Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
Int J Mol Sci ; 24(5)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36902186

ABSTRACT

There is an increasing urgency in the search for new drugs to target high-grade cancers such as osteosarcomas (OS), as these have limited therapeutic options and poor prognostic outlook. Even though key molecular events leading to tumorigenesis are not well understood, it is widely agreed that OS tumours are Wnt-driven. ETC-159, a PORCN inhibitor that inhibits the extracellular secretion of Wnt, has recently progressed on to clinical trials. In vitro and in vivo murine and chick chorioallantoic membrane xenograft models were established to examine the effect of ETC-159 on OS. Consistent with our hypothesis, we noted that ETC-159 treatment not only resulted in markedly decreased ß-catenin staining in xenografts, but also increased tumour necrosis and a significant reduction in vascularity-a hereby yet undescribed phenotype following ETC-159 treatment. Through further understanding the mechanism of this new window of vulnerability, therapies can be developed to potentiate and maximize the effectiveness of ETC-159, further increasing its clinical utility for the treatment of OS.


Subject(s)
Acyltransferases , Bone Neoplasms , Neovascularization, Pathologic , Osteosarcoma , Wnt Signaling Pathway , Animals , Humans , Mice , Acyltransferases/antagonists & inhibitors , beta Catenin/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/drug therapy , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Membrane Proteins/antagonists & inhibitors , Necrosis , Osteosarcoma/blood supply , Osteosarcoma/drug therapy , Wnt Signaling Pathway/drug effects , Neovascularization, Pathologic/drug therapy
2.
J Zhejiang Univ Sci B ; 22(11): 885-892, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34783219

ABSTRACT

Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. It is an aggressive tumor with a tendency to spread to the lung, which is the most common site of metastasis. Patients with advanced OS with metastases have poor prognoses despite the application of chemotherapy, thus highlighting the need for novel therapeutic targets. The tumor microenvironment (TME) of OS is confirmed to be essential for and supportive of tumor growth and dissemination. The immune component of the OS microenvironment is mainly composed of tumor-associated macrophages (TAMs). In OS, TAMs promote tumor growth and angiogenesis and upregulate the cancer stem cell-like phenotype. However, TAMs inhibit the metastasis of OS. Therefore, much attention has been paid to investigating the mechanism of TAMs in OS development and the progression of immunotherapy for OS. In this article, we aim to summarize the roles of TAMs in OS and the major findings on the application of TAMs in OS treatment.


Subject(s)
Bone Neoplasms/immunology , Osteosarcoma/immunology , Tumor-Associated Macrophages/physiology , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Humans , Neoplasm Metastasis , Neoplastic Stem Cells/physiology , Neovascularization, Pathologic/etiology , Osteosarcoma/blood supply , Osteosarcoma/pathology , Tumor Microenvironment/physiology
3.
Life Sci ; 256: 118011, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32592723

ABSTRACT

Melatonin is recognized as an anti-angiogenic agent, but its function in the tumor microenvironment especially in osteosarcoma remains uncertain. Among the selected miRNAs, miR-205, miR-424, miR-140, miR-106, and miR-519 were upregulated by melatonin in osteosarcoma cells. The functional role of miR-424-5p in osteosarcoma was further analyzed using miR-424-5p mimic/inhibitor. VEGFA mRNA and protein expression were altered by miR-424-5p mimic/inhibitor transfection with and without melatonin treatment and it was further identified that the VEGFA 3'UTR is directly targeted by miR-424-5p using the luciferase reporter gene system. The conditioned medium from SaOS2 and MG63 cells treated with melatonin and/or transfected with miR-424-5p mimic/inhibitor was exposed to endothelial cells, and cell proliferation and migration was analyzed. MG-63 and SaOS2 cells are also transfected with miR-424-5p inhibitors and positioned on CAM vascular bed to study the angiogenic activity at both morphological and molecular level under melatonin treatment. Our observations demonstrate for the first time that, melatonin upregulated the expression of miR-424-5p in osteosarcoma inhibiting VEGFA. Furthermore, it suppresses tumor angiogenesis, modulating surrounding endothelial cell proliferation and migration as well as the morphology of blood vessels, and angiogenic growth factors. These findings suggest that melatonin could play a pivotal role in tumor suppression via miR-424-5p/VEGFA axis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bone Neoplasms/drug therapy , Melatonin/pharmacology , Osteosarcoma/drug therapy , Animals , Bone Neoplasms/blood supply , Cell Line, Tumor , Chickens , Egg Yolk , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , MicroRNAs/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , RNA, Messenger/genetics , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Am J Case Rep ; 21: e922257, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32439833

ABSTRACT

BACKGROUND Osteosarcoma is the most common type of malignant bone tumor arising from mesenchymal stem cell. When occurring on the proximal humerus, it is associated with poor outcomes; there are numerous neurovascular structures around proximal humerus. Unfortunately, the degree of vascular involvement in osteosarcoma is rarely evaluated and reported. Thus, we would like to highlight our case. CASE REPORT We reported a case of left proximal humerus osteosarcoma causing dead limb in a 14-year-old boy. The dead limb progressed in the span of 3 weeks. An emergency forequarter amputation (FQA) was conducted to prevent further complications such as septicemia and mortality. Two months after the surgery, the patient had improved quality of life. One year after, the patient had no local recurrence. However, there was a lung metastasis detected 9 months after the surgery. The patient died 13 months after the surgery. CONCLUSIONS Osteosarcoma of the proximal humerus can potentially compromise vascular structures. Early diagnosis and treatment are mandatory to prevent such complications.


Subject(s)
Amputation, Surgical , Bone Neoplasms/blood supply , Bone Neoplasms/surgery , Humerus/blood supply , Humerus/surgery , Osteosarcoma/blood supply , Osteosarcoma/surgery , Adolescent , Fatal Outcome , Humans , Lung Neoplasms/secondary , Male , Quality of Life
5.
Ultrasound Med Biol ; 45(11): 3028-3041, 2019 11.
Article in English | MEDLINE | ID: mdl-31474384

ABSTRACT

Ultrasound and microbubbles have been found to improve the delivery of drugs and nanoparticles to tumor tissue. To obtain new knowledge on the influence of vascular parameters on extravasation and to elucidate the effect of acoustic pressure on extravasation and penetration of nanoscale particles into the extracellular matrix, real-time intravital multiphoton microscopy was performed during sonication of tumors growing in dorsal window chambers. The impact of vessel diameter, vessel structure and blood flow was characterized. Fluorescein isothiocyanate-dextran (2 MDa) was injected to visualize blood vessels. Mechanical indexes (MI) of 0.2-0.8 and in-house-made, nanoparticle-stabilized microbubbles or Sonovue were applied. The rate and extent of penetration into the extracellular matrix increased with increasing MI. However, to achieve extravasation, smaller vessels required MIs (0.8) higher than those of blood vessels with larger diameters. Ultrasound changed the blood flow rate and direction. Interestingly, the majority of extravasations occurred at vessel branching points.


Subject(s)
Extravasation of Diagnostic and Therapeutic Materials , Nanoparticles/chemistry , Osteosarcoma/blood supply , Osteosarcoma/diagnostic imaging , Sonication , Ultrasonography/methods , Animals , Cell Line, Tumor , Contrast Media/chemistry , Dextrans , Disease Models, Animal , Fluorescein-5-isothiocyanate/analogs & derivatives , Humans , Male , Mice , Mice, Inbred BALB C , Microbubbles , Phospholipids/chemistry , Sulfur Hexafluoride/chemistry
6.
Int J Oncol ; 55(1): 167-178, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31180533

ABSTRACT

Vascular endothelial growth inhibitor (VEGI; also referred to as TNFSF15 or TL1A) is involved in the modulation of vascular homeostasis. VEGI is known to operate via two receptors: Death receptor­3 (DR3) and decoy receptor­3 (DcR3). DR3, which is thus far the only known functional receptor for VEGI, contains a death domain and induces cell apoptosis. DcR3 is secreted as a soluble protein and antagonizes VEGI/DR3 interaction. Overexpression of DcR3 and downregulation of VEGI have been detected in a number of cancers. The aim of the present study was to investigate the effects of sodium valproate (VPA), a histone deacetylase inhibitor, in combination with hydralazine hydrochloride (Hy), a DNA methylation inhibitor, on the expression of VEGI and its related receptors in human osteosarcoma (OS) cell lines and human microvascular endothelial (HMVE) cells. Combination treatment with Hy and VPA synergistically induced the expression of VEGI and DR3 in both OS and HMVE cells, without inducing DcR3 secretion. In addition, it was observed that the combination of VPA and Hy significantly enhanced the inhibitory effect on vascular tube formation by VEGI/DR3 autocrine and paracrine pathways. Furthermore, the VEGI/VEGF­A immune complex was pulled down by immunoprecipitation. Taken together, these findings suggest that DNA methyltransferase and histone deacetylase inhibitors not only have the potential to induce the re­expression of tumor suppressor genes in cancer cells, but also exert anti­angiogenic effects, via enhancement of the VEGI/DR3 pathway and VEGI/VEGF­A interference.


Subject(s)
Bone Neoplasms/drug therapy , Hydralazine/pharmacology , Osteosarcoma/drug therapy , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Valproic Acid/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Drug Synergism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Member 25/biosynthesis , Receptors, Tumor Necrosis Factor, Member 25/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
7.
J Cell Biochem ; 120(8): 12473-12488, 2019 08.
Article in English | MEDLINE | ID: mdl-30825232

ABSTRACT

Osteosarcoma (OS) is the most common highly malignant bone tumor in teens. Vasculogenic mimicry (VM) is defined as de novo extracellular matrix-rich vascular-like networks formed by highly aggressive tumor cells. We previously reported the presence of VM and it is an unfavorable prognostic factor in OS patients. Long noncoding RNAs (lncRNAs) are aberrantly expressed in OS and involved in cancer cell VM. However, lncRNAs in VM formation of OS have not been investigated. We, therefore, profiled the expression of lncRNAs in highly aggressive OS cell line 143B compared with its parental poorly aggressive cell line HOS. The differentially expressed (DE) lncRNAs and messenger RNA (mRNAs) were subjected to constructed lncRNA-mRNA coexpressed network. The top-ranked hub gene lncRNA n340532 knockdown 143B cells were used for in vitro and in vivo VM assays. The annotation of DE lncRNAs was performed according to the coexpressed mRNAs by Gene Ontology and pathway analysis. A total of 1360 DE lncRNAs and 1353 DE mRNAs were screened out. lncRNA MALAT1 and FTX, which have known functions related to VM formation and tumorigenesis were identified in our data. The coexpression network composed of 226 lncRNAs and 118 mRNAs in which lncRNA n340532 had the highest degree number. lncRNA n340532 knockdown reduced VM formation in vitro. The suppression of n340532 also exhibited potent anti-VM and antimetastasis effect in vivo, suggesting its potential role in OS VM and metastasis. Furthermore, n340532 coexpressed with 10 upregulation mRNAs and 3 downregulation mRNAs. The enriched transforming growth factor-ß signaling pathway, angiogenesis and so forth were targeted by those coexpressed mRNAs, implying n340532 may facilitate VM formation in OS through these pathways and gene functions. Our findings provide evidence for the potential role of lncRNAs in VM formation of OS that could be used in the clinic for anti-VM therapy in OS.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/blood supply , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , RNA, Long Noncoding/genetics , Animals , Apoptosis , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Proliferation , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Res Vet Sci ; 122: 1-6, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30439557

ABSTRACT

Combretastatin A-4 phosphate (CA4P) induces tumor necrosis by selectively inhibiting tumor blood flow. However, the detailed mechanisms by which CA4P selectively disrupts tumor blood vessels are not well understood. Our previous study indicated that the selective blocking effect of CA4P might be related to a vascular endothelial cadherin (VE-cadherin) dysfunction in the tumor vasculature. In this study, we evaluated the vascular disrupting effect of CA4P on canine osteosarcomas xenografted into mice, focusing on VE-cadherin. Even though 30 mg/kg CA4P only partially inhibited blood flow in the xenografted tumor, a combination of an anti-VE-cadherin neutralizing antibody and 30 mg/kg CA4P inhibited most of the tumor blood flow. In addition, the combination of antibody and drug significantly inhibited tumor growth compared to the control. These results strongly suggested a relationship between the expression of VE-cadherin in tumor blood vessels and the selective blocking mechanisms of CA4P.


Subject(s)
Antigens, CD/metabolism , Bibenzyls/pharmacology , Cadherins/metabolism , Neoplasms, Experimental/drug therapy , Osteosarcoma/blood supply , Osteosarcoma/drug therapy , Animals , Antigens, CD/genetics , Cadherins/genetics , Dog Diseases/metabolism , Dogs , Endothelium, Vascular/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Mice , Neovascularization, Pathologic/pathology
9.
Eur J Radiol ; 105: 49-55, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30017298

ABSTRACT

PURPOSE: To evaluate the frequency of tumor thrombus in the large veins draining primary pelvic osteosarcoma on early cross-sectional imaging studies and its effect on patient survival. MATERIALS AND METHODS: Our retrospective study included all patients with primary pelvic osteosarcoma treated at our facility between January 2000 and May 2014, who were ≤ 45 years of age, and had adequate imaging studies and clinical follow up. Four radiologists evaluated for tumor in the large draining veins on initial CT, MRI and PET/CTs. A consensus evaluation by the four radiologists together with findings on operative reports, pathology reports or follow-up imaging was used as the reference standard. RESULTS: Thirty-nine patients with primary pelvic osteosarcoma met final inclusion criteria. Tumor thrombus was identified in the large draining veins in 10 of the 22 (45%) patients who underwent tumor resection and 10 of the 17 (59%) who did not. In the 22 patients who underwent tumor resection, tumor thrombus was significantly associated with worse overall survival (p = 0.03). CONCLUSIONS: Tumor thrombus in the large draining veins is identified in a significant proportion of initial imaging studies in patients with pelvic osteosarcoma, and is associated with worse overall survival in patients who undergo tumor resection.


Subject(s)
Bone Neoplasms/blood supply , Osteosarcoma/blood supply , Pelvic Bones , Thrombophlebitis/pathology , Adolescent , Adult , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Child , Cross-Sectional Studies , Female , Humans , Magnetic Resonance Angiography/methods , Magnetic Resonance Angiography/mortality , Male , Middle Aged , Multimodal Imaging/methods , Multimodal Imaging/mortality , Osteosarcoma/mortality , Osteosarcoma/pathology , Positron Emission Tomography Computed Tomography/methods , Positron Emission Tomography Computed Tomography/mortality , Retrospective Studies , Survival Analysis , Thrombophlebitis/mortality , Tomography, X-Ray Computed/methods , Tomography, X-Ray Computed/mortality , Veins , Young Adult
10.
BMC Cancer ; 18(1): 536, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29739381

ABSTRACT

BACKGROUND: Osteosarcoma is the most common primary bone cancer in children and young adults. It is highly aggressive and patients that present with metastasis have a poor prognosis. Angiopoietin-like 4 (ANGPTL4) drives the progression and metastasis of many solid tumours, but has not been described in osteosarcoma tissue. ANGPTL4 also enhances osteoclast activity, which is required for osteosarcoma growth in bone. We therefore investigated the expression and function of ANGPTL4 in human osteosarcoma tissue and cell lines. METHODS: Expression of ANGPTL4 in osteosarcoma tissue microarrays was determined by immunohistochemistry. Hypoxic secretion of ANGPTL4 was tested by ELISA and Western blot. Regulation of ANGPTL4 by hypoxia-inducible factor (HIF) was investigated using isoform specific HIF siRNA (HIF-1α, HIF-2α). Effects of ANGPTL4 on cell proliferation, migration (scratch wound assay), colony formation and osteoblastogenesis were assessed using exogenous ANGPTL4 or cells stably transfected with ANGPTL4. Osteoclastogenic differentiation of CD14+ monocytes was assessed by staining for tartrate-resistant acid phosphatase (TRAP), bone resorption was assessed by lacunar resorption of dentine. RESULTS: ANGPTL4 was immunohistochemically detectable in 76/109 cases. ANGPTL4 was induced by hypoxia in 6 osteosarcoma cell lines, under the control of the HIF-1α transcription factor. MG-63 cells transfected with an ANGPTL4 over-expression plasmid exhibited increased proliferation and migration capacity and promoted osteoclastogenesis and osteoclast-mediated bone resorption. Individually the full-length form of ANGPTL4 could increase MG-63 cell proliferation, whereas N-terminal ANGPTL4 mediated the other pro-tumourigenic phenotypes. CONCLUSIONS: This study describes a role(s) for ANGPTL4 in osteosarcoma and identifies ANGPTL4 as a treatment target that could potentially reduce tumour progression, inhibit angiogenesis, reduce bone destruction and prevent metastatic events.


Subject(s)
Angiopoietin-Like Protein 4/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Osteogenesis/genetics , Osteosarcoma/pathology , Angiopoietin-Like Protein 4/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Resorption/genetics , Bone Resorption/pathology , Carcinogenesis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Osteoclasts/physiology , Osteosarcoma/blood supply , Osteosarcoma/genetics , RNA, Small Interfering/metabolism , Tissue Array Analysis
11.
Sci Rep ; 8(1): 5415, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29615744

ABSTRACT

There is increasing evidence that the complement system is activated in various cancer tissues. Besides being involved in innate immunity against pathogens, the complement system also participates in inflammation and the modulation of tumor microenvironment. Recent studies suggest that complement activation promotes tumor progression in various ways. Among some cancer cell lines, we found that human bone osteosarcoma epithelial cells (U2-OS) can activate the alternative pathway of the complement system by pooled normal human serum. Interestingly, U2-OS cells showed less expression of complement regulatory proteins, compared to other cancer cell lines. Furthermore, the activated complement system enhanced the production of growth factors, which promoted angiogenesis of human endothelial cells. Our results demonstrated a direct linkage between the complement system and angiogenesis using the in vitro model, which suggest the complement system and related mechanisms might be potential targets for cancer treatment.


Subject(s)
Bone Neoplasms/pathology , Complement System Proteins/metabolism , Fibroblast Growth Factor 1/metabolism , Neovascularization, Pathologic/metabolism , Osteosarcoma/pathology , Vascular Endothelial Growth Factor A/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Cell Line, Tumor , Endothelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Phosphorylation
12.
Anticancer Res ; 38(1): 159-164, 2018 01.
Article in English | MEDLINE | ID: mdl-29277768

ABSTRACT

BACKGROUND: We previously developed a color-coded imaging model that can quantify the length of nascent blood vessels using Gelfoam® implanted in nestin-driven green fluorescent protein (ND-GFP) nude mice. In this model, nascent blood vessels selectively express GFP. We also previously showed that osteosarcoma cells promote angiogenesis in this assay. We have also previously demonstrated the tumor-targeting bacteria Salmonella typhimurium A1-R (S. typhimurium A1-R) can inhibit or regress all tested tumor types in mouse models. The aim of the present study was to determine if S. typhimurium A1-R could inhibit osteosarcoma angiogenesis in the in vivo Gelfoam® color-coded imaging assay. MATERIALS AND METHODS: Gelfoam® was implanted subcutaneously in ND-GFP nude mice. Skin flaps were made 7 days after implantation and 143B-RFP human osteosarcoma cells expressing red fluorescent protein (RFP) were injected into the implanted Gelfoam. After establishment of tumors in the Gelfoam®, control-group mice were treated with phosphate buffered saline via tail-vein injection (iv) and the experimental group was treated with S. typhimurium A1-R iv Skin flaps were made at day 7, 14, 21, and 28 after implantation of the Gelfoam® to allow imaging of vascularization in the Gelfoam® using a variable-magnification small-animal imaging system and confocal fluorescence microscopy. RESULTS: Nascent blood vessels expressing ND-GFP extended into the Gelfoam® over time in both groups. However, the extent of nascent blood-vessel growth was significantly inhibited by S. typhimurium A1-R treatment by day 28. CONCLUSION: The present results indicate S. typhimurium A1-R has potential for anti-angiogenic targeted therapy of osteosarcoma.


Subject(s)
Bone Neoplasms/therapy , Neovascularization, Pathologic/therapy , Osteosarcoma/therapy , Salmonella typhimurium , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice, Nude , Mice, Transgenic , Osteosarcoma/blood supply , Osteosarcoma/pathology
13.
Biomaterials ; 149: 29-40, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28988062

ABSTRACT

Cancer stem cells (CSCs) are believed to have a critical role in tumorigenesis, metastasis, therapeutic resistance or recurrence. Therefore, strategies designed to specifically target and eliminate CSCs have become one of the most promising and desirable ways for tumor treatment. Osteosarcoma stem cells (OSCs), the CSCs in osteosarcoma (OS), are critically associated with OS progression. Here, we show that single-walled carbon nanotubes (SWCNTs), including unmodified SWCNT (SWCNT-Raw) and SWCNT-COOH, have the ability to specifically inhibit the process of TGFß1-induced OS cells dedifferentiation, prevent the stem cell phenotypes acquisition in OS cells and reduce the OSC viability under conditions which mimic the OS microenvironment. Concurrently, SWCNT treatment significantly down-regulates the expression of OSC markers in OS, and markedly reduces the tumor microvessel density and tumor growth. Furthermore, we found that SWCNT could suppress the TGFß1-induced activation of TGFß type I receptor and downstream signaling, which are key for the OSC formation and maintenance. Our results reveal an unexpected function of SWCNT in negative modulation of OSCs, and provide significant implications for the potential CSCs-targeted therapeutic applications of SWCNT.


Subject(s)
Bone Neoplasms/drug therapy , Nanotubes, Carbon/chemistry , Neoplastic Stem Cells/drug effects , Osteosarcoma/drug therapy , Transforming Growth Factor beta1/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Dedifferentiation , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Humans , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Osteosarcoma/pathology , Particle Size , Signal Transduction , Tumor Microenvironment
14.
Exp Cell Res ; 360(2): 138-145, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28867479

ABSTRACT

Angiogenesis is now well known for being involved in tumor progression, aggressiveness, emergence of metastases, and also resistance to cancer therapies. In this study, to better mimic tumor angiogenesis encountered in vivo, we used 3D culture of osteosarcoma cells (MG-63) that we deposited on 2D endothelial cells (HUVEC) grown in monolayer. We report that endothelial cells combined with tumor cells were able to form a well-organized network, and that tubule-like structures corresponding to new vessels infiltrate tumor spheroids. These vessels presented a lumen and expressed specific markers as CD31 and collagen IV. The combination of 2D endothelial cells and 3D microtissues of tumor cells also increased expression of angiogenic factors as VEGF, CXCR4 and ICAM1. The cell environment is the key point to develop tumor vascularization in vitro and to be closer to tumor encountered in vivo.


Subject(s)
Bone Neoplasms/pathology , Cell Culture Techniques/methods , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/physiology , Neovascularization, Pathologic/pathology , Osteosarcoma/pathology , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Neovascularization, Pathologic/genetics , Osteosarcoma/blood supply , Osteosarcoma/genetics , Tissue Scaffolds/chemistry
15.
Mar Drugs ; 15(6)2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28632184

ABSTRACT

The marine origin polysaccharide fucoidan combines multiple biological activities. As demonstrated by various studies in vitro and in vivo, fucoidans show anti-viral, anti-tumor, anti-oxidant, anti-inflammatory and anti-coagulant properties, although the detailed molecular action remains to be elucidated. The aim of the present study is to assess the impact of crude fucoidan extracts, on the formation of vascular structures in co-culture models relevant for bone vascularization during bone repair and for vascularization processes in osteosarcoma. The co-cultures consisted of bone marrow derived mesenchymal stem cells, respectively the osteosarcoma cell line MG63, and human blood derived outgrowth endothelial cells (OEC). The concentration dependent effects on the metabolic activity on endothelial cells and osteoblast cells were first assessed using monocultures of OEC, MSC and MG63 suggesting a concentration of 100 µg/mL as a suitable concentration for further experiments. In co-cultures fucoidan significantly reduced angiogenesis in MSC/OEC but also in MG63/OEC co-cultures suggesting a potential application of fucoidan to lower the vascularization in bone tumors such as osteosarcoma. This was associated with a decrease in VEGF (vascular endothelial growth factor) and SDF-1 (stromal derived factor-1) on the protein level, both related to the control of angiogenesis and furthermore discussed as crucial factors in osteosarcoma progression and metastasis. In terms of bone formation, fucoidan slightly lowered on the calcification process in MSC monocultures and MSC/OEC co-cultures. In summary, these data suggest the suitability of lower fucoidan doses to limit angiogenesis for instance in osteosarcoma.


Subject(s)
Bone Neoplasms/blood supply , Bone Regeneration/drug effects , Chemokine CXCL12/analysis , Neovascularization, Pathologic/prevention & control , Osteosarcoma/blood supply , Polysaccharides/pharmacology , Vascular Endothelial Growth Factor A/analysis , Bone Neoplasms/drug therapy , Cells, Cultured , DNA/analysis , Endothelial Cells/physiology , Humans , Osteosarcoma/drug therapy
16.
Cell Death Dis ; 8(4): e2750, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28406476

ABSTRACT

In recent years, much research has focused on the role of angiogenesis in osteosarcoma, which occurs predominantly in adolescents and young adults. The vascular endothelial growth factor-A (VEGF-A) pathway is the key regulator of angiogenesis and in osteosarcoma. VEGF-A expression has been recognized as a prognostic marker in angiogenesis. Aberrant WNT1-inducible signaling pathway protein-1 (WISP-1) expression is associated with various cancers. However, the function of WISP-1 in osteosarcoma angiogenesis is poorly understood. We demonstrate a positive correlation between WISP-1 and VEGF-A expression in human osteosarcoma. Moreover, we show that WISP-1 promotes VEGF-A expression in human osteosarcoma cells, subsequently inducing human endothelial progenitor cell (EPC) migration and tube formation. The focal adhesion kinase (FAK), Jun amino-terminal kinase (JNK), and hypoxia-inducible factor (HIF)-1α signaling pathways were activated after WISP-1 stimulation, while FAK, JNK, and HIF-1α inhibitors or small interfering RNA (siRNA) abolished WISP-1-induced VEGF-A expression and angiogenesis. In vitro and in vivo studies revealed down-regulation of microRNA-381 (miR-381) in WISP-1-induced VEGF-A expression and angiogenesis. Our findings reveal that WISP-1 enhances VEGF-A expression and angiogenesis through the FAK/JNK/HIF-1α signaling pathways, as well as via down-regulation of miR-381 expression. WISP-1 may be a promising target in osteosarcoma angiogenesis.


Subject(s)
Bone Neoplasms , CCN Intercellular Signaling Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic , Osteosarcoma , Proto-Oncogene Proteins/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Adolescent , Adult , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Chick Embryo , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Male , MicroRNAs/biosynthesis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Neoplasm/biosynthesis , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology
17.
Anticancer Drugs ; 28(5): 514-521, 2017 06.
Article in English | MEDLINE | ID: mdl-28225457

ABSTRACT

GTPase RhoA and its downstream Rho-associated coiled-coil-containing protein kinases (ROCKs) are frequently overexpressed in human cancers. Inhibition of the RhoA/ROCK pathway blocks angiogenesis mediated by the vascular endothelial growth factor, which led us to investigate the role of this pathway in vasculogenic mimicry (VM) - a process by which aggressive cancer cells form vessel-like structures that provide adequate blood supply for tumor growth. We showed that the expression of RhoA and its effector kinases ROCK1/2 was much higher in human osteosarcoma (OS) tissues and the human OS cell line U2OS than in nontumorous tissues and cell line hFOB 1.19 using western blot analysis and real-time PCR. Inhibition of the RhoA/ROCK signaling pathway by the pharmacological inhibitor fasudil reduced vascular-like channels of U2OS cells in Matrigel. Furthermore, we used rhodamine-phalloidin immunofluorescence, wound healing assay, and transwell migration assay to examine the effect of fasudil on tumor cell plasticity and motility, both of which play key roles in VM formation. Finally, we explored the underlying mechanisms of fasudil-induced VM destruction. In this context, we showed that the RhoA/ROCK signaling pathway is a novel regulator in VM of U2OS OS cells and suggest that fasudil in conjunction with established treatments may present a novel therapeutic strategy for OS.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Plasticity/drug effects , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/pathology , Fluorescent Antibody Technique , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , rho-Associated Kinases/biosynthesis , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/biosynthesis , rhoA GTP-Binding Protein/metabolism
18.
Biochem Biophys Res Commun ; 482(4): 987-993, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27908732

ABSTRACT

Osteosarcoma (OS) has become one of the most common primary malignant tumors in the children and adolescents with a poor prognosis owing to its high malignant and metastatic potential. Although increasing evidence indicates that miR-451 could inhibit the growth and metastasis of OS, its effect on angiogenesis in OS is still very poor. What is more, the mechanism by which miR-451 affects the OS has not been fully elucidated. In the present study, miR-451 was reduced in human osteosarcoma tissues compared with the adjacent bone tissues, and the introduction of miR-451 dramatically inhibited the growth, migration and angiogenesis in OS. Additionally, it was suggested that IL 6R is a direct target gene of miR-451. Silencing of IL 6R suppressed the growth, migration and angiogenesis of OS, which was consistent with the effect of overexpression of miR-451. In conclusion, our data demonstrate that miR-451 may function as a potential suppressor of tumor growth, migration and angiogenesis in OS via down-regulating IL 6R, suggesting a promising therapeutic avenue for managing OS.


Subject(s)
Bone Neoplasms/genetics , Bone and Bones/pathology , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Osteosarcoma/genetics , Receptors, Interleukin-6/genetics , Animals , Base Sequence , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Bone and Bones/blood supply , Bone and Bones/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , Osteosarcoma/pathology
19.
Oncotarget ; 7(34): 54474-54487, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27323409

ABSTRACT

The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility and angiogenesis by interacting with the formyl peptide receptor type 1 (FPR1). In this study, we present evidence that the cyclization of the uPAR88-92 sequence generates a new potent inhibitor of migration, and extracellular matrix invasion of human osteosarcoma and chondrosarcoma cells expressing comparable levels of FPR1 on cell surface. In vitro, the cyclized peptide [SRSRY] prevents formation of capillary-like tubes by endothelial cells co-cultured with chondrosarcoma cells and trans-endothelial migration of osteosarcoma and chondrosarcoma cells. When chondrosarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density and circulating tumor cells in blood samples collected before the sacrifice, were significantly reduced in animals treated daily with i.p-administration of 6 mg/Kg [SRSRY] as compared to animals treated with vehicle only. Our findings indicate that [SRSRY] prevents three key events occurring during the metastatic process of osteosarcoma and chondrosarcoma cells: the extracellular matrix invasion, the formation of a capillary network and the entry into bloodstream.


Subject(s)
Bone Neoplasms/blood supply , Chondrosarcoma/blood supply , Neovascularization, Pathologic/drug therapy , Osteosarcoma/blood supply , Peptides, Cyclic/therapeutic use , Receptors, Urokinase Plasminogen Activator/therapeutic use , Animals , Cell Line, Tumor , Cell Movement , Chondrosarcoma/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Osteosarcoma/pathology , Receptors, Formyl Peptide/physiology
20.
Int J Exp Pathol ; 97(1): 86-92, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27028305

ABSTRACT

Periostin (PN), originally named as osteoblast-specific factor-2 (OSF-2), has been involved in regulating adhesion and differentiation of osteoblasts. Recently many studies have shown that high-level expression of PN is correlated significantly with tumour angiogenesis and prognosis in many kinds of human cancer. However, whether and how periostin expression influences prognosis in osteosarcoma remains unknown. This study aimed to examine the expression of PN in patients with osteosarcoma and explore the relationship of PN expression with clinicopathologic factors, tumour angiogenesis and prognosis. Immunohistochemistry was performed to determine the expression of PN in osteosarcoma and osteochondroma respectively. Vascular endothelial growth factor (VEGF) and CD34 were also examined in tissues from the osteosarcoma patients mentioned above. The results showed that PN expression was significantly (P < 0.05) higher in osteosarcoma (80.9%) than in osteochondroma (14.7%). Increased PN protein expression was associated with histological subtype (P = 0.000), Enneking stage (P = 0.027) and tumour size (P = 0.009). The result also showed that high expression of PN correlated with VEGF expression (r = 0.285; P = 0.019) and that tumours with PN-positive expression significantly had higher microvessal density (44.6 ± 13.7 vs. 20.6 ± 6.5; P = 0.000) compared to those in normal bone tissues. Additionally, the expression of PN was found to be an independent prognostic factor in osteosarcoma patients. In conclusion, our findings suggest that PN may have an important role in tumour progression and may be used as a prognostic biomarker for patients with osteosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Neovascularization, Pathologic , Osteosarcoma/diagnosis , Osteosarcoma/metabolism , Adolescent , Adult , Biomarkers, Tumor/metabolism , Bone Neoplasms/diagnosis , Disease Progression , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Osteosarcoma/blood supply , Osteosarcoma/pathology , Prognosis , Vascular Endothelial Growth Factor A/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...