Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.348
Filter
1.
Sci Rep ; 13(1): 19783, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37957249

ABSTRACT

Studies indicate EGFL7 as an important gene in controlling angiogenesis and cancer growth, including in colorectal cancer (CRC). Anti-EGFL7 agents are being explored, yet without promising results. Therefore, the role of EGFL7 in CRC carcinogenesis should be investigated. This study aimed to evaluate the prognostic value of EGFL7 expression in CRC and the signaling pathways influenced by this gene. EGFL7 expression was evaluated through immunohistochemistry in 463 patients diagnosed with CRC and further associated with clinicopathological data, angiogenesis markers and survival. In silico analyzes were performed with colon adenocarcinoma data from The Cancer Genome Atlas. Analysis of enriched gene ontology and pathways were performed using the differentially expressed genes. 77.7% of patients presented low EGFL7 expression, which was associated with higher lymph node spread and invasion of lymphatic vessels, with no impact on survival. Additionally, low EGFL7 expression was associated with high VEGFR2 expression. Finally, we found in silico that EGFL7 expression was associated with cell growth, angiogenesis, and important pathways such as VEGF, Rap-1, MAPK and PI3K/Akt. Expression of EGFL7 in tumor cells may be associated with important pathways that can alter functions related to tumor invasive processes, preventing recurrence and metastatic process.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Colorectal Neoplasms , Lymphatic Vessels , Humans , Phosphatidylinositol 3-Kinases/metabolism , Endothelial Growth Factors/genetics , EGF Family of Proteins/metabolism , Neoplastic Processes , Intercellular Signaling Peptides and Proteins/metabolism , Transcription Factors/metabolism , Lymph Nodes/metabolism , Lymphatic Vessels/metabolism , Colorectal Neoplasms/genetics , Calcium-Binding Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 120(29): e2303740120, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37428914

ABSTRACT

Defining reliable surrogate markers and overcoming drug resistance are the most challenging issues for improving therapeutic outcomes of antiangiogenic drugs (AADs) in cancer patients. At the time of this writing, no biomarkers are clinically available to predict AAD therapeutic benefits and drug resistance. Here, we uncovered a unique mechanism of AAD resistance in epithelial carcinomas with KRAS mutations that targeted angiopoietin 2 (ANG2) to circumvent antivascular endothelial growth factor (anti-VEGF) responses. Mechanistically, KRAS mutations up-regulated the FOXC2 transcription factor that directly elevated ANG2 expression at the transcriptional level. ANG2 bestowed anti-VEGF resistance as an alternative pathway to augment VEGF-independent tumor angiogenesis. Most colorectal and pancreatic cancers with KRAS mutations were intrinsically resistant to monotherapies of anti-VEGF or anti-ANG2 drugs. However, combination therapy with anti-VEGF and anti-ANG2 drugs produced synergistic and potent anticancer effects in KRAS-mutated cancers. Together, these data demonstrate that KRAS mutations in tumors serve as a predictive marker for anti-VEGF resistance and are susceptible to combination therapy with anti-VEGF and anti-ANG2 drugs.


Subject(s)
Carcinoma , Endothelial Growth Factors , Humans , Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Angiopoietin-1/metabolism
3.
Turk J Gastroenterol ; 34(2): 118-127, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36445057

ABSTRACT

BACKGROUND: Rat sarcoma virus mutational status guides first-line treatment in metastatic colorectal cancer. This study was a multi center, multi-country ambispective, observational study in the Middle East and North Africa assessing regional rat sarcoma virus testing practices in newly diagnosed patients. METHODS: The retrospective arm (2011-2014) included adults with metastatic colorectal cancer who had initiated first-line therapy with ≥1 post-baseline visit and survival data. The prospective arm (2014-2019) enrolled newly diagnosed patients with histologically proven metastatic colorectal cancer with ≥1 measurable lesion per Response Evaluation Criteria in Solid Tumors, and tissue availability for biomarker analysis. Data look-back and follow-up were 2 years; the rate of RAS mutation was evaluated. RESULTS: RAS testing was ordered for patients in retrospective (326/417) and prospective (407/500) studies. In the former, testing was typically prescribed after first-line treatment initiation, significantly more in patients with stage IV disease (P < .005), resulting in the addition of targeted therapy (41.8% anti-epidermal growth factor receptor, 30.2% anti-vascular endothelial growth factor) in wild-type metastatic colorectal cancer, and significantly impacted the treatment of left-sided tumors (P = .037). In the latter, 58.4% were RAS wild-type; 41.6% were RAS mutant. Non-prescription of RAS testing was attributed to test unavailability, financial, or medical rea sons; predictors of testing prescription were older age, primary tumor in ascending colon, and high tumor grade. RAS status knowledge resulted in the addition of anti-vascular endothelial growth factor (20.4%) or anti-epidermal growth factor receptor therapy (21.2%). CONCLUSION: Before 2014, RAS testing in patients with colorectal cancer in the Middle East and North Africa was often performed after first-line treatment. Testing is more routine in newly diagnosed patients, potentially shifting early treatment patterns.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Rectal Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms/genetics , Endothelial Growth Factors/genetics , Endothelial Growth Factors/therapeutic use , Mutation , Prospective Studies , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Growth Factor/genetics , Receptors, Growth Factor/therapeutic use , Retrospective Studies , Registries
4.
Medicine (Baltimore) ; 101(38): e30595, 2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36197222

ABSTRACT

Sorsby fundus dystrophy (SFD) is a rare autosomal dominant disorder with complete penetrance affecting the macula. This is caused by a mutation in the TIMP-3. This objective narrative review aims to provide an overview of the pathophysiology, current treatment modalities, and future perspectives. A literature search was performed using "PubMed," "Web of Science," "Scopus," "ScienceDirect," "Google Scholar," "medRxiv," and "bioRxiv." The molecular mechanisms underlying SFD are not completely understood. Novel advancements in cell culture techniques, including induced pluripotent stem cells, may enable more reliable modeling of SFD. These cell culture techniques aim to shed more light on the pathophysiology of SFD, and hopefully, this may lead to the future development of treatment strategies for SFD. Currently, no gene therapy is available. The main treatment is the use of anti-vascular endothelial growth factors (anti-VEGF) to treat secondary choroidal neovascular membrane (CNV), which is a major complication observed in this condition. If CNV is detected and treated promptly, patients with SFD have a good chance of maintaining a functional central vision. Other treatment modalities have been tried but have shown limited benefit, and therefore, have not managed to be more widely accepted. In summary, although there is no definitive cure yet, the use of anti-VEGF treatment for secondary CNV has provided the opportunity to maintain functional vision in individuals with SFD, provided CNV is detected and treated early.


Subject(s)
Macular Degeneration , Tissue Inhibitor of Metalloproteinase-3 , Endothelial Growth Factors/genetics , Humans , Macular Degeneration/genetics , Macular Degeneration/therapy , Mutation , Tissue Inhibitor of Metalloproteinase-3/genetics
5.
Cell Death Dis ; 13(10): 910, 2022 10 29.
Article in English | MEDLINE | ID: mdl-36309484

ABSTRACT

Accumulating evidence supports evolutionary trait of drug resistance. Like resilience in other systems, most tumor cells experience drug-tolerant state before full resistance acquired. However, the underlying mechanism is still poorly understood. Here, we identify that EGF like domain multiple 7 (EGFL7) is a responsive gene to epidermal growth factor receptor (EGFR) kinase inhibition during a period when tumors are decimated. Moreover, our data reveal that the adaptive increase of EGFL7 during this process is controlled by the depression of nonsense-mediated mRNA decay (NMD) pathway. Upregulation of EGFL7 activates NOTCH signaling in lung cancer cells, which slows down the decrease of c-Myc caused by EGFR inhibition, thereby helping the survival of cancer cells. Our data, taken together, demonstrate that EGFL7 is a driver gene for resistance to EGFR kinase inhibition, and suggest that targeting EGFL7/NOTCH signaling may improve the clinical benefits of EGFR inhibitors in patients with EGFR mutant tumors.


Subject(s)
Endothelial Growth Factors , Lung Neoplasms , Humans , EGF Family of Proteins/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Calcium-Binding Proteins , ErbB Receptors/metabolism , Transcription Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor
6.
Genet Test Mol Biomarkers ; 25(5): 334-345, 2021 May.
Article in English | MEDLINE | ID: mdl-33970702

ABSTRACT

Background: Vascular endothelial growth factors (VEGFs) are important for glioblastoma multiforme (GBM) growth and development. However, the effects of VEGF-targeting drugs in primary GBM remain poorly understood. Aim: We aimed to explore the key genes correlated with VEGF expression and prognosis and elucidate their potential implications in GBM anti-VEGF therapy. Materials and Methods: RNA-seq data with the corresponding clinicopathological information was retrieved from The Cancer Genome Atlas and the Chinese Glioma Genome Atlas. Weighted gene coexpression network analyses was performed on differentially expressed genes to construct coexpression modules and investigate their correlation with VEGFs. Functional enrichment analyses were performed based on the coexpressed genes from the most promising modules. CytoHubba and Kaplan-Meier analyses were implemented to identify the key genes in the modules of interest. The oncomine database, quantitative reverse transcription PCR, and the Human Protein Atlas were used to investigate the expression characteristics of the identified key genes. Results: Four modules (cyan, green, purple, and tan) correlated significantly with VEGF expression. Enrichment analyses suggested that extracellular matrix-receptor interaction, growth factor binding, and the PI3K-Akt pathways were involved in VEGF expression. Four hub genes (COL6A1, SNRPG, COL3A1, and AHI1) associated with VEGF were identified. Among them, COL6A1 was regarded as the key gene associated with anti-VEGF therapy. Further, COL6A1 was upregulated in GBM compared to that in normal brain tissues. COL6A1 overexpression was associated with a poor prognosis. Conclusion: COL6A1 was identified as the key gene associated with anti-VEGF therapy and may provide novel insight into GBM targeted therapy.


Subject(s)
Collagen Type VI/metabolism , Glioblastoma/genetics , Vascular Endothelial Growth Factors/antagonists & inhibitors , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , China , Collagen Type VI/genetics , Databases, Genetic , Endothelial Growth Factors/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks , Glioblastoma/metabolism , Glioma/genetics , Humans , Kaplan-Meier Estimate , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Protein Interaction Maps , Transcriptome/genetics , Vascular Endothelial Growth Factors/drug effects , Vascular Endothelial Growth Factors/metabolism , snRNP Core Proteins/genetics
7.
J Vis Exp ; (168)2021 02 26.
Article in English | MEDLINE | ID: mdl-33720132

ABSTRACT

In response to specific external cues and the activation of certain transcription factors, endothelial cells can differentiate into a mesenchymal-like phenotype, a process that is termed endothelial to mesenchymal transition (EndMT). Emerging results have suggested that EndMT is causally linked to multiple human diseases, such as fibrosis and cancer. In addition, endothelial-derived mesenchymal cells may be applied in tissue regeneration procedures, as they can be further differentiated into various cell types (e.g., osteoblasts and chondrocytes). Thus, the selective manipulation of EndMT may have clinical potential. Like epithelial-mesenchymal transition (EMT), EndMT can be strongly induced by the secreted cytokine transforming growth factor-beta (TGF-ß), which stimulates the expression of so-called EndMT transcription factors (EndMT-TFs), including Snail and Slug. These EndMT-TFs then up- and downregulate the levels of mesenchymal and endothelial proteins, respectively. Here, we describe methods to investigate TGF-ß-induced EndMT in vitro, including a protocol to study the role of particular TFs in TGF-ß-induced EndMT. Using these techniques, we provide evidence that TGF-ß2 stimulates EndMT in murine pancreatic microvascular endothelial cells (MS-1 cells), and that the genetic depletion of Snail using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated gene editing, abrogates this phenomenon. This approach may serve as a model to interrogate potential modulators of endothelial biology, and can be used to perform genetic or pharmacological screens in order to identify novel regulators of EndMT, with potential application in human disease.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Endothelial Cells/metabolism , Gene Editing , Mesoderm/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Fluorescent Antibody Technique , Mice , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/genetics
8.
J Assist Reprod Genet ; 37(11): 2883-2892, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32945993

ABSTRACT

PURPOSE: To investigate if rare gene variants in women with severe ovarian hyperstimulation syndrome (OHSS) provide clues to the mechanisms involved in the syndrome. METHODS: Among participants in a prospective randomized study (Toftager et al. 2016), six women with predicted low and six women with predicted high risk of OHSS developing severe OHSS (grades 4 and 5, Golan classification) were selected. In the same cohort, six plus six matched controls developing no signs of OHSS (Golan grade 0) were selected. Whole-exome sequencing was performed. Analysis using a predefined in silico OHSS gene panel, variant filtering, and pathway analyses was done. RESULTS: We found no convincing monogenetic association with the development of OHSS using the in silico gene panel. Pathway analysis of OHSS variant lists showed substantial overlap in highly enriched top pathways (p value range p < 0.0001 and p > 9.8E-17) between the low- and high-risk group developing severe OHSS, i.e., "the integrin-linked kinase (ILK) signaling pathway" and the "axonal guidance signaling pathway," both being connected to vasoactive endothelial growth factor (VEGF) and endothelial function. CONCLUSION: Rare variants in OHSS cases with two distinct risk profiles enrich the same signaling pathways linked to VEGF and endothelial function. Clarification of the mechanism as well as potentially defining genetic predisposition of the high vascular permeability is important for future targeted treatment and prevention of OHSS; the potential roles of ILK signaling and the axonal guidance signaling need to be validated by functional studies.


Subject(s)
Fertilization in Vitro , Ovarian Hyperstimulation Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Vascular Endothelial Growth Factor A/genetics , Adult , Chorionic Gonadotropin/genetics , Cohort Studies , Endothelial Growth Factors/genetics , Female , Humans , Ovarian Hyperstimulation Syndrome/pathology , Prospective Studies , Signal Transduction/genetics , Exome Sequencing
9.
Int J Mol Sci ; 20(22)2019 Nov 10.
Article in English | MEDLINE | ID: mdl-31717665

ABSTRACT

It is well known that Prospero homeobox 1 (PROX1) is a crucial regulator of lymphangiogenesis, that reprograms blood endothelial cells to lymphatic phenotype. However, the role of PROX1 in tumor progression, especially in angiogenesis remains controversial. Herein, we studied the role of PROX1 in angiogenesis in cell lines derived from follicular thyroid cancer (FTC: FTC-133) and squamous cell carcinoma of the thyroid gland (SCT: CGTH-W-1) upon PROX1 knockdown. The genes involved in angiogenesis were selected by RNA-seq, and the impact of PROX1 on vascularization potential was investigated using human umbilical vein endothelial cells (HUVECs) cultured in conditioned medium collected from FTC- or SCT-derived cancer cell lines after PROX1 silencing. The angiogenic phenotype was examined in connection with the analysis of focal adhesion and correlated with fibroblast growth factor 2 (FGF2) levels. Additionally, the expression of selected genes involved in angiogenesis was detected in human FTC tissues. As a result, we demonstrated that PROX1 knockdown resulted in upregulation of factors associated with vascularization, such as metalloproteinases (MMP1 and 3), FGF2, vascular endothelial growth factors C (VEGFC), BAI1 associated protein 2 (BAIAP2), nudix hydrolase 6 (NUDT6), angiopoietin 1 (ANGPT1), and vascular endothelial growth factor receptor 2 (KDR). The observed molecular changes resulted in the enhanced formation of capillary-like structures by HUVECs and upregulated focal adhesion in FTC-133 and CGTH-W-1 cells. The signature of selected angiogenic genes' expression in a series of FTC specimens varied depending on the case. Interestingly, PROX1 and FGF2 showed opposing expression levels in FTC tissues and seven thyroid tumor-derived cell lines. In summary, our data revealed that PROX1 is involved in the spreading of thyroid cancer cells by regulation of angiogenesis.


Subject(s)
Adenocarcinoma, Follicular/pathology , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Neovascularization, Pathologic/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Adenocarcinoma, Follicular/blood supply , Adenocarcinoma, Follicular/metabolism , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Cells, Cultured , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Neovascularization, Pathologic/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/blood supply , Thyroid Neoplasms/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
10.
Medicina (Kaunas) ; 55(8)2019 08 11.
Article in English | MEDLINE | ID: mdl-31405227

ABSTRACT

BACKGROUND AND OBJECTIVES: Four single nucleotide polymorphisms (SNPs); rs6921438 and rs4416670 in LOC100132354-C6orf223, rs6993770 in ZFPM2, and rs10738760 in VLDLR-KCNV2 were reported to explain up to 50% of the heritability of vascular endothelial growth factor circulating levels. These SNPs were also studied for possible associations with circulating lipid levels in supposedly healthy European individuals and in a limited number of Iranian individuals with metabolic syndrome. To go further, the association of those four SNPs with plasma lipid parameters, hypercholesterolemia and metabolic syndrome (MetS) was assessed. MATERIALS AND METHODS: A cross-sectional study was conducted on 460 individuals chosen from the general population. Demographic and clinical data were collected and DNA was extracted and genotyped using Kompetitive allele specific PCR (KASP™). A meta-analysis followed, combining our participants with the Iranian individuals (n = 336). RESULTS: Whereas rs10738760 was associated with total cholesterol (Tchol) (p = 0.01), rs6993770 showed significant associations with both Tchol and low-density lipoprotein cholesterol (LDL-C) levels (p = 0.007 and p = 0.01 respectively). Using a multivariate logistic regression model adjusted for different confounding factors, we found that rs6993770 was associated with hypercholesterolemia, specifically high Tchol (p = 0.01) and LDL-C levels (p = 0.01). Furthermore, rs10738760 was positively associated with the risk of MetS in these individuals (p = 0.02) and in the meta-analysis (OR = 1.67, p = 0.01). CONCLUSION: Our results suggest that whereas rs6993770 in ZFPM2 was positively associated with hypercholesterolemia, rs10738760 (VLDLR-KCNV2) has a possible implication in MetS in two Middle Eastern populations.


Subject(s)
Endothelial Growth Factors/genetics , Hypercholesterolemia/genetics , Metabolic Syndrome/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, LDL/analysis , Adult , Cross-Sectional Studies , Female , Humans , Iran , Male , Middle Aged , Receptors, LDL/blood
11.
J Cell Physiol ; 234(6): 8221-8232, 2019 06.
Article in English | MEDLINE | ID: mdl-30317627

ABSTRACT

Aberrant blood vessel functioning and systemic circulation are key causes for vascular disorders; cardiovascular, cerebrovascular, renal artery stenosis, and peripheral artery diseases. Epidemiological and basic science evidence supported genetic reasons, compounded by obesity, hypercholesterolemia, hypertension, diabetes, and smoking as risk factors. This is an umbrella review of risk factors and therapies in vascular disorders, exploring systematic reviews and meta-analyses studies in PubMed, Cochrane, Embase, and Central published in January 2000-May 2018. We made qualitative eligibility gradation of the articles based on inclusion criteria, and independently extracted descriptive and methodologic data to compile their outcomes. We considered 95% confidence interval and the between-study heterogeneity, designated by I 2 . Overall, we extracted 217 studies of impressive quality and at low risk of bias, including 124, 30, 23, 32, and 8, respectively, for the search terms "cardiovascular," "renal," "cerebral," and "limb ischemia" each in combination with "risk factors" and "therapeutics." Our search on genome-wide analyses revealed genes associated with HDL-cholesterol, matrix metalloproteases, angiogenesis, notch3, renin-angiotensin, apolipoprotein E, insulin, and cytokine levels as critical participants in the pathogenesis of vascular diseases. Hypertension and endothelial growth factor-linked polymorphisms were found to contribute to vascular damage. The studies reinforced that lifestyle and dietary patterns influenced susceptibility of circulatory system diseases. Additionally, endovascular medicines, surgical vascularization, angioplasty, and renal artery stenting appeared as major therapeutic approaches in vascular patients. Altogether, our review offers up-to-date information on pathophysiology of vascular diseases and provides insight into existing research, clinical management and clinical gaps in the field.


Subject(s)
Cardiovascular Diseases/genetics , Diabetes Complications/genetics , Hypertension/genetics , Obesity/genetics , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Diabetes Complications/epidemiology , Endothelial Growth Factors/genetics , Genome-Wide Association Study , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/epidemiology , Hypercholesterolemia/genetics , Hypertension/complications , Hypertension/epidemiology , Life Style , Obesity/complications , Obesity/epidemiology , Risk Factors
12.
Neoplasma ; 66(2): 187-196, 2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30569717

ABSTRACT

Melanoma is the main cause of death in patients with skin cancer. While the pathogenesis of cutaneous melanoma is poorly understood, increasing evidence shows that epidermal growth factor (EGF) may be involved. Herein, we tested the hypothesis that down-regulation of EGFL7 inhibits development and progression of human cutaneous melanoma (CM). Initially, we performed immunohistochemical analysis of EGFL7 in 130 specimens and the findings indicated that EGFL7 was highly expressed in CM. The expressions of EGFL7 and Notch signaling pathway-related genes in CM were then measured by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot assay. In order to assess biological functions of EGFL7 in CM we up-regulated or down-regulated endogenous EGFL7 using EGFL7-OE or shRNA against EGFL7 in the A375 CM cell line. To better understand the pivotal role of Notch signaling pathway in CM, we blocked this pathway in A375 cells by inhibitor treatment. Finally, tumor xenograft in nude mice was performed to test the in vivo tumorigenesis of the transfected A375 cells. While EGFL7 activated the Notch signaling pathway in CM, gain- and loss-of-function studies established that decreased EGFL7 inhibited cell proliferation and promoted apoptosis in A375 cells. Moreover, down-regulated EGFL7 suppressed in vivo tumorigenesis. Most importantly, we determined that down-regulating EGFL7 inhibited CM development by suppressing the Notch signaling pathway. The combined findings define potential roles of decreased EGFL7 as inhibitors of CM development by suppressing the Notch signaling pathway, and EGFL7 may therefore be a novel therapeutic target in cutaneous melanoma patients.


Subject(s)
Endothelial Growth Factors/genetics , Gene Silencing , Melanoma/genetics , Receptors, Notch/metabolism , Signal Transduction , Skin Neoplasms/pathology , Animals , Apoptosis , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation , EGF Family of Proteins , Humans , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Proteins , Skin Neoplasms/genetics
13.
J Cell Biochem ; 119(12): 10327-10337, 2018 12.
Article in English | MEDLINE | ID: mdl-30129142

ABSTRACT

Epidermal growth factor-like domain multiple 7 (EGFL7) is an important sport stimulating factor and motility related factors significantly enhanced the tumor cell metastasis and overexpressed in many cancers, including hepatocellular carcinoma (HCC), associated with tumorigenesis. However, the molecular mechanism by which EGFL7 regulates HCC cell proliferation and apoptosis and the correlation between EGFL7 and cyclin-dependent kinases regulatory subunit 2 (CKS2), which is essential for biological function, have not fully explained. In this study, EGFL7 and CKS2 expression in patients with HCC was measured by real-time polymerase chain reaction and immunohistochemistry. After HCC cells respectively transfected with pLKO.1-EGFL7-shRNA, pLVX-Puro-EGFL7 recombined vector or CKS2 small interfering RNA, cell counting kit-8 and flow cytometry was performed to examine the cell proliferation and apoptosis, respectively, and the expression of ß-catenin, CKS2, CDK2, and cleaved caspase-3 was measured by Western blot analysis. We found that EGFL7 and CKS2 were overexpressed in HCC tissues and a positive correlation was found between them. EGFL7 knockdown markedly inhibited proliferation and promoted apoptosis of HCC cells, along with decreased expression of CKS2 and CDK2, but increased cleaved caspase-3 expression, while EGFL7 overexpression showed an opposite effect. EGFL7 silencing in nude mice also showed decreased tumor growth and altered protein expression similar to its effect in HCC cells in vitro. Importantly, CKS2 silencing significantly inhibited EGFL7-induced HCC cell proliferation and protein expression, and Wnt/ß-catenin signaling pathway inhibitor IWR-1-endo significantly inhibited CKS2 expression in HCC cells. Taken together, EGFL7 promotes HCC cell proliferation and inhibits cell apoptosis through increasing CKS2 expression by activating Wnt/ß-catenin signaling.


Subject(s)
CDC2-CDC28 Kinases/genetics , Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Endothelial Growth Factors/genetics , Liver Neoplasms/genetics , Apoptosis/genetics , Calcium-Binding Proteins , Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , EGF Family of Proteins , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Wnt Signaling Pathway/genetics
14.
J Cancer Res Ther ; 14(4): 867-872, 2018.
Article in English | MEDLINE | ID: mdl-29970668

ABSTRACT

OBJECTIVE: Epidermal growth factor-like domain-containing protein 7 (EGFL7) is an endothelial cell-derived secreted factor that regulates vascular tube formation. In human cancer, the specificity of expression is lost as EGFL7 has been detected in tumor cells, in addition to endothelial cells. This study evaluated the intricate relationship between hypoxia-inducible factor 1-alpha (HIF-1α) and EGFL7 under both hyperoxia and hypoxia states. MATERIALS AND METHODS: In the present study, immunohistochemical staining and ELISA were applied to examine the relative level of EGFL7 in 182 cases of hepatocellular carcinoma (HCC) formalin-fixed and paraffin-embedded tissues and 110 cases of HCC serum samples. Quantitative polymerase chain reaction and Western blotting were applied to verify the correlation between serum EGFL7 level and anoxic microenvironment. Immunohistochemical staining was performed to determine the correlation between EGFL7 and HIF1-α. RESULTS: The correlations between EGFL7 expression and patients' age, tumor size, gender, N-stage, history of cirrhosis, M-stage, history of hepatitis C, and history of hepatitis B were statistically insignificant (P = 0.28, 0.34, 0.71, 0.15, 0.8, 0.2, 0.052, and 0.14, respectively). High level of EGFL7 was significantly correlated with overall survival as well as disease-free survival in 182 HCC patients (P = 0.0016 and P < 0.001, respectively). The correlations between serum EGFL7 and vascular invasion and extrahepatic metastasis were statistically significant (P < 0.0001). Among the 35 HIF1-α-positive HCC patients, 69% were medium positive and 31% were strong positive. EGFL7 protein expression level was oxygen dependent in HCC line (P < 0.05). CONCLUSIONS: EGFL7 was found to be a potential predictor for HCC survival and metastasis state; EGFL7 may be a promising biomarker and therapeutic target in human HCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Endothelial Growth Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Adult , Aged , Calcium-Binding Proteins , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , EGF Family of Proteins , Endothelial Growth Factors/genetics , Female , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Oxygen/metabolism , Prognosis
15.
J Cell Physiol ; 233(11): 8526-8537, 2018 11.
Article in English | MEDLINE | ID: mdl-29923200

ABSTRACT

Epidermal growth factor-like domain-containing protein 7 (EGFL7), a member of the epidermal growth factor (EGF)-like protein family, is a potent angiogenic factor expressed in many different cell types. EGFL7 plays a vital role in controlling vascular angiogenesis during embryogenesis, organogenesis, and maintaining skeletal homeostasis. It regulates cellular functions by mediating the main signaling pathways (Notch, integrin) and EGF receptor cascades. Accumulating evidence suggests that Egfl7 plays a crucial role in cancer biology by modulating tumor angiogenesis, metastasis, and invasion. Dysregulation of Egfl7 has been frequently found in several types of cancers, such as malignant glioma, colorectal carcinoma, oral and oesophageal cancers, gastric cancer, hepatocellular carcinoma, pancreatic cancer, breast cancer, lung cancer, osteosarcoma, and acute myeloid leukemia. In addition, altered expression of miR-126, a microRNA associated with Egfl7, was found to play an important role in oncogenesis. More recently, our study has shown that EGFL7 is expressed in both the osteoclast and osteoblast lineages and promotes endothelial cell activities via extracellular signal-regulated kinase (ERK), signal transducer and activator of transcription 3 (STAT3), and integrin signaling cascades, indicative of its angiogenic regulation in the bone microenvironment. Thus, understanding the role of EGFL7 may provide novel insights into the development of improved diagnostics and therapeutic treatment for cancers and skeletal pathological disorders, such as ischemic osteonecrosis and bone fracture healing.


Subject(s)
Endothelial Growth Factors/genetics , MicroRNAs/genetics , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Calcium-Binding Proteins , Cell Lineage/genetics , Cell Movement/genetics , EGF Family of Proteins , Fractures, Bone/genetics , Fractures, Bone/pathology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/classification , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Osteonecrosis/genetics , Osteonecrosis/pathology , Signal Transduction
16.
Sci China Life Sci ; 61(8): 893-901, 2018 08.
Article in English | MEDLINE | ID: mdl-29951953

ABSTRACT

Currently, the primary therapeutic strategy for most growth hormone-producing pituitary adenomas (GHPA) is surgery. Due to the invasiveness of GHPA, high recurrence has limited the benefit of complete adenoma removal surgery. Epidermal growth factor-like domain 7 (EGFL7) is a secreted factor implicated in tumor angiogenesis, growth, invasiveness and metastasis in GHPA. Herein, we observed that the expression level of EGFL7 and p-EGFR in invasive GHPA was much higher than that of non-invasive GHPA. The overexpression of EGFL7 was positively correlated with activation of EGFR (p-EGFR). Noticeably, EGFL7 knockdown significantly inhibited activation of EGFR signaling cascades, including p-ERGR, p-AKT and p-ERK. Further studies showed that EGFL7 knockdown or pharmacological inhibition of EGFR-pathway, using EGFR inhibitor Tyrphostin AG-1478, significantly suppressed migration and invasion of GH3 and GT1-1 cells. In summary, our findings suggest that EGFL7 is a key factor for regulation of EGFR signaling pathway and plays an important role in migration and invasion of invasive GHPA.


Subject(s)
Adenoma/metabolism , Cell Movement , Endothelial Growth Factors/metabolism , ErbB Receptors/metabolism , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Adenoma/genetics , Adenoma/pathology , Animals , Calcium-Binding Proteins , Cell Line, Tumor , EGF Family of Proteins , Endothelial Growth Factors/genetics , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Growth Hormone-Secreting Pituitary Adenoma/genetics , Growth Hormone-Secreting Pituitary Adenoma/pathology , Humans , Mice , Neoplasm Invasiveness , Quinazolines/pharmacology , RNA Interference , Rats , Signal Transduction/drug effects , Tyrphostins/pharmacology
17.
Cancer Genet ; 224-225: 37-40, 2018 08.
Article in English | MEDLINE | ID: mdl-29778234

ABSTRACT

DNA methylation is one of the epigenetic mechanisms associated with gene expression and plays a key role as in activation and deactivation of oncogenes and tumor suppressor genes, respectively. This study employed DNA methylation array to identify methylated genes which are highly correlated with various phenotypes of epithelial ovarian cancer (EOC) in Thai patients and to quantify promoter CpG-island methylation of candidate genes. Tissues from patients with serous and non-serous EOC showed significantly higher promoter methylation of EGFL7 and RASSF1 compared to benign cases. These results indicate the potential of investigating promoter CpG-island methylation of cancer-associated genes as biomarkers of disease progression and even possibly of early detection.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , DNA Methylation/genetics , Endothelial Growth Factors/genetics , Tumor Suppressor Proteins/genetics , Calcium-Binding Proteins , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , EGF Family of Proteins , Female , Humans , Microarray Analysis
18.
Int J Mol Sci ; 19(3)2018 Feb 28.
Article in English | MEDLINE | ID: mdl-29495622

ABSTRACT

Minoxidil directly promotes hair growth via the stimulation of dermal papilla (DP) and epithelial cells. Alternatively, there is little evidence for indirect promotion of hair growth via stimulation of adipose-derived stem cells (ASCs). We investigated whether minoxidil stimulates ASCs and if increased growth factor secretion by ASCs facilitates minoxidil-induced hair growth. Telogen-to-anagen induction was examined in mice. Cultured DP cells and vibrissae hair follicle organ cultures were used to further examine the underlying mechanisms. Subcutaneous injection of minoxidil-treated ASCs accelerated telogen-to-anagen transition in mice, and increased hair weight at day 14 post-injection. Minoxidil did not alter ASC proliferation, but increased migration and tube formation. Minoxidil also increased the secretion of growth factors from ASCs, including chemokine (C-X-C motif) ligand 1 (CXCL1), platelet-derived endothelial cell growth factor (PD-ECGF), and platelet-derived growth factor-C (PDGF-C). Minoxidil increased extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation, and concomitant upregulation of PD-ECGF and PDGF-C mRNA levels were attenuated by an ERK inhibitor. Subcutaneous injection of CXCL1, PD-ECGF, or PDGF-C enhanced anagen induction in mice, and both CXCL1 and PDGF-C increased hair length in ex vivo organ culture. Treatment with CXCL1, PD-ECGF, or PDGF-C also increased the proliferation index in DP cells. Finally, topical application of CXCL1, PD-ECGF, or PDGF-C with 2% minoxidil enhanced anagen induction when compared to minoxidil alone. Minoxidil stimulates ASC motility and increases paracrine growth factor signaling. Minoxidil-stimulated secretion of growth factors by ASCs may enhance hair growth by promoting DP proliferation. Therefore, minoxidil can be used as an ASC preconditioning agent for hair regeneration.


Subject(s)
Adipose Tissue/cytology , Hair Follicle/drug effects , Intercellular Signaling Peptides and Proteins/biosynthesis , Minoxidil/pharmacology , Stem Cells/cytology , Stem Cells/metabolism , Biomarkers , Cell Proliferation/drug effects , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Hair/growth & development , Humans , Lymphokines/genetics , Lymphokines/metabolism , MAP Kinase Signaling System , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism
19.
Nat Commun ; 9(1): 819, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483510

ABSTRACT

Extracellular matrix (ECM) proteins secreted by blood-brain barrier (BBB) endothelial cells (ECs) are implicated in cell trafficking. We discovered that the expression of ECM epidermal growth factor-like protein 7 (EGFL7) is increased in the CNS vasculature of patients with multiple sclerosis (MS), and in mice with experimental autoimmune encephalomyelitis (EAE). Perivascular CD4 T lymphocytes colocalize with ECM-bound EGFL7 in MS lesions. Human and mouse activated T cells upregulate EGFL7 ligand αvß3 integrin and can adhere to EGFL7 through integrin αvß3. EGFL7-knockout (KO) mice show earlier onset of EAE and increased brain and spinal cord parenchymal infiltration of T lymphocytes. Importantly, EC-restricted EGFL7-KO is associated with a similar EAE worsening. Finally, treatment with recombinant EGFL7 improves EAE, reduces MCAM expression, and tightens the BBB in mouse. Our data demonstrate that EGFL7 can limit CNS immune infiltration and may represent a novel therapeutic avenue in MS.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Endothelial Growth Factors/genetics , Spinal Cord/drug effects , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , CD146 Antigen/genetics , CD146 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Calcium-Binding Proteins , EGF Family of Proteins , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Growth Factors/deficiency , Endothelial Growth Factors/immunology , Endothelial Growth Factors/pharmacology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Female , Gene Expression Regulation , Humans , Integrin alpha5/genetics , Integrin alpha5/immunology , Integrin beta3/genetics , Integrin beta3/immunology , Lymphocyte Activation , Male , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spinal Cord/immunology
20.
Stem Cell Rev Rep ; 14(1): 82-91, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28980146

ABSTRACT

Egfl7 (VE-statin) is a secreted protein mostly specific to the endothelial lineage during development and in the adult and which expression is enhanced during angiogenesis. Egfl7 involvement in human postnatal vasculogenesis remains unresolved yet. Our aim was to assess Egfl7 expression in several angiogenic cell types originating from human bone marrow, peripheral blood, or cord blood. We found that only endothelial colony forming cells (ECFC), which are currently considered as the genuine endothelial precursor cells, expressed large amounts of Egfl7. In order to assess its potential roles in ECFC, Egfl7 was repressed in ECFC by RNA interference and ECFC angiogenic capacities were tested in vitro and in vivo. Cell proliferation, differentiation, and migration were significantly improved when Egfl7 was repressed in ECFC in vitro, whereas miR-126-3p levels remained unchanged. In vivo, repression of Egfl7 in ECFC significantly improved post-ischemic revascularization in a model of mouse hind-limb ischemia. In conclusion, ECFC are the sole postnatal angiogenic cells which express large amounts of Egfl7 and whose angiogenic properties are repressed by this factor. Thus, Egfl7 inhibition may be considered as a therapeutic option to improve ECFC-mediated postnatal vasculogenesis and to optimize in vitro ECFC expansion in order to develop an optimized cell therapy approach.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelial Progenitor Cells/cytology , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Endothelial Growth Factors/genetics , Endothelial Progenitor Cells/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Physiologic/physiology , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...