Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Front Pharmacol ; 15: 1421470, 2024.
Article in English | MEDLINE | ID: mdl-39050762

ABSTRACT

Background: Vasculogenic Mimicry (VM) can reduce the efficacy of anti-angiogenesis and promote distant metastasis in hepatocellular carcinoma (HCC). Our previous studies have found that Celastrus orbiculatus extract (COE) can inhibit the VM formation in HCC by reducing EphA2 expression. However the underlying mechanism related to EphA2 in VM formation is unclear. Purpose: This study aimed to confirm that EphA2 is one of the potential targets of COE, and to explore the effect of EphA2 in VM formation in hypoxia context in HCC. Methods: TCM Systems Pharmacology database and proteomics analysis were used to explore the key targets of COE in HCC treatment. CD31-PAS double staining and VE-CAD staining were used to indicate vasculogenic mimicry. The localization of EphA2 and VE-CAD was examined through fluorescent microscopy. CCK8 assay, cell invasion assay, and tube formation assay were used to indicate the formation of VM under hypoxic conditions. The regulatory relationship of EphA2 upstream and downstream molecules were evaluated through COIP and Western Blot. The nude mouse xenograft tumor models were used to observe the VM formation after knocking down or overexpressing EphA2. Results: EphA2 is identified to the target of COE, and the driving gene of HCC. In HCC surgical specimens, EphA2 expression is closely associated with the VM formation of HCC. COE-regulated EphA2 is involved in hypoxia-induced VM formation in HCC cells in vitro. EphA2 is regulated by HIF directly or indirectly by C-MYC. Overexpression of EphA2 can promote the VM formation of HCC in nude mice, while knocking down EphA2 can inhibit the VM formation. Conclusion: EphA2, as a target of COE, plays a crucial regulatory role in the formation of vasculogenic mimicry in HCC, involving upstream HIF/MYC transcriptional promotion and downstream PI3K/FAK/VE-CAD expression regulation.

2.
Cell Biol Toxicol ; 40(1): 44, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862832

ABSTRACT

BACKGROUND: Vasculogenic mimicry (VM) is an enigmatic physiological feature that influences blood supply within glioblastoma (GBM) tumors for their sustained growth. Previous studies identify NFATC3, FOSL1 and HNRNPA2B1 as significant mediators of VEGFR2, a key player in vasculogenesis, and their molecular relationships may be crucial for VM in GBM. AIMS: The aim of this study was to understand how NFATC3, FOSL1 and HNRNPA2B1 collectively influence VM in GBM. METHODS: We have investigated the underlying gene regulatory mechanisms for VM in GBM cell lines U251 and U373 in vitro and in vivo. In vitro cell-based assays were performed to explore the role of NFATC3, FOSL1 and HNRNPA2B1 in GBM cell proliferation, VM and migration, in the context of RNA interference (RNAi)-mediated knockdown alongside corresponding controls. Western blotting and qRT-PCR assays were used to examine VEGFR2 expression levels. CO-IP was employed to detect protein-protein interactions, ChIP was used to detect DNA-protein complexes, and RIP was used to detect RNA-protein complexes. Histochemical staining was used to detect VM tube formation in vivo. RESULTS: Focusing on NFATC3, FOSL1 and HNRNPA2B1, we found each was significantly upregulated in GBM and positively correlated with VM-like cellular behaviors in U251 and U373 cell lines. Knockdown of NFATC3, FOSL1 or HNRNPA2B1 each resulted in decreased levels of VEGFR2, a key growth factor gene that drives VM, as well as the inhibition of proliferation, cell migration and extracorporeal VM activity. Chromatin immunoprecipitation (ChIP) studies and luciferase reporter gene assays revealed that NFATC3 binds to the promoter region of VEGFR2 to enhance VEGFR2 gene expression. Notably, FOSL1 interacts with NFATC3 as a co-factor to potentiate the DNA-binding capacity of NFATC3, resulting in enhanced VM-like cellular behaviors. Also, level of NFATC3 protein in cells was enhanced through HNRNPA2B1 binding of NFATC3 mRNA. Furthermore, RNAi-mediated silencing of NFATC3, FOSL1 and HNRNPA2B1 in GBM cells reduced their capacity for tumor formation and VM-like behaviors in vivo. CONCLUSION: Taken together, our findings identify NFATC3 as an important mediator of GBM tumor growth through its molecular and epistatic interactions with HNRNPA2B1 and FOSL1 to influence VEGFR2 expression and VM-like cellular behaviors.


Subject(s)
Cell Movement , Cell Proliferation , Glioblastoma , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , NFATC Transcription Factors , Neovascularization, Pathologic , Proto-Oncogene Proteins c-fos , Humans , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/blood supply , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Animals , Cell Proliferation/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Cell Movement/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Gene Expression Regulation, Neoplastic , Mice , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/blood supply , Mice, Nude
3.
Cancers (Basel) ; 15(19)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37835469

ABSTRACT

Among the deadliest human cancers is glioblastoma (GBM) for which new treatment approaches are urgently needed. Here, the effects of the cyclic decapeptide, uPAcyclin, are investigated using the U87-MG, U251-MG, and U138-MG human GBM and C6 rat cell models. All GBM cells express the αV-integrin subunit, the target of uPAcyclin, and bind specifically to nanomolar concentrations of the decapeptide. Although peptide exposure affects neither viability nor cell proliferation rate, nanomolar concentrations of uPAcyclin markedly inhibit the directional migration and matrix invasion of all GBM cells, in a concentration- and αV-dependent manner. Moreover, wound healing rate closure of U87-MG and C6 rat glioma cells is reduced by 50% and time-lapse videomicroscopy studies show that the formation of vascular-like structures by U87-MG in three-dimensional matrix cultures is markedly inhibited by uPAcyclin. A strong reduction in the branching point numbers of the U87-MG, C6, and U251-MG cell lines undergoing vasculogenic mimicry, in the presence of nanomolar peptide concentrations, was observed. Lysates from matrix-recovered uPAcyclin-exposed cells exhibit a reduced expression of VE-cadherin, a prominent factor in the acquisition of vascular-like structures. In conclusion, these results indicate that uPAcyclin is a promising candidate to counteract the formation of new vessels in novel targeted anti-GBM therapies.

4.
Curr Eye Res ; 47(10): 1381-1388, 2022 10.
Article in English | MEDLINE | ID: mdl-35923150

ABSTRACT

PURPOSE: To explore the effect of doxycycline on vasculogenic mimicry (VM) formation and the potential mechanism in human pterygium fibroblasts in order to find novel targets for pterygium therapy. METHODS: First, we demonstrate the existence of VM in 73 pterygium specimens by CD31 and periodic acid Schiff (PAS) dual staining. Then we used cell counting kit-8, clone formation assay and flow cytometry to prove the inhibitory effect of doxycycline on cell proliferation and apoptosis. The VM formation was evaluated through wound healing assay, cell transwell assay and three-dimensional cell culture combined with PAS staining. Finally, we used Western blot to testify the correlation of the VM and the factors in protein level preliminarily. RESULTS: Our results showed that VM existed in human pterygium specimens exactly. Otherwise, in human pterygium fibroblasts, doxycycline induced a dose-dependent inhibitory effect on cell proliferation and apoptosis induction. Besides, doxycycline significantly suppressed vasculogenic mimicry tube formation, cell migration and invasion. Furthermore, doxycycline impaired the expression of MMP-9, MMP-2 and VEGF which may related to pterygium VM formation. CONCLUSIONS: Doxycycline decelerated pterygium progression might be through inhibiting VM formation according to the downregulation of MMP-9, MMP-2 and VEGF, which may provide the basis of further studies involving doxycycline for pterygium treatment.


Subject(s)
Matrix Metalloproteinase 2 , Pterygium , Cell Line, Tumor , Conjunctiva/abnormalities , Doxycycline/pharmacology , Fibroblasts/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/metabolism , Periodic Acid , Pterygium/drug therapy , Vascular Endothelial Growth Factor A/metabolism
6.
Transl Cancer Res ; 10(6): 3044-3052, 2021 Jun.
Article in English | MEDLINE | ID: mdl-35116612

ABSTRACT

OBJECTIVE: To discuss the role and mechanism of vasculogenic mimicry (VM) and to provide reference for the further research of VM in head and neck tumors. BACKGROUND: Head and neck tumors are common in the clinic, and tumor metastasis is clinically difficult to treat. VM is another tumor blood supply mode that is different from angiogenesis and plays an important role in tumor growth, metastasis, and invasion. At present, studies on VM have mainly focused on breast cancer, melanoma, glioblastoma, and other cancers. With time, VM has become a hotspot in head and neck tumor research. METHODS: We searched published English literatures from 2015 to 2020 on PubMed. In this paper, we review the progress of VM in head and neck tumors from 7 different perspectives. VM has two distinct types, namely tubular type and patterned matrix type. VM is associated with high tumor grade, tumor progression, invasion, metastasis, and poor prognosis in patients with head and neck tumors. We discuss the recent studies on the effects of immune cells and Epstein-Barr virus on VM in head and neck tumors. Furthermore, we also summarize the molecular mechanism of VM formation in head and neck tumors. Finally, we discussed the possibility of VM-targeted therapy in the clinical treatment of head and neck tumors. CONCLUSIONS: VM plays a critical role in tumor invasion, metastasis, and poor prognosis in patients with head and neck tumors. There is potential for VM as a potential new antitumor target. VM has become a hotspot in head and neck tumor research.

7.
Front Oncol ; 11: 746411, 2021.
Article in English | MEDLINE | ID: mdl-35251951

ABSTRACT

With the onset of resistance, ovarian cancer cells display almost unpredictable adaptive potential. This may derive from the tumor genetic ancestry and can be additionally tailored by post translational protein modifications (PTMs). In this study, we took advantage of high-end (phospho)-proteome analysis combined with multiparametric morphometric profiling in high-grade serous (OVCAR-3) and non-serous (SKOV-3) ovarian carcinoma cells. For functional experiments, we applied two different protocols, representing typical conditions of the abdominal cavity and of the growing tumor tissue: on the one side hypoxia (oxygen 1%) which develops within the tumor mass or is experienced during migration/extravasation in non-vascularized areas. On the other hand, fluid shear stress (250 rpm, 2.8 dyn/cm2) which affects tumor surface in the peritoneum or metastases in the bloodstream. After 3 hours incubation, treatment groups were clearly distinguishable by PCA analysis. Whereas basal proteome profiles of OVCAR-3 and SKOV-3 cells appeared almost unchanged, phosphoproteome analysis revealed multiple regulatory events. These affected primarily cellular structure and proliferative potential and consolidated in the proteome signature after 24h treatment. Upon oxygen reduction, metabolism switched toward glycolysis (e.g. upregulation hexokinase-2; HK2) and cell size increased, in concerted regulation of pathways related to Rho-GTPases and/or cytoskeletal elements, resembling a vasculogenic mimicry response. Shear stress regulated proteins governing cell cycle and structure, as well as the lipid metabolism machinery including the delta(14)-sterol reductase, kinesin-like proteins (KIF-22/20A) and the actin-related protein 2/3 complex. Independent microscopy-based validation experiments confirmed cell-type specific morphometric responses. In conclusion, we established a robust workflow enabling the description of the adaptive potential of ovarian cancer cells to physical and chemical stressors typical for the abdominal cavity and supporting the identification of novel molecular mechanisms sustaining tumor plasticity and pharmacologic resistance.

8.
Ann Palliat Med ; 9(6): 3985-3992, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33302660

ABSTRACT

BACKGROUND: To explore the relationship between CD177 and the vasculogenic mimicry (VM), clinicopathological parameters, and prognosis of epithelial ovarian cancer. METHODS: Tumor tissue samples and clinicopathological data were collected from 98 patients with epithelial ovarian cancer. The expression of CD177 in tumor tissues was detected by immunohistochemical streptavidin-peroxidase conjugate (SP) method, while the VM structure in tumor tissues was identified by CD31/periodic acid-Schiff (PAS) double staining in order to analyze the relationship between CD177, VM, clinicopathological parameters, and the prognosis of epithelial ovarian cancer. RESULTS: The proportion of the positive expression of CD177 (CD177+) in 98 ovarian cancer tissues was higher than that of the negative expression of CD177 (CD177-) (65.31% vs. 34.69%, P<0.05). Univariate analysis showed that CD177+ was associated with VM formation, tumor differentiation degree, tumor diameter, tumor stages, and platinum sensitivity (P<0.05), and was not associated with age, tumor types, or lymph node metastasis (P>0.05). Correlation analysis showed that CD177+ was positively correlated with VM formation, tumor differentiation degree, tumor diameter, and tumor stages (P<0.05), and was negatively correlated with platinum sensitivity (P<0.05). Kaplan-Meier survival analysis showed that the survival time of CD177+ patients was significantly shorter than that of CD177- patients (P<0.05). CONCLUSIONS: CD177+ is associated with the tumor malignancy of patients with epithelial ovarian cancer, and may participate in the formation of VM structure in epithelial ovarian cancer tissues. It can thus serve as important indicator for the prognosis of patients.


Subject(s)
Isoantigens , Ovarian Neoplasms , Receptors, Cell Surface , Carcinoma, Ovarian Epithelial , Female , GPI-Linked Proteins , Humans , Lymphatic Metastasis , Neovascularization, Pathologic , Prognosis
9.
Front Oncol ; 10: 18, 2020.
Article in English | MEDLINE | ID: mdl-32117706

ABSTRACT

Aberrant extra-vascular expression of VE-cadherin has been observed in metastasis associated with Vasculogenic Mimicry (VM); we have recently shown that in VM prone cells VE-cadherin is mainly in the form of phospho-VE-cadherin in Y658 allowing increased plasticity that potentiates VM development in malignant cells. In the current study, we present results to show that human malignant melanoma cells VM+, express the VE-cadherin phosphatase VE-PTP. VE-PTP forms a complex with VE-Cadherin and p120-catenin and the presence of this complex act as a safeguard to prevent VE-Cadherin protein degradation by autophagy. Indeed, VE-PTP silencing results in complete degradation of VE-cadherin with the features of autophagy. In summary, this study shows that VE-PTP is involved in VM formation and disruption of VE-PTP/VE-Cadherin/p120 complex results in enhanced autophagy in aggressive VM+ cells. Thus, we identify VE-PTP as a key player in VM development by regulating VE-cadherin protein degradation through autophagy.

10.
Front Oncol ; 9: 680, 2019.
Article in English | MEDLINE | ID: mdl-31428573

ABSTRACT

The term vasculogenic mimicry (VM) refers to the capacity of certain cancer cells to form fluid-conducting structures within a tumor in an endothelial cell (EC)-free manner. Ever since its first report by Maniotis in 1999, the existence of VM has been an extremely contentious issue. The overwhelming consensus of the literature suggests that VM is frequently observed in highly aggressive tumors and correlates to lower patient survival. While the presence of VM in vivo in animal and patient tumors are claimed upon the strong positive staining for glycoproteins (Periodic Acid Schiff, PAS), it is by no means universally accepted. More controversial still is the existence of an in vitro model of VM that principally divides the scientific community. Original reports demonstrated that channels or tubes occur in cancer cell monolayers in vitro when cultured in matrigel and that these structures may support fluid movement. However, several years later many papers emerged stating that connections formed between cancer cells grown on matrigel represented VM. We speculate that this became accepted by the cancer research community and now the vast majority of the scientific literature reports both presence and mechanisms of VM based on intercellular connections, not the presence of fluid conducting tubes. In this opinion paper, we call upon evidence from an exhaustive review of the literature and original data to argue that the majority of in vitro studies presented as VM do not correspond to this phenomenon. Furthermore, we raise doubts on the validity of concluding the presence of VM in patient samples and animal models based solely on the presence of PAS+ staining. We outline the requirement for new biomarkers of VM and present criteria by which VM should be defined in vitro and in vivo.

11.
J Cell Biochem ; 120(2): 1068-1079, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30431663

ABSTRACT

The epithelial-mesenchymal transition (EMT) program, which loosens cell-cell adhesion complexes, endows cells with enhanced migratory and invasive properties. Furthermore, this process facilitates both the development of drug resistance and immunosuppression by tumor cells, which preclude the successful treatment of cancer. Recent research has demonstrated that many signaling pathways are involved in EMT progression. In addition, cancer stem cells (CSCs), vasculogenic mimicry (VM) and the tumor-related immune microenvironment all play important roles in tumor formation. However, there are few reports on the relationships between EMT and these factors. In addition, in recent years, traditional Chinese medicine (TCM) has developed a unique system for treating cancer. In this review, we summarize the crucial signaling pathways associated with the EMT process in cancer patients and discuss the interconnections between EMT and other molecular factors (such as CSCs, VM, and the tumor-related immune microenvironment). We attempt to identify common regulators that might be potential therapeutic targets to thereby optimize tumor treatment. In addition, we outline recent research on TCM approaches that target EMT and thereby provide a foundation for further research on the exact mechanisms by which TCMs affect EMT in cancer.

12.
Biomed Pharmacother ; 96: 982-992, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29208325

ABSTRACT

Breast cancer is one of the most serious malignant tumors that harm to women's health. Vasculogenic mimicry (VM) is an alternative type of blood supplement independent of endothelial vessels, which refers to the formation of tumor cell-lined vessels and is associated with tumor invasion, metastasis and poor cancer patient prognosis. Prior antiangiogenic therapy just focused on vascular endothelial cells and did not significantly affect VM. DMBT, 6, 6'-bis (2, 3-dimethoxybenzoyl)-a, a-D-trehalose, has shown to have multiple anti-tumor invasion and metastasis activities; however the exact mechanism is not thoroughly elucidated. In this study, we examined key molecular mechanisms underlying VM by using breast cancer cells MDA-MB-231 and MCF-7. We found that following the hypoxia treatment, the cells were easily to form VM networks and DMBT could inhibit VM formation of both MDA-MB-231 and MCF-7 cells in hypoxic condition. When tumor cells exposed to hypoxia environment, the expression of VM related proteins such as HIF-1α, VE-cadherin, MMP-9, Cdc42, and EGFR, p-Akt, p-mTOR were increased but decreased when exposed to hypoxia medium with DMBT. In MDA-MB-231 cells, DMBT inhibit hypoxia-induced VM by suppress HIF-1α/VE-cadherin/MMPs signaling pathway and in MCF-7 cells, DMBT had little effect on HIF-1α or VE-cadherin but could inhibit cell autophagy to suppress VM formation. These results suggested that DMBT could serve as a therapeutic agent to inhibit VM formation in human breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Hypoxia/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antigens, CD/metabolism , Breast Neoplasms/metabolism , Cadherins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
13.
Oncotarget ; 8(29): 47801-47815, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28599281

ABSTRACT

Vasculogenic mimicry (VM) is a blood supply modality that occurs independently of endothelial cell angiogenesis. Hypoxia and the epithelial-mesenchymal transition (EMT) induce VM formation by remodeling the extracellular matrix. Our previous study demonstrated that hypoxia-inducible factor-2 alpha (HIF-2α) promotes the progress of EMT in pancreatic cancer; however, whether HIF-2α promotes VM formation in pancreatic cancer remains unknown. In this study, we investigated HIF-2α expression and VM by immunohistochemistry in 70 pancreatic cancer patients as well as the role of Twist1and Twist2 in HIF-2α-induced VM in vitro and in vivo. We found that the overexpression of HIF-2α and VM were correlated with poor tumor differentiation, late clinical stage and lymph node metastasis, and a poor prognosis in pancreatic cancer. Moreover, the upregulation of HIF-2α in SW1990 cells induced VM formation, whereas the opposite results were found after silencing HIF-2α in AsPC-1 cells. A mechanistic study indicated that HIF-2α might regulate the binding of twist1 to vascular endothelial cadherin (VE-cadherin) to promote VM formation in pancreatic cancer cells, and that the P1 (-421bp) and P4 (-2110bp) regions of the Twist1 binding sequences are positive regulatory elements for VE-cadherin. In addition, we confirmed that the overexpression of HIF-2α increased Twist1 expression and promoted tumor growth and VM formation in pancreatic cancer xenografts in nude mice. These findings indicated that HIF-2α might play a critical role in VM and that HIF-2α and the pathway of HIF-2α inducing VM formation are potential therapeutic targets for pancreatic cancer.


Subject(s)
Antigens, CD/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadherins/genetics , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , Twist-Related Protein 1/metabolism , Aged , Aged, 80 and over , Animals , Antigens, CD34/metabolism , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Female , Heterografts , Humans , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Protein Binding
14.
Oncotarget ; 8(2): 2501-2513, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27705934

ABSTRACT

Hypervascularity is one of the main characteristics of hepatocellular carcinoma (HCC). However, the mechanisms of angiogenesis in HCC remain controversial. In this study, we investigate the role of Notch1 in angiogenesis of HCC. We found that Notch1 expression was correlated with formation of vasculogenic mimicry (VM) and expression of biomarkers of epithelial-to-mesenchymal transition (EMT) in the tumor specimens. Two HCC cell lines, HepG2 and MHCC97-H, with low and high Notch1 expression, respectively, were used to study the mechanism of VM formation both in vitro and in vivo. It was found that MHCC97-H cells, but not HepG2 cells form VM when they grow on matrigel in vitro. HepG2 cells gained the power of forming VM when they were overexpressed with Notch1, while knockdown Notch1 expression in MHCC97-H cells led to the loss of VM forming ability of the cells. Similar results were found in in vivo study. High expression of Notch1 in HepG2 promoted xenograft growth in nude mice, with abundant VM formation in the tumor samples. Moreover, we observed Notch1 was associated with the EMT and malignant behavior of hepatocellular carcinoma by analyzing clinical specimens, models for in vitro and in vivo experiments. HepG2 presented EMT phenomenon when induced by TGF-ß1, accompanied by Notch1 activation while MHCC97-H with knockdown of Notch1 lost the responsiveness to TGF-ß1 induction. Our results suggest that Notch1 promotes HCC progression through activating EMT pathway and forming VM. Our results will guide targeting Notch1 in new drug development.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Receptor, Notch1/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Receptor, Notch1/genetics , Up-Regulation
15.
Oncotarget ; 7(47): 77987-77997, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27793002

ABSTRACT

Portal vein invasion (PVI) is common in hepatocellular carcinoma (HCC) and largely contributes to tumor recurrence after radical tumor resection or liver transplantation. Vasculogenic mimicry (VM) was an independent vascular system lined with tumor cells and associated with poor prognosis of HCC. The present study was conducted to evaluate the relationship between VM and portal vein invasion. A total of 44 HCC cases receiving anatomic liver resection were included in the study and were divided into groups with and without PVI. The prevalence of VM in each group was examined by CD34-PAS dual staining. The regulatory molecules of VM formation such as Notch1, Vimentin and matrix metalloproteinases (MMPs) were investigated by immunohistochemical staining. Analysis was performed to explore the association of PVI, VM and the VM regulatory molecules. PVI was found in 40.91% (18/44) cases and VM was found in 38.64% (17/44) cases in total samples. The incidence of VM was 72.22% (13/18) in PVI group while it was 15.38% (4/26) in non-PVI group (P<0.001), VM formation was positively correlated with PVI (r=0.574, P<0.001). The VM forming regulatory molecules such as Notch1, Vimentin, MMP-2 and MMP-9 were found to be correlated with PVI in HCC patients. Taken together, our results suggested that VM formation, alone with its regulatory molecules, is the promoting factor of PVI in hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Liver Neoplasms/blood supply , Portal Vein/pathology , Carcinoma, Hepatocellular/pathology , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neovascularization, Pathologic/pathology
16.
Oncotarget ; 7(20): 29051-62, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27074560

ABSTRACT

Cancer stem cells (CSCs) have gained much attention due to their roles in the invasion and metastasis of numerous kinds of human cancers. Here, we showed that the positive expression of CD133, the stemness marker, was positively associated with vasculogenic mimicry (VM) formation, local regional recurrence, distant metastasis and poorer prognosis in salivary adenoid cystic carcinoma (ACC) specimens. Compared with CD133- ACC cells, CD133+ cancer stem-like cells had more migration and invasion capabilities, as well as more VM formation. The levels of endothelial cell marker VE-cadherin, MMP-2 and MMP-9 expression in CD133+ cancer stem-like cells and xenograft tumors of nude mice injected with CD133+ cells were significantly higher than those with CD133- cells. The data indicated that CD133+ cancer stem-like cells might contribute to the migration and invasion of ACC through inducing VM formation.


Subject(s)
Carcinoma, Adenoid Cystic/pathology , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/pathology , Salivary Gland Neoplasms/pathology , AC133 Antigen/metabolism , Adult , Aged , Animals , Cell Line, Tumor , Cell Movement , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Middle Aged
17.
Biomark Res ; 3: 8, 2015.
Article in English | MEDLINE | ID: mdl-26085929

ABSTRACT

Glioblastoma (GBM) is a highly angiogenic malignancy that is resistant to standard therapy; neo-formed vessels of this aggressive malignancy are thought to arise by sprouting of pre-existing brain capillaries. However, the conventional anti-angiogenic therapy, which seemed promising initially, shows transitory and incomplete efficacy. The discovery of vasculogenic mimicry (VM) has offered a new horizon for understanding tumor vascularization. VM is a tumor cell-constituted, matrix-embedded fluid-conducting meshwork that is independent of endothelial cells and is positively correlated with poor prognosis. Therefore, a better understanding of GBM vasculature is needed to optimize anti-angiogenic therapy. This review focuses on the signaling molecules and cascades involved in VM in relation to ongoing glioma research, as well as the clinical translational advances in GBM that have been offered by the development of optimized anti-angiogenesis treatment modalities.

SELECTION OF CITATIONS
SEARCH DETAIL