Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
Add more filters

Country/Region as subject
Publication year range
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 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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Carcinogenesis ; 36(1): 104-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25330803

ABSTRACT

Chemokines modulate angiogenesis and metastasis that dictate cancer development in tumor microenvironment. Osteosarcoma is the most frequent bone tumor and is characterized by a high metastatic potential. Chemokine CCL5 (previously called RANTES) has been reported to facilitate tumor progression and metastasis. However, the crosstalk between chemokine CCL5 and vascular endothelial growth factor (VEGF) as well as tumor angiogenesis in human osteosarcoma microenvironment has not been well explored. In this study, we found that CCL5 increased VEGF expression and production in human osteosarcoma cells. The conditioned medium (CM) from CCL5-treated osteosarcoma cells significantly induced tube formation and migration of human endothelial progenitor cells. Pretreatment of cells with CCR5 antibody or transfection with CCR5 specific siRNA blocked CCL5-induced VEGF expression and angiogenesis. CCL5/CCR5 axis demonstrably activated protein kinase Cδ (PKCδ), c-Src and hypoxia-inducible factor-1 alpha (HIF-1α) signaling cascades to induce VEGF-dependent angiogenesis. Furthermore, knockdown of CCL5 suppressed VEGF expression and attenuated osteosarcoma CM-induced angiogenesis in vitro and in vivo. CCL5 knockdown dramatically abolished tumor growth and angiogenesis in the osteosarcoma xenograft animal model. Importantly, we demonstrated that the expression of CCL5 and VEGF were correlated with tumor stage according the immunohistochemistry analysis of human osteosarcoma tissues. Taken together, our findings provide evidence that CCL5/CCR5 axis promotes VEGF-dependent tumor angiogenesis in human osteosarcoma microenvironment through PKCδ/c-Src/HIF-1α signaling pathway. CCL5 may represent a potential therapeutic target against human osteosarcoma.


Subject(s)
Bone Neoplasms/blood supply , Chemokine CCL5/metabolism , Neovascularization, Pathologic/metabolism , Osteosarcoma/blood supply , Receptors, CCR5/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chick Embryo , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Chromatin Immunoprecipitation , Culture Media, Conditioned/pharmacology , Flow Cytometry , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, CCR5/chemistry , Receptors, CCR5/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
10.
Int J Cancer ; 137(1): 73-85, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25471534

ABSTRACT

Interleukin-34 (IL-34) was recently characterized as the M-CSF "twin" cytokine, regulating the proliferation/differentiation/survival of myeloid cells. The implication of M-CSF in oncology was initially suspected by the reduced metastatic dissemination in knock-out mice, due to angiogenesis impairment. Based on this observation, our work studied the involvement of IL-34 in the pathogenesis of osteosarcoma. The in vivo effects of IL-34 were assessed on tissue vasculature and macrophage infiltration in a murine preclinical model based on a paratibial inoculation of human osteosarcoma cells overexpressing or not IL-34 or M-CSF. In vitro investigations using endothelial cell precursors and mature HUVEC cells were performed to analyse the involvement of IL-34 in angiogenesis and myeloid cell adhesion. The data revealed that IL-34 overexpression was associated with the progression of osteosarcoma (tumor growth, lung metastases) and an increase of neo-angiogenesis. In vitro analyses demonstrated that IL-34 stimulated endothelial cell proliferation and vascular cord formation. Pre-treatment of endothelial cells by chondroitinases/heparinases reduced the formation of vascular tubes and abolished the associated cell signalling. In addition, IL-34 increased the in vivo recruitment of M2 tumor-associated macrophages into the tumor tissue. IL-34 increased in vitro monocyte/CD34(+) cell adhesion to activated HUVEC monolayers under physiological shear stress conditions. This work also demonstrates that IL-34 is expressed by osteosarcoma cells, is regulated by TNF-α, IL-1ß, and contributes to osteosarcoma growth by increasing the neo-angiogenesis and the recruitment of M2 macrophages. By promoting new vessel formation and extravasation of immune cells, IL-34 may play a key role in tumor development and inflammatory diseases.


Subject(s)
Bone Neoplasms/pathology , Interleukins/metabolism , Lung Neoplasms/pathology , Macrophages/metabolism , Neovascularization, Pathologic/metabolism , Osteosarcoma/pathology , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Interleukin-1beta/metabolism , Interleukins/pharmacology , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Mice , Neoplasm Transplantation , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Cancer Sci ; 106(10): 1394-401, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26250694

ABSTRACT

Angiogenesis plays an important role in tumor growth and metastasis and has been reported to be inversely correlated with overall survival of osteosarcoma patients. It has been shown that apurinic/apyrimidinic endonuclease 1 (APE1), a dually functional protein possessing both base excision repair and redox activities, is involved in tumor angiogenesis, although these mechanisms are not fully understood. Our previous study showed that the expression of transforming growth factor ß (TGFß) was significantly reduced in APE1-deficient osteosarcoma cells. Transforming growth factor ß promotes cancer metastasis through various mechanisms including immunosuppression, angiogenesis, and invasion. In the current study, we initially revealed that APE1, TGFß, and microvessel density (MVD) have pairwise correlation in osteosarcoma tissue samples, whereas TGFß, tumor size, and MVD were inversely related to the prognosis of the cohort. We found that knocking down APE1 in osteosarcoma cells resulted in TGFß downregulation. In addition, APE1-siRNA led to suppression of angiogenesis in vitro based on HUVECs in Transwell and Matrigel tube formation assays. Reduced secretory protein level of TGFß of culture medium also resulted in decreased phosphorylation of Smad3 of HUVECs. In a mouse xenograft model, siRNA-mediated silencing of APE1 downregulated TGFß expression, tumor size, and MVD. Collectively, the current evidence indicates that APE1 regulates angiogenesis in osteosarcoma by controlling the TGFß pathway, suggesting a novel target for anti-angiogenesis therapy in human osteosarcoma.


Subject(s)
Bone Neoplasms/blood supply , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , Transforming Growth Factor beta/metabolism , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Female , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/enzymology , Osteosarcoma/pathology , Phosphorylation , Prognosis , RNA Interference , RNA, Small Interfering , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/biosynthesis , Xenograft Model Antitumor Assays
12.
Ann Surg Oncol ; 22(2): 489-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25155396

ABSTRACT

BACKGROUND: Osteosarcoma is considered a highly vascularized bone tumor with early metastatic dissemination through intratumoral blood vessels mostly into the lung. Novel targets for therapy such as tumor vascularization are highly warranted since little progress has been achieved in the last 30 years. However, proof of relevance for vascularization as a major prognostic parameter has been hampered by tumor heterogeneity, difficulty in detecting microvessels by immunohistochemistry, and small study cohorts. Most recently, we demonstrated that highly standardized whole-slide imaging could overcome these limitations (Kunz et al., PloS One 9(3):e90727, 2014). In this study, we applied this method to a multicenter cohort of 131 osteosarcoma patients to test osteosarcoma vascularization as a prognostic determinant. METHODS: Computer-assisted whole-slide analysis, together with enzymatic epitope retrieval, was used for CD31-based microvessel quantification in 131 pretreatment formalin-fixed and paraffin-embedded biopsies from three bone tumor centers. Kaplan-Meier-estimated survival and chemoresponse were determined and multivariate analysis was performed. Conventional hot-spot-based microvessel density (MVD) determination was compared with whole-slide imaging. RESULTS: We detected high estimated overall (p ≤ 0.008) and relapse-free (p ≤ 0.004) survival in 25 % of osteosarcoma patients with low osteosarcoma vascularization in contrast to other patient groups. Furthermore, all patients with low osteosarcoma vascularization showed a good response to neoadjuvant chemotherapy. Comparison of conventional MVD determination with whole-slide imaging suggests false high quantification or even exclusion of samples with low osteosarcoma vascularization due to difficult CD31 detection in previous studies. CONCLUSION: Low intratumoral vascularization at the time of diagnosis is a strong predictor for prolonged survival and good response to neoadjuvant chemotherapy in osteosarcoma.


Subject(s)
Bone Neoplasms/blood supply , Bone Neoplasms/mortality , Osteosarcoma/blood supply , Osteosarcoma/mortality , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Adolescent , Adult , Aged , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Chemotherapy, Adjuvant , Child , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Middle Aged , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Prognosis , Young Adult
13.
Clin Orthop Relat Res ; 473(1): 305-10, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25193692

ABSTRACT

BACKGROUND: The survival of patients who present with nonmetastatic extremity osteosarcoma has dramatically improved, but there are some patients who do not respond to chemotherapy. The ability to identify patients with a poorer prognosis might allow us to target different therapy for these patients. Glucose transporter protein-1 (Glut-1), one of the key factors in glucose metabolism, has been reported to be an independent prognostic factor in various tumors. However, little is known about the role of the Glut-1 pathway in osteosarcoma. QUESTIONS/PURPOSES: We asked (1) if Glut-1 expression is a prognostic marker for survival in patients with osteosarcoma, and (2) if there is a relationship between Glut-1 expression and tumor angiogenesis. PATIENTS AND METHODS: Thirty-seven patients with resectable high-grade osteosarcomas treated between 1982 and 2007 were reviewed retrospectively. Patients were excluded if representative biopsy material and followup data were not available. The expression of Glut-1 and the number of CD34-positive microvessels for angiogenic activity were measured immunohistochemically. The median followup was 6 years 6 months (range, 11-211 months). Survival analyses were evaluated using the Kaplan-Meier method and the Cox proportional hazards model. The association between Glut-1 expression and microvessel density was analyzed using Student's t-test and chi-square test. For 12 (32.4%) of 37 patients with osteosarcoma, the expression of Glut-1 was positive, with four patients (10.8%) showing strong expression of Glut-1 protein. RESULTS: The expression of Glut-1 correlated with a shorter disease-free survival period (relative risk, 20.13; 95% CI, 1.77-229.3; p=0.0016). The microvessel density mean value of positive Glut-1 expression (mean±SD, 26.5±19.4) was lower than that of negative expression (mean±SD, 46.4±35.3; Student's t-test, p=0.038). When more than 50 was defined as a high microvessel density, positive expression of Glut-1 was significantly associated with low microvessel density (chi-square test, p=0.049). CONCLUSIONS: These findings indicate that Glut-1 is a potential predictor of survival in patients with osteosarcoma and that glucose metabolism may be negatively associated with angiogenesis. If substantiated in larger numbers of patients, these findings might stimulate the development of novel treatments for patients with a poorer prognosis. LEVEL OF EVIDENCE: Level III, prognostic study. See the Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/blood supply , Bone Neoplasms/chemistry , Glucose Transporter Type 1/analysis , Neovascularization, Pathologic , Osteosarcoma/blood supply , Osteosarcoma/chemistry , Adolescent , Adult , Antigens, CD34/analysis , Biopsy , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/surgery , Child , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Microvessels/chemistry , Microvessels/pathology , Middle Aged , Osteosarcoma/mortality , Osteosarcoma/pathology , Osteosarcoma/surgery , Osteotomy , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Time Factors , Treatment Outcome , Young Adult
14.
J Cell Biochem ; 115(9): 1490-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24590470

ABSTRACT

We previously described 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 ND-GFP mice, nascent blood vessels are labeled with GFP. We report here that osteosarcoma cells promote angiogenesis in the Gelfoam® angiogenesis assay in ND-GFP mice. Gelfoam® was initially transplanted subcutaneously in the flank of transgenic ND-GFP nude mice. Seven days after transplantation of Gelfoam®, skin flaps were made and human 143B osteosarcoma cells expressing green fluorescent protein (GFP) in the nucleus and red fluorescent protein (RFP) in cytoplasm were injected into the transplanted Gelfoam®. The control-group mice had only implanted Gelfoam®. Skin flaps were made at days 14, 21, and 28 after transplantation of the Gelfoam® to allow imaging of vascularization in the Gelfoam® using a variable-magnification small animal imaging system and confocal fluorescence microscopy. ND-GFP expressing nascent blood vessels penetrated and spread into the Gelfoam® in a time-dependent manner in both control and osteosarcoma-implanted mice. ND-GFP expressing blood vessels in the Gelfoam® of the osteosarcoma-implanted mice were associated with the cancer cells and larger and longer than in the Gelfoam®-only implanted mice (P < 0.01). The results presented in this report demonstrate strong angiogenesis induction by osteosarcoma cells and suggest this process is a potential therapeutic target for this disease.


