Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell Commun Signal ; 22(1): 225, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605348

ABSTRACT

The endothelial glycocalyx, located at the luminal surface of the endothelium, plays an important role in the regulation of leukocyte adhesion, vascular permeability, and vascular homeostasis. Endomucin (EMCN), a component of the endothelial glycocalyx, is a mucin-like transmembrane glycoprotein selectively expressed by venous and capillary endothelium. We have previously shown that knockdown of EMCN impairs retinal vascular development in vivo and vascular endothelial growth factor 165 isoform (VEGF165)-induced cell migration, proliferation, and tube formation by human retinal endothelial cells in vitro and that EMCN is essential for VEGF165-stimulated clathrin-mediated endocytosis and signaling of VEGF receptor 2 (VEGFR2). Clathrin-mediated endocytosis is an essential step in receptor signaling and is of paramount importance for a number of receptors for growth factors involved in angiogenesis. In this study, we further investigated the molecular mechanism underlying EMCN's involvement in the regulation of VEGF-induced endocytosis. In addition, we examined the specificity of EMCN's role in angiogenesis-related cell surface receptor tyrosine kinase endocytosis and signaling. We identified that EMCN interacts with AP2 complex, which is essential for clathrin-mediated endocytosis. Lack of EMCN did not affect clathrin recruitment to the AP2 complex following VEGF stimulation, but it is necessary for the interaction between VEGFR2 and the AP2 complex during endocytosis. EMCN does not inhibit VEGFR1 and FGFR1 internalization or their downstream activities since EMCN interacts with VEGFR2 but not VEGFR1 or FGFR1. Additionally, EMCN also regulates VEGF121-induced VEGFR2 phosphorylation and internalization.


Subject(s)
Endothelial Cells , Vascular Endothelial Growth Factor A , Humans , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Sialomucins/metabolism , Endocytosis , Clathrin/metabolism
3.
Food Sci Anim Resour ; 43(6): 1017-1030, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37969322

ABSTRACT

Fetal bovine serum (FBS), which contains various nutrients, comprises 20% of the growth medium for cell-cultivated meat. However, ethical, cost, and scientific issues, necesitates identification of alternatives. In this study, we investigated commercially manufactured serum-free media capable of culturing Hanwoo satellite cells (HWSCs) to identify constituent proliferation enhancing factors. Six different serum-free media were selected, and the HWSC proliferation rates in these serum-free media were compared with that of control medium supplemented with 20% FBS. Among the six media, cell proliferation rates were higher only in StemFlexTM Medium (SF) and Mesenchymal Stem Cell Growth Medium DXF (MS) than in the control medium. SF and MS contain high fibroblast growth factor 2 (FGF2) concentrations, and we found upregulated FGF2 protein expression in cells cultured in SF or MS. Activation of the fibroblast growth factor receptor 1 (FGFR1)-mediated signaling pathway and stimulation of muscle satellite cell proliferation-related factors were confirmed by the presence of related biomarkers (FGFR1, FRS2, Raf1, ERK, p38, Pax7, and MyoD) as indicated by quantitative polymerase chain reaction, western blotting, and immunocytochemistry. Moreover, PD173074, an FGFR1 inhibitor suppressed cell proliferation in SF and MS and downregulated related biomarkers (FGFR1, FRS2, Raf1, and ERK). The promotion of cell proliferation in SF and MS was therefore attributed to FGF2, which indicates that FGFR1 activation in muscle satellite cells may be a target for improving the efficiency of cell-cultivated meat production.

6.
Acta Biochim Biophys Sin (Shanghai) ; 55(11): 1819-1832, 2023 11 25.
Article in English | MEDLINE | ID: mdl-37867436

ABSTRACT

Despite substantial advancements in screening, surgery, and chemotherapy, colorectal cancer remains the second most lethal form of the disease. Nuclear factor kappa B (NF-κB) signaling is a critical driver facilitating the malignant transformation of chronic inflammatory bowel diseases. In this study, deregulated miRNAs that could play a role in colon cancer are analyzed and investigated for specific functions in vitro using cancer cells and in vivo using a subcutaneous xenograft model. miRNA downstream targets are analyzed, and predicted binding and regulation are verified. miR-1262, an antitumor miRNA, is downregulated in colon cancer tissue samples and cell lines. miR-1262 overexpression suppresses colon cancer malignant behaviors in vitro and tumor development and metastasis in a subcutaneous xenograft model and a lung metastasis mouse model in vivo. miR-1262 directly targets fibroblast growth factor receptor 1 (FGFR1) and inhibits FGFR1 expression. FGFR1 overexpression shows oncogenic functions through the regulation of cancer cell proliferation, invasion, and migration; when cotransfected, lv-FGFR1 partially attenuates the antitumor effects of agomir-1262. NF-κB binds to the miR-1262 promoter region and inhibits transcription activity. The NF-κB inhibitor CAPE exerts antitumor effects; miR-1262 inhibition partially reverses CAPE effects on colon cancer cells. Conclusively, miR-1262 serves as an antitumor miRNA in colon cancer by targeting FGFR1. The NF-κB/miR-1262/FGFR1 axis modulates colon cancer cell phenotypes, including proliferation, invasion, and migration.


Subject(s)
Colonic Neoplasms , MicroRNAs , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colonic Neoplasms/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
7.
Mol Divers ; 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37566198

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are a family of cell surface receptors that bind to fibroblast growth factor (FGF) and mediate various cellular functions (translocating proteins, tissue repair, cell proliferation, development, and differentiation) through complex signaling pathways. The FGFR1 growth receptor is essential in the pathogenesis of numerous malignancies, including but not limited to breast cancer, bladder cancer, hepatocellular carcinoma (HCC), and cholangiocarcinoma. The higher levels of FGFR1 expression on the surface of cancer cells cause overly active signaling, which leads to rapid cell proliferation, resulting in a high spread of cancer cells. The kinases that FGFR1 activates migrate across the cell nucleus, activating genes and kinase proteins necessary for the growth and survival of cancerous cells. Therefore, FGFR1 targeting shows therapeutic promise in some diseases, including cancer. Inhibitors of FGFR1s are being developed and studied for their potential to block aberrant FGFR1 signaling and inhibit cancer growth. Since the discovery of new FGFR1 inhibitors in the laboratory is difficult, expensive, time-consuming, and labor-intensive, only a small number of FGFR1 inhibitors have been approved by the FDA for use in the treatment of cancer. To accelerate drug discovery by efficiently exploring the vast chemical space, and identifying potential candidates with higher accuracy and reduced cost, we developed artificial intelligence (AI)-based prediction models for FGFR1 inhibitors using a dataset of 2356 chemical compounds. Four machine learning (ML) algorithms (SVM, RF, k-NN, and ANN) were used to train different prediction models based on molecular descriptors (1D and 2D, with and without molecular fingerprints). Among all trained models, the random forest (RF)-based prediction model achieved the highest accuracy on the training (98.9%), test (89.8%), and external test (90.3%) datasets. The developed inhibitor prediction model (FGFR1Pred) provides a valuable tool for identifying potential FGFR1 inhibitors, expediting the drug discovery process and ultimately facilitating the development of new therapeutics. The model is made available at https://github.com/PGlab-NIPER/FGFR1Pred.git.

9.
Transl Cancer Res ; 11(9): 3222-3234, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36237262

ABSTRACT

Background: Fibroblast growth factor receptor 1 (FGFR1) and fibroblast growth factor receptor 2 (FGFR2) may be of significance in the development of laryngeal squamous cell carcinoma (SCC) tissues. Examination of the expression results of these factors may offer new insights into treatment of the disease, such as genetic and histological targeted target therapy. Methods: We selected tissue from 30 cases of laryngeal SCC, 23 cases of adjacent normal mucosa, and 26 cases of benign laryngeal mucosal tissues from patients who received surgery at the Otolaryngology Department of the Affiliated Hospital of Chengde Medical College between September 2020 and January 2022. The laryngeal cancers included nine cases of supraglottic, 20 glottic (vocal cord), and one case of subglottic cancer, while all benign laryngeal mucosal lesions were obtained from vocal cord polyps. The expression of FGFR1 and FGFR2 was detected in 30 laryngeal cancers, 23 adjacent normal mucosa, and 26 vocal cord polyps by immunohistochemical technology [immunohistochemistry (IHC)], and the correlation analysis of their expression in laryngeal cancer was performed. P<0.05 was represented statistically significant. Results: The expression of FGFR1 and FGFR2 was significantly different in laryngeal SCC and the normal tissue >0.5 cm from the tumor margin (P<0.05), and between laryngeal SCC and vocal polyps (P<0.05). There was no difference in FGFR1 and FGFR2 expression (P>0.05) between normal mucosal margins and vocal cord polyp tissue, and no correlation between FGFR1 and FGFR2 in laryngeal SCC and sex, age, smoking history, alcohol consumption history, tumor diameter, tumor lymph node metastasis, tumor differentiation degree, and Tumor-Node-Metastasis (TNM) stage (P>0.05), A moderate positive correlation between FGFR1 expression and FGFR2 expression in laryngeal SCC was seen (Rs=0.499, P<0.01). Conclusions: FGFR1 and FGFR2 may participate in the occurrence of SCC of the throat: (I) positive FGFR1 and FGFR2 expressions are not associated with gender, age, smoking history, alcohol consumption history, tumor diameter, lymph node metastasis, degree of differentiation, or TNM stage. (II) FGFR2 increases successively with higher FGFR1 expression and with a positive correlation in laryngeal SCC.

10.
Respir Res ; 23(1): 269, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36183124

ABSTRACT

BACKGROUND: The defects and imbalance in lung repair and structural maintenance contribute to the pathogenesis of chronic obstructive pulmonary diseases (COPD), yet the molecular mechanisms that regulate lung repair process are so far incompletely understood. We hypothesized that cigarette smoking causes glycocalyx impairment and endothelial apoptosis in COPD, which could be repaired by the stimulation of fibroblast growth factor 10 (FGF10)/FGF receptor 1 (FGFR1) signaling. METHODS: We used immunostaining (immunohistochemical [IHC] and immunofluorescence [IF]) and enzyme-linked immunosorbent assay (ELISA) to detect the levels of glycocalyx components and endothelial apoptosis in animal models and in patients with COPD. We used the murine emphysema model and in vitro studies to determine the protective and reparative role of FGF10/FGFR1. RESULTS: Exposure to cigarette smoke caