-31717420

Clinical trials have demonstrated the safety and efficacy of autologous endothelial progenitor cell (EPC) therapy in various diseases. Since EPCs' functions are influenced by genetic, systemic and environmental factors, the therapeutic potential of each individual EPCs is unknown and may affect treatment outcome. Therefore, our aim was to compare EPCs function among healthy donors in order to predict blood vessel formation (angiogenesis) before autologous EPC transplantation. Human EPCs were isolated from the blood of ten volunteers. EPCs proliferation rate, chemoattractant ability, and CXCR4 mRNA levels were different among donors (p < 0.0001, p < 0.01, p < 0.001, respectively). A positive correlation was found between SDF-1, CXCR4, and EPCs proliferation (R = 0.736, p < 0.05 and R = 0.8, p < 0.01, respectively). In-vivo, blood vessels were counted ten days after EPCs transplantation in a subcutaneous mouse model. Mean vessel density was different among donors (p = 0.0001); nevertheless, donors with the lowest vessel densities were higher compared to control (p < 0.05). Finally, using a linear regression model, a mathematical equation was generated to predict blood vessel density relying on: (i) EPCs chemoattractivity, and (ii) VEGFR-2 mRNA levels. Results reveal differences in EPCs functions among healthy individuals, emphasizing the need for a potency assay to pave the way for standardized research and clinical use of human EPCs.


Endothelial Progenitor Cells/transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Blood Vessels/metabolism , Blood Vessels/transplantation , Cell Movement/genetics , Cell Proliferation/genetics , Chemotactic Factors/genetics , Endothelial Progenitor Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Mice , RNA, Messenger/genetics , Stem Cell Transplantation
8.
Clin Implant Dent Relat Res ; 21(4): 593-601, 2019 Aug.
Article En | MEDLINE | ID: mdl-31025823

BACKGROUND: Transforming growth factor-ß (TGF-ß1 ) enhances mesenchymal stem cell (MSC) differentiation into osteoblasts. PURPOSE: The aim of the study was to assess whether TGF-ß1 loaded onto ß-tricalcium phosphate (ß-TCP) synthetic scaffold enhances bone regeneration in a rat calvaria model. The release kinetics of TGF-ß1 from ß-TCP scaffold was evaluated in vitro. MATERIALS AND METHODS: TGF-ß1 in various concentrations (1-40 ng/mL) was loaded onto the ß-TCP scaffold, and release kinetics was monitored by ELISA. The effect of TGF-ß1 on the proliferation of MSCs was assessed using AlamarBlue, and MSC differentiation was evaluated by Alizarin Red quantification assay.Bone augmentation following transplantation of TGF-ß1 loaded onto ß-TCP in a rat calvaria model was evaluated in vivo. RESULTS: Greater TGF-ß1 release from the 40 ng/mL concentration was found. A suppressive effect of TGF-ß on the MSCs proliferation was observed with maximum inhibition obtained with 40 ng/mL compared to the control group (P = .028). A positive effect on MSCs osteogenic differentiation was found.Bone height and bone area fraction in vivo were similar with or without TGF-ß1 ; however, blood vessel density and degradation of the scaffold were significantly higher in the TGF-ß1 group. CONCLUSION: TGF-ß1 adsorbed to ß-TCP stimulated angiogenesis and scaffold degradation that may enhance bone formation.


Calcium Phosphates , Osteogenesis , Transforming Growth Factor beta , Animals , Bone Regeneration , Cell Differentiation , Rats , Skull , Tissue Engineering , Transforming Growth Factor beta1 , Transforming Growth Factors
9.
Thromb Haemost ; 100(5): 878-85, 2008 Nov.
Article En | MEDLINE | ID: mdl-18989533

Microvesicles (MVs) which include microparticles (MPs) and exosomes are found in blood circulation in normal physiologic conditions and are increased in a variety of diseases. This study evaluated the effects of MVs on human umbilical vein endothelial cells (HUVEC) by morphologic changes, apoptosis, and thrombogenicty, in vitro. Stimulation of monocyte cell line (THP-1) by starvation or by endotoxin and calcium ionophore A23187 resulted in the release of MVs which express exosome marker Tsg 101, negative phospholipids in their leaflets, monocyte markers (CD18, CD14) and active tissue factor (TF). MVs were found to disrupt EC integrity and rapidly induce membrane blebbing. Brief exposure (2-4 hours) to MVs resulted in EC membrane phospholipids "flip-flop" while longer stimulation (20 hours) led to two contradicting outcomes - tube formation as well as apoptosis, as assessed by nuclear fragmentation. Additionally, MVs exposure resulted in increased cell surface thrombogenicity and perturbation of the endothelial haemostatic balance, which were enhanced during longer exposure time. Activity, antigen level and mRNA expression of the coagulation initiator TF were elevated due to (i) adherence of MVs derived TF to the EC membrane, and (ii) an increase in endothelial TF expression. Furthermore, levels of the anticoagulant tissue factor pathway inhibitor (TFPI) and thrombomodulin (TM) were decreased. These findings demonstrate that monocyte MVs increase endothelial thrombogenicity and apoptosis. In addition, they induce tube formation which may indicate their angiogenic effect. These findings may clarify, in part, the role of MVs in EC dysfunction associated with inflammatory diseases and hypercoagulable states.


Apoptosis , Blood Coagulation , Cell-Derived Microparticles/metabolism , Endothelial Cells/metabolism , Exosomes/metabolism , Monocytes/metabolism , Blood Coagulation/genetics , Cell Differentiation , Cell Shape , Cells, Cultured , Endothelial Cells/pathology , Humans , Lipoproteins/metabolism , Neovascularization, Physiologic , RNA, Messenger/metabolism , Thrombomodulin/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism , Time Factors
10.
Plant Physiol ; 131(4): 1868-76, 2003 Apr.
Article En | MEDLINE | ID: mdl-12692346

The aroma of roses (Rosa hybrida) is due to more than 400 volatile compounds including terpenes, esters, and phenolic derivatives. 2-Phenylethyl acetate, cis-3-hexenyl acetate, geranyl acetate, and citronellyl acetate were identified as the main volatile esters emitted by the flowers of the scented rose var. "Fragrant Cloud." Cell-free extracts of petals acetylated several alcohols, utilizing acetyl-coenzyme A, to produce the corresponding acetate esters. Screening for genes similar to known plant alcohol acetyltransferases in a rose expressed sequence tag database yielded a cDNA (RhAAT1) encoding a protein with high similarity to several members of the BAHD family of acyltransferases. This cDNA was functionally expressed in Escherichia coli, and its gene product displayed acetyl-coenzyme A:geraniol acetyltransferase enzymatic activity in vitro. The RhAAT1 protein accepted other alcohols such as citronellol and 1-octanol as substrates, but 2-phenylethyl alcohol and cis-3-hexen-1-ol were poor substrates, suggesting that additional acetyltransferases are present in rose petals. The RhAAT1 protein is a polypeptide of 458 amino acids, with a calculated molecular mass of 51.8 kD, pI of 5.45, and is active as a monomer. The RhAAT1 gene was expressed exclusively in floral tissue with maximum transcript levels occurring at stage 4 of flower development, where scent emission is at its peak.


Acetyltransferases/metabolism , Esters/metabolism , Monoterpenes/metabolism , Oils, Volatile/metabolism , Rosa/enzymology , Rosa/growth & development , Terpenes/metabolism , Acetyltransferases/genetics , Acetyltransferases/isolation & purification , Acyclic Monoterpenes , Amino Acid Sequence , Flowers/enzymology , Flowers/genetics , Flowers/growth & development , Flowers/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Kinetics , Molecular Sequence Data , Molecular Structure , Odorants , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rosa/genetics , Rosa/metabolism , Sequence Homology , Substrate Specificity
...