Subject(s)
Green Fluorescent Proteins/biosynthesis , Luminescent Proteins/biosynthesis , Neovascularization, Pathologic/metabolism , Osteosarcoma/blood supply , Animals , Cell Line, Tumor , Female , Gelatin Sponge, Absorbable , Green Fluorescent Proteins/genetics , Humans , Implants, Experimental , Luminescent Proteins/genetics , Mice , Mice, Nude , Mice, Transgenic , Microscopy, Confocal , Neoplasm Transplantation , Red Fluorescent Protein
15.
Cancer Sci ; 105(2): 186-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24329908

ABSTRACT

Tumor angiogenesis contributes to inferior prognosis in osteosarcoma. Apurinic/apyrimidinic endonuclease 1 (APE1) and fibroblast growth factor 2 (FGF2) and its receptor 3 (FGFR3) signaling pathway plays an important role in the angiogenic process. In this study we observed that high expression of APE1, FGF2 and FGFR3, and microvessel density are positively correlated with poor prognosis of osteosarcoma patients. Furthermore, the Cox model showed that the tumor size, FGF2 and its receptor 3 (FGFR3), and microvessel density were adverse prognostic factors. Based on our clinical data, and the fact that APE1 is involved in tumor angiogenesis, we hypothesize that it is very likely that APE1 may indirectly promote angiogenesis by upregulating fibroblast FGF2 and FGFR3. Our preliminary data show small interfering RNA-mediated silence of APE1 experiments, which further supports this hypothesis. APE1-small interfering RNA significantly inhibited tumor angiogenesis by downregulating in vitro expression of FGF2 and FGFR3 in human umbilical vein endothelial cells in Matrigel tube formation assay, and further inhibited tumor growth in vivo in a mouse xenograft model. Thus, the proposed APE1-FGF2 and FGFR3 pathway may provide a novel mechanism for regulation of FGF2 and FGFR3 by APE1 in tumor angiogenesis.


Subject(s)
Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Fibroblast Growth Factor 2/genetics , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Adolescent , Adult , Aged , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Child , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Down-Regulation/genetics , Female , Fibroblast Growth Factor 2/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microvessels/pathology , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/genetics , Osteosarcoma/pathology , Prognosis , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Up-Regulation , Young Adult
16.
Adv Exp Med Biol ; 804: 285-9, 2014.
Article in English | MEDLINE | ID: mdl-24924180

ABSTRACT

Recent advances have shown that cell surface receptors are expressed differentially in normal and pathological conditions. Novel organ specific and disease specific proteins expressed on tumor vasculature have been identified by in vivo phage display technology and the diversity of the tumor-associated vasculature has provided the basis for the development of targeted therapeutics. Investigators recently screened a phage display library in a human cancer patient. An IL-11 mimic phage displaying the cyclic peptide CGRRAGGSC (single letter amino acid code) specifically bound to immobilized IL-11Rα. It has been demonstrated that the expression of the IL-11Rα is increased in several other types of tumors including osteosarcoma. The ability to selectively target the IL-11Rα may provide an alternative treatment of for a disease where new treatment options are truly needed.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/therapy , Lung Neoplasms/therapy , Osteosarcoma/therapy , Peptides, Cyclic/pharmacology , Receptors, Interleukin-11/genetics , T-Lymphocytes, Cytotoxic/immunology , Bone Neoplasms/blood supply , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Cell Engineering , Gene Expression , Humans , Immunotherapy , Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Molecular Targeted Therapy , Neovascularization, Pathologic , Osteosarcoma/blood supply , Osteosarcoma/immunology , Osteosarcoma/pathology , Peptide Library , Receptors, Interleukin-11/antagonists & inhibitors , Receptors, Interleukin-11/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/transplantation
17.
Adv Exp Med Biol ; 804: 67-92, 2014.
Article in English | MEDLINE | ID: mdl-24924169

ABSTRACT

The Notch pathway has been described as an oncogene in osteosarcoma, but the myriad functions of all the members of this complex signaling pathway, both in malignant cells and nonmalignant components of tumors, make it more difficult to define Notch as simply an oncogene or a tumor suppressor. The cell-autonomous behaviors caused by Notch pathway manipulation may vary between cell lines but can include changes in proliferation, migration, invasiveness, oxidative stress resistance, and expression of markers associated with stemness or tumor-initiating cells. Beyond these roles, Notch signaling also plays a vital role in regulating tumor angiogenesis and vasculogenesis, which are vital aspects of osteosarcoma growth and behavior in vivo. Further, osteosarcoma cells themselves express relatively low levels of Notch ligand, making it likely that nonmalignant cells, especially endothelial cells and pericytes, are the major source of Notch activation in osteosarcoma tumors in vivo and in patients. As a result, Notch pathway expression is not expected to be uniform across a tumor but likely to be highest in those areas immediately adjacent to blood vessels. Therapeutic targeting of the Notch pathway is likewise expected to be complicated. Most pharmacologic approaches thus far have focused on inhibition of gamma secretase, a protease of the presenilin complex. This enzyme, however, has numerous other target proteins that would be expected to affect osteosarcoma behavior, including CD44, the WNT/ß-catenin pathway, and Her-4. In addition, Notch plays a vital role in tissue and organ homeostasis in numerous systems, and toxicities, especially GI intolerance, have limited the effectiveness of gamma secretase inhibitors. New approaches are in development, and the downstream targets of Notch pathway signaling also may turn out to be good targets for therapy. In summary, a full understanding of the complex functions of Notch in osteosarcoma is only now unfolding, and this deeper knowledge will help position the field to better utilize novel therapies as they are developed.


Subject(s)
Bone Neoplasms/blood supply , Gene Expression Regulation, Neoplastic , Lung Neoplasms/blood supply , Osteosarcoma/blood supply , Receptors, Notch/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Inhibitors/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Neovascularization, Pathologic , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/secondary , Receptors, Notch/agonists , Receptors, Notch/antagonists & inhibitors , Signal Transduction
18.
Oncol Res ; 32(7): 1163-1172, 2024.
Article in English | MEDLINE | ID: mdl-38948019

ABSTRACT

Background: Osteosarcoma is the most common malignant primary bone tumor. The prognosis for patients with disseminated disease remains very poor despite recent advancements in chemotherapy. Moreover, current treatment regimens bear a significant risk of serious side effects. Thus, there is an unmet clinical need for effective therapies with improved safety profiles. Taurolidine is an antibacterial agent that has been shown to induce cell death in different types of cancer cell lines. Methods: In this study, we examined both the antineoplastic and antiangiogenic effects of taurolidine in animal models of osteosarcoma. K7M2 murine osteosarcoma cells were injected, both intramuscular and intraperitoneal, into 60 BALB/c mice on day zero. Animals were then randomized to receive treatment with taurolidine 2% (800 mg/kg), taurolidine 1% (400 mg/kg), or NaCl 0.9% control for seven days by intravenous or intraperitoneal administration. Results: After 35 days, mice were euthanized, and the tumors were harvested for analysis. Eighteen mice were excluded from the analysis due to complications. Body weight was significantly lower in the 2% taurolidine intraperitoneal treatment group from day 9 to 21, consistent with elevated mortality in this group. Intraperitoneal tumor weight was significantly lower in the 1% (p = 0.003) and 2% (p = 0.006) intraperitoneal taurolidine treatment groups compared to the control. No antineoplastic effects were observed on intramuscular tumors or for intravenous administration of taurolidine. There were no significant differences in microvessel density or mitotic rate between treatment groups. Reduced body weight and elevated mortality in the 2% taurolidine intraperitoneal group suggest that the lower 1% dose is preferable. Conclusions: In conclusion, there is no evidence of antiangiogenic activity, and the antitumor effects of taurolidine on osteosarcoma observed in this study are limited. Moreover, its toxic profile grants further evaluation. Given these observations, further research is necessary to refine the use of taurolidine in osteosarcoma treatment.


Subject(s)
Bone Neoplasms , Disease Models, Animal , Osteosarcoma , Taurine , Thiadiazines , Tumor Burden , Animals , Taurine/analogs & derivatives , Taurine/pharmacology , Taurine/therapeutic use , Thiadiazines/pharmacology , Thiadiazines/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/blood supply , Mice , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Tumor Burden/drug effects , Microvascular Density/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Humans , Neovascularization, Pathologic/drug therapy
19.
J Cell Physiol ; 228(4): 846-52, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23042366

ABSTRACT

Understanding the mechanisms inducing endothelial cell (EC) proliferation following tumor microenvironment stimuli may be important for the development of antiangiogenic therapies. Here, we show that cyclin-dependent kinase 2 and 5 (Cdk2, Cdk5) are important mediators of neoangiogenesis in in vitro and in vivo systems. Furthermore, we demonstrate that a specific Yin Yang 1 (YY1) protein-dependent signal from osteosarcoma (SaOS) cells determines proliferation of human aortic endothelial cells (HAECs). Following tumor cell stimuli, HAECs overexpress Cdk2 and Cdk5, display increased Cdk2 activity, undergo enhanced proliferation, and form capillary-like structures. Moreover, Roscovitine, an inhibitor of Cdks, blunted overexpression of Cdk2 and Cdk5 and Cdk2 activity induced by the YY1-dependent signal secreted by SaOS cells. Furthermore, Roscovitine decreased HAEC proliferation and angiogenesis (the latter by 70% in in vitro and 50% in in vivo systems; P < 0.01 vs. control). Finally, the finding that Roscovitine triggers apoptosis in SaOS cells as well as in HAECs by activating caspase-3/7 indicates multiple mechanisms for the potential antitumoral effect of Roscovitine. Present work suggests that Cdk2 and Cdk5 might be pharmacologically accessible targets for both antiangiogenic and antitumor therapy.


Subject(s)
Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Endothelial Cells/pathology , Osteosarcoma/blood supply , Osteosarcoma/metabolism , Animals , Antineoplastic Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apoptosis/drug effects , Apoptosis/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Culture Media , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Purines/pharmacology , Roscovitine , Up-Regulation , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
20.
J Pathol ; 227(2): 245-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22297719

ABSTRACT

About 40% of osteosarcoma patients die of metastases. Novel strategies to improve treatment of metastatic patients require a better understanding of the processes involved, like angiogenesis, migration, and the immune response. However, the rarity of osteosarcoma and its heterogeneity make this neoplasm difficult to study. Recently we reported malignant transformation of mouse mesenchymal stem cells (MSCs) which formed osteosarcoma upon transplantation into mice. Here we studied these cells in zebrafish embryos and found that transformed MSCs induced angiogenesis and migrated through the bodies of the embryos, but this was never observed with non-transformed normal MSCs (progenitors of the transformed MSCs). Whole genome expression analysis of both the cells and the host showed that angiogenesis and migration-related genes matrix metalloproteinase 19 (Mmp-19) and erythroblastosis virus E26 oncogene homologue 1 (Ets-1) were overexpressed in transformed MSCs compared to normal MSCs. Investigating the host response, embryos injected with transformed MSCs showed decreased expression of immune response-related genes, especially major histocompatibility complex class 1 (mhc1ze), as compared to embryos injected with normal MSCs. These findings contribute to the identification of genetic events involved in angiogenesis, migration, and host response providing targets as well as an appropriate model for high-throughput drug screens.


Subject(s)
Bone Neoplasms/enzymology , Cell Movement , Matrix Metalloproteinases, Secreted/metabolism , Neovascularization, Pathologic/enzymology , Osteosarcoma/enzymology , Proto-Oncogene Protein c-ets-1/metabolism , Tumor Escape , Zebrafish , Animals , Animals, Genetically Modified , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Carbocyanines/metabolism , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Fluorescent Dyes/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Matrix Metalloproteinases, Secreted/genetics , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Oligonucleotide Array Sequence Analysis , Osteosarcoma/blood supply , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/secondary , Proto-Oncogene Protein c-ets-1/genetics , Time Factors , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL