Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters








Publication year range
1.
J Transl Med ; 22(1): 113, 2024 01 29.
Article in English | MEDLINE | ID: mdl-38281945

ABSTRACT

BACKGROUND: Ovarian cancer poses a serious threat to women's health. Due to the difficulty of early detection, most patients are diagnosed with advanced-stage disease or peritoneal metastasis. We found that LncRNA MEG3 is a novel tumor suppressor, but its role in tumor occurrence and development is still unclear. METHODS: We investigated the expression level of MEG3 in pan-cancer through bioinformatics analysis, especially in gynecological tumors. Function assays were used to detect the effect of MEG3 on the malignant phenotype of ovarian cancer. RIP, RNA pull-down, MeRIP-qPCR, actinomycin D test were carried out to explore the m6A methylation-mediated regulation on MEG3. Luciferase reporter gene assay, PCR and Western blot were implemented to reveal the potential mechanism of MEG3. We further confirmed the influence of MEG3 on tumor growth in vivo by orthotopic xenograft models and IHC assay. RESULTS: In this study, we discovered that MEG3 was downregulated in various cancers, with the most apparent downregulation in ovarian cancer. MEG3 inhibited the proliferation, migration, and invasion of ovarian cancer cells. Overexpression of MEG3 suppressed the degradation of VASH1 by negatively regulating miR-885-5p, inhibiting the ovarian cancer malignant phenotype. Furthermore, we demonstrated that MEG3 was regulated at the posttranscriptional level. YTHDF2 facilitated MEG3 decay by recognizing METTL3­mediated m6A modification. Compared with those injected with vector control cells, mice injected with MEG3 knockdown cells showed larger tumor volumes and faster growth rates. CONCLUSION: We demonstrated that MEG3 is influenced by METTL3/YTHDF2 methylation and restrains ovarian cancer proliferation and metastasis by binding miR-885-5p to increase VASH1 expression. MEG3 is expected to become a therapeutic target for ovarian cancer.


Subject(s)
MicroRNAs , Ovarian Neoplasms , RNA, Long Noncoding , Animals , Female , Humans , Mice , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Methylation , Methyltransferases/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
2.
Cancer Biol Ther ; 24(1): 2285817, 2023 12 31.
Article in English | MEDLINE | ID: mdl-38010374

ABSTRACT

The function of Vasohibin-1 (VASH1) in human cancer has not been thoroughly or comprehensively examined. Here, we identified the tumor suppressor part of VASH1 across cancers, including epithelial ovarian tumors. Our study carefully contrasted the expression of VASH1 in pancancer and nontumorous tissues in a public database to explore its regulatory role in clinical prognosis, diagnosis, tumor purity, and immune cell infiltration. Next, we explored the antitumor mechanism of VASH1 through drug sensitivity, functional enrichment, and phenotypic experiments in ovarian cancer. Research suggests that the expression of VASH1 in neoplastic tissues is lower than that in normal tissues. VASH1 affects the OS and RFS of several tumor types. In addition, VASH1 expression resulted in a high OS and RFS in the diagnosis of tumor and nontumor tissues and negatively regulated tumor purity. Moreover, VASH1 controls the tumor microenvironment by regulating immunocyte infiltration. In ovarian cancer, VASH1 can serve as a biomarker to estimate the efficacy of chemotherapy. Functional enrichment analysis suggests that VASH1 plays a tumor suppressor role by regulating the extracellular matrix receptor pathway. VASH1 inhibition of the malignant phenotype of ovarian cancer cells was further confirmed by in vivo experiments. These results indicate that VASH1 acts as a cancer-inhibiting factor and potential therapeutic target in ovarian cancer.


Subject(s)
Ovarian Neoplasms , Humans , Female , Prognosis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Tumor Microenvironment
3.
Tumour Virus Res ; 16: 200269, 2023 12.
Article in English | MEDLINE | ID: mdl-37499979

ABSTRACT

PROBLEM: Human papillomavirus infection is integral to developing invasive cervical cancer in the majority of patients. In a recent genome-wide association study, rs9357152 and rs4243652 have been associated with seropositivity for HPV16 or HPV18, respectively. It is unknown whether these variants also associate with cervical cancer triggered by either HPV16 or HPV18. METHODS: We investigate whether the two HPV susceptibility variants show association with type-specific cervical cancer in a genetic case-control study with cases stratified by HPV16 or HPV18, respectively. We further tested whether rs9357152 modulates gene expression of any of 36 genes at the human leukocyte antigen locus in 256 cervical tissues. RESULTS: rs9357152 was associated with invasive HPV16-positive cervical cancer (OR 1.33, 95%CI 1.03-1.70, p = 0.03), and rs4243652 was associated with HPV18-positive adenocarcinomas (OR 2.96, 95%CI 1.18-7.41, p = 0.02). These associations remained borderline significant after testing against different sets of controls. rs9357152 was found to be an eQTL for HLA-DRB1 in HPV-positive cervical tissues (pANOVA = 0.0009), with the risk allele lowering mRNA levels. CONCLUSIONS: We find evidence that HPV seropositivity variants at chromosome 6 and 14 may modulate type-specific cervical cancer risk. rs9357152 may exert its effect through regulating HLA-DRB1 induction in the presence of HPV. In regard of multiple testing, these results need to be confirmed in larger studies.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/genetics , HLA-DRB1 Chains/genetics , Papillomavirus Infections/complications , Case-Control Studies , Genome-Wide Association Study , Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Genomics
4.
Reprod Biol ; 22(1): 100592, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34995817

ABSTRACT

Endometriosis is a benign gynecological disease. Accumulating evidence has revealed the participation of dysregulated miRNAs in the progression of endometriosis. Here, the function and molecular mechanism of miR-143-3p in endometriosis were investigated. The levels of vasohibin 1 (VASH1) and miR-143-3p in endometrial tissues and endometriotic stromal cells (ESCs) were detected by RT-qPCR. Migrative and invasive phenotypes of ESCs were tested by Transwell assays. The protein expression of VASH1, TGF-ß signaling markers, and epithelial to mesenchymal transition (EMT) markers was examined by western blotting. The targeted relationship between miR-143-3p and VASH1 was confirmed by bioinformatics analysis and luciferase reporter assay. We found that miR-143-3p expression was significantly upregulated in ectopic endometrial tissues compared to that in eutopic and normal endometrial tissues. MiR-143-3p knockdown restrained EMT process, invasive and migrative behaviors of ESCs. Mechanically, miR-143-3p targeted VASH1 and negatively regulated VASH1. VASH1 downregulation reserved the effects of miR-143-3p knockdown in ESCs. MiR-143-3p activated TGF-ß signaling via targeting VASH1. Furthermore, activation of TGF-ß signaling counteracted the miR-143-3p knockdown-caused suppression of migration, invasion and EMT process in ESCs. Overall, miR-143-3p activates TGF-ß signaling by targeting VASH1 to facilitate migration and invasion of ESCs.


Subject(s)
Endometriosis , MicroRNAs , Stromal Cells , Cell Cycle Proteins/metabolism , Cell Movement/genetics , Endometriosis/genetics , Endometriosis/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism
5.
Development ; 148(4)2021 02 19.
Article in English | MEDLINE | ID: mdl-33547133

ABSTRACT

Previous studies have shown that Vasohibin 1 (Vash1) is stimulated by VEGFs in endothelial cells and that its overexpression interferes with angiogenesis in vivo Recently, Vash1 was found to mediate tubulin detyrosination, a post-translational modification that is implicated in many cell functions, such as cell division. Here, we used the zebrafish embryo to investigate the cellular and subcellular mechanisms of Vash1 on endothelial microtubules during formation of the trunk vasculature. We show that microtubules within venous-derived secondary sprouts are strongly and selectively detyrosinated in comparison with other endothelial cells, and that this difference is lost upon vash1 knockdown. Vash1 depletion in zebrafish specifically affected secondary sprouting from the posterior cardinal vein, increasing endothelial cell divisions and cell number in the sprouts. We show that altering secondary sprout numbers and structure upon Vash1 depletion leads to defective lymphatic vessel formation and ectopic lymphatic progenitor specification in the zebrafish trunk.


Subject(s)
Cell Cycle Proteins/genetics , Embryonic Development/genetics , Lymphangiogenesis/genetics , Zebrafish/embryology , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Conserved Sequence , Evolution, Molecular , Gene Expression Regulation, Developmental , Immunohistochemistry , Microtubules/metabolism , Models, Biological
6.
Acta Crystallogr D Struct Biol ; 76(Pt 10): 993-1000, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33021501

ABSTRACT

Vasohibins regulate angiogenesis, tumor growth, metastasis and neuronal differentiation. They form a complex with small vasohibin-binding protein (SVBP) and show tubulin tyrosine carboxypeptidase activity. Recent crystal structure determinations of vasohibin-SVBP complexes have provided a molecular basis for complex formation, substrate binding and catalytic activity. However, the regulatory mechanism and dynamics of the complex remain elusive. Here, the crystal structure of the VASH1-SVBP complex and a molecular-dynamics simulation study are reported. The overall structure of the complex was similar to previously reported structures. Importantly, however, the structure revealed a domain-swapped heterotetramer that was formed between twofold symmetry-related molecules. This heterotetramerization was stabilized by the mutual exchange of ten conserved N-terminal residues from the VASH1 structural core, which was intramolecular in other structures. Interestingly, a comparison of this region with previously reported structures revealed that the patterns of hydrogen bonding and hydrophobic interactions vary. In the molecular-dynamics simulations, differences were found between the heterotetramer and heterodimer, where the fluctuation of the N-terminal region in the heterotetramer was suppressed. Thus, heterotetramer formation and flexibility of the N-terminal region may be important for enzyme activity and regulation.


Subject(s)
Carrier Proteins/chemistry , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Protein Domains , Protein Multimerization
7.
Mol Cancer ; 18(1): 181, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31823788

ABSTRACT

BACKGROUND: Brain metastasis (BM) is one of the principal causes of mortality for lung cancer patients. While the molecular events that govern BM of lung cancer remain frustrating cloudy. METHODS: The miRNA expression profiles are checked in the paired human BM and primary lung cancer tissues. The effect of miR-143-3p on BM of lung cancer cells and its related mechanisms are investigated. RESULTS: miR-143-3p is upregulated in the paired BM tissues as compared with that in primary cancer tissues. It can increase the invasion capability of in vitro blood brain barrier (BBB) model and angiogenesis of lung cancer by targeting the three binding sites of 3'UTR of vasohibin-1 (VASH1) to inhibit its expression. Mechanistically, VASH1 can increase the ubiquitylation of VEGFA to trigger the proteasome mediated degradation, further, it can endow the tubulin depolymerization through detyrosination to increase the cell motility. m6A methyltransferase Mettl3 can increase the splicing of precursor miR-143-3p to facilitate its biogenesis. Moreover, miR-143-3p/VASH1 axis acts as adverse prognosis factors for in vivo progression and overall survival (OS) rate of lung cancer. CONCLUSIONS: Our work implicates a causal role of the miR-143-3p/VASH1 axis in BM of lung cancers and suggests their critical roles in lung cancer pathogenesis.


Subject(s)
Adenosine/analogs & derivatives , Brain Neoplasms/secondary , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Animals , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Mice , Models, Biological , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA Interference , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
8.
Technol Cancer Res Treat ; 18: 1533033819879905, 2019.
Article in English | MEDLINE | ID: mdl-31757187

ABSTRACT

BACKGROUND: Hepatocellular carcinoma is a highly vascularized tumor, so it is critical to study its angiogenesis. Cancer-associated fibroblasts and enhancer of zeste homolog 2 play an important role in tumor angiogenesis and became significant hallmarks of cancer. But the relationship between enhancer of zeste homolog-2 and cancer-associated fibroblasts in response to angiogenesis and its precise mechanism remains unclear. METHODS: The separation of cancer-associated fibroblasts was identified by immunofluorescence. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis was used to reveal the proliferation of human umbilical vein endothelial cells. Vascular endothelial growth factor level was quantified by enzyme-linked immunosorbent assay. The wound healing, transwell, and vascular tube formation assays were used to identify the capability of migration, invasion, and tube formation of human umbilical vein endothelial cells in vitro. The detection of tumor angiogenesis was also performed in vivo. Finally, the level of enhancer of zeste homolog-2 and vasohibin 1 were determined by quantitative real-time polymerase chain reaction and Western blotting. RESULTS: In comparison to control and condition medium noncancerous fibroblasts groups, the condition medium cancer-associated fibroblasts could significantly promote the proliferation, migration, invasion, and angiogenesis of human umbilical vein endothelial cells. We found that cancer-associated fibroblasts promoted angiogenesis of human umbilical vein endothelial cells via vascular endothelial growth factor secretion in vitro and in vivo. The upregulation of enhancer of zeste homolog 2 by vascular endothelial growth factor inhibited the expression of vasohibin 1, thus promoting the proliferation and angiogenesis of human umbilical vein endothelial cells. Taken together, the cancer-associated fibroblasts of hepatocellular carcinoma regulate the enhancer of zeste homolog-2/vasohibin 1 pathway via vascular endothelial growth factor secretion, thereby promoting the proliferation and angiogenesis of human umbilical vein endothelial cells. CONCLUSION: Our study identified the relationship between cancer-associated fibroblasts and enhancer of zeste homolog-2 and confirmed the pivotal role of cancer-associated fibroblasts in angiogenesis of hepatocellular carcinoma. Cancer-associated fibroblasts promote angiogenesis of hepatocellular carcinoma by vascular endothelial growth factor-mediated enhancer of zeste homolog-2/vasohibin 1 pathway and may be a potentially useful therapeutic target for hepatocellular carcinoma.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Liver Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Biopsy , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Models, Animal , Heterografts , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Mice
9.
Genet Med ; 21(8): 1790-1796, 2019 08.
Article in English | MEDLINE | ID: mdl-30607023

ABSTRACT

PURPOSE: Identifying and characterizing novel causes of autosomal recessive intellectual disability based on systematic clinical and genetic evaluation, followed by functional experiments. METHODS: Clinical examinations, genome-wide positional mapping, and sequencing were followed by quantitative polymerase chain reaction and western blot of the protein SVBP and its interaction partners. We then knocked down the gene in rat primary hippocampal neurons and evaluated the consequences on synapses. RESULTS: We identified a founder, homozygous stop-gain variant in SVBP (c.82C>T; p.[Gln28*]) in four affected individuals from two independent families with intellectual disability, microcephaly, ataxia, and muscular hypotonia. SVBP encodes a small chaperone protein that transports and stabilizes two angiogenesis regulators, VASH1 and VASH2. The altered protein is unstable and nonfunctional since transfected HeLa cells with mutant SVBP did not reveal evidence for immunoreactive SVBP protein fragments and cotransfection with VASH1 showed a severe reduction of VASH1 in medium and cell lysate. Knocking down Svbp in rat primary hippocampal neurons led to a significant decrease in the number of excitatory synapses. CONCLUSION: SVBP is not only involved in angiogenesis, but also has vital functions in the central nervous system. Biallelic loss-of-function variants in SVBP lead to intellectual disability.


Subject(s)
Carrier Proteins/genetics , Genes, Recessive/genetics , High-Throughput Nucleotide Sequencing , Intellectual Disability/genetics , Angiogenic Proteins , Animals , Ataxia/epidemiology , Ataxia/genetics , Ataxia/pathology , Cell Cycle Proteins , Female , Genotype , HeLa Cells , Homozygote , Humans , Intellectual Disability/epidemiology , Intellectual Disability/pathology , Loss of Function Mutation/genetics , Male , Microcephaly/epidemiology , Microcephaly/genetics , Microcephaly/pathology , Muscle Hypotonia/epidemiology , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Pedigree , Rats
10.
J Cell Physiol ; 234(5): 6654-6666, 2019 05.
Article in English | MEDLINE | ID: mdl-30341900

ABSTRACT

Multiple studies have reported different methods in treating gestational diabetes mellitus (GDM); however, the relationship between miR-335-5p and GDM still remains unclear. Here, this study explores the effect of miR-335-5p on insulin resistance and pancreatic islet ß-cell secretion via activation of the TGFß signaling pathway by downregulating VASH1 expression in GDM mice. The GDM mouse model was established and mainly treated with miR-335-5p mimic, miR-335-5p inhibitor, si-VASH1, and miR-335-5p inhibitor + si-VASH1. Oral glucose tolerance test (OGTT) was conducted to detect fasting blood glucose (FBG) fasting insulin (FINS). The OGTT was also used to calculate a homeostasis model assessment of insulin resistance (HOMA-IR). A hyperglycemic clamp was performed to measure the glucose infusion rate (GIR), which estimated ß-cell function. Expressions of miR-335-5p, VASH1, TGF-ß1, and c-Myc in pancreatic islet ß-cells were determined by RT-qPCR, western blot analysis, and insulin release by ELISA. The miR-335-5p mimic and si-VASH1 groups showed elevated blood glucose levels, glucose area under the curve (GAUC), and HOMA-IR, but a reduced GIR and positive expression of VASH1. Overexpression of miR-335-5p and inhibition of VASH1 contributed to activated TGFß1 pathway, higher c-Myc, and lower VASH1 expressions, in addition to downregulated insulin and insulin release levels. These findings provided evidence that miR-335-5p enhanced insulin resistance and suppressed pancreatic islet ß-cell secretion by inhibiting VASH1, eventually activating the TGF-ß pathway in GDM mice, which provides more clinical insight on the GDM treatment.


Subject(s)
Blood Glucose/genetics , Diabetes, Gestational/genetics , Insulin Resistance/genetics , MicroRNAs/genetics , Transforming Growth Factor beta/deficiency , Animals , Blood Glucose/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Glucose Tolerance Test/methods , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Male , Mice , Pregnancy , Transforming Growth Factor beta/metabolism
11.
Oncol Lett ; 16(4): 5265-5274, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30250596

ABSTRACT

Vasohibin (VASH) -1 and -2 are novel angiogenic regulators. The aim of the present study was to assess the prognostic values of VASH1 expression and VASH2 expression in esophageal squamous cell carcinoma (ESCC). A total of 209 patients with ESCC were investigated. Resected tumor specimens were immunostained using anti-CD34 antibody, anti-VASH1 antibody and anti-VASH2 antibody. The ratio of the microvessels density and the VASH1 density as the VASH1-positive ratio were defined and the patients were divided into two groups (a high VASH1 group and a low VASH1 group) according to the average value. The patients were also divided into two groups (a high VASH2 group and a low VASH2 group) according to VASH2 expression upon immunostaining. The clinical outcomes of these two groups were then evaluated. The high VASH1 group contained 106 patients (50.7%). The high VASH2 group contained 48 patients (23.0%). Long-term survival was significantly poorer in the high VASH1 group compared with that in the low VASH1 group. A slight correlation between VASH1 expression and VASH2 expression was observed. The low VASH1/low VASH2 group had a better prognosis than the other three groups with different combinations of VASH1 and VASH2 expression levels. The present study showed that high VASH1 expression and high VASH2 expression may be novel independent predictors of a poor prognosis in patients with ESCC and that a slight correlation between VASH1 and VASH2 expression existed. The present findings suggest that combined evaluation of VASH1 and VASH2 expression should provide an improved understanding of their clinicopathological features.

12.
Life Sci ; 148: 1-8, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26860892

ABSTRACT

AIMS: An abnormal ratio of vasohibin-2/vasohibin-1 may be involved in the abnormal angiogenesis and vascular remodeling during pulmonary arterial hypertension (PAH). MAIN METHODS: To evaluate the pharmacological actions of Ponatinib (AP) in experimental model of PAH, the effects of AP on TGF-ß1-mediated endothelial-mesenchymal transition (EndoMT) in human pulmonary microvascular endothelial cells (HPMEC), and the hypoxic human pulmonary artery smooth muscle cells (HPASMC) proliferation and HPMEC in vitro, and on bleomycin (BLM)-induced PAH in vivo were investigated. KEY FINDINGS: AP treatment resulted in a reduction of EndoMT in HPMECs with a decrease of vimentin, whereas an increase of VE-cadherin, reduction of fibroblast growth factor (FGF-2), vascular endothelial growth factor (VEGF) and vasohibin-2 (VASH-2), whereas an increase of vasohibin-1 (VASH-1) in the hypoxic HPMEC, a reduction of the HPASMC proliferation with decreases of wnt5a, ß-catenin and cyclin D1 expression. AP ameliorated BLM-induced PAH in rats with reductions of FGF-2, VEGF, von Willebrand factor (vWF) and VASH-2 expression, whereas an increase of VASH-1 expression. AP ameliorated BLM-induced PAH in rats with reductions of the pathological score and the collagen deposition. In addition, AP ameliorated hemodynamics and right ventricular hypertrophy. SIGNIFICANCES: Our results identified a therapeutic potential of AP in PAH therapy might be modulated VASH-2/VASH-1 and the Wnt signaling.


Subject(s)
Angiogenic Proteins/metabolism , Cell Cycle Proteins/metabolism , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/prevention & control , Imidazoles/therapeutic use , Pyridazines/therapeutic use , Wnt Signaling Pathway/physiology , Angiogenic Proteins/antagonists & inhibitors , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Imidazoles/pharmacology , Male , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridazines/pharmacology , Rats , Rats, Sprague-Dawley , Wnt Signaling Pathway/drug effects
13.
Oncotarget ; 6(10): 7880-98, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25797264

ABSTRACT

Vasohibin-1 (VASH1) is an endogenous angiogenesis inhibitor.However, the clinical relevance of VASH1 in colon cancer and its regulations on cancer angiogenesis and cancer cell biological characteristics are still unknown. Here we showed that stromal VASH1 levels were negatively correlated with tumor size, advanced clinical stage and distant metastases in colon cancer patients. Overexpression of VASH1 in colon cancer cells induced apoptosis and senescence, inhibiting cancer cell growth and colony formation in vitro and tumor growth in vivo. In addition, knockdown of VASH1 in cancer cells promoted cell growth, adhesion and migration in vitro, and enhanced tumorigenesis and metastasis in vivo.


Subject(s)
Cell Cycle Proteins/metabolism , Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Aged , Animals , Apoptosis/physiology , Cell Adhesion/physiology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Movement/physiology , Cellular Senescence/physiology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Nude , Mice, Transgenic , Middle Aged , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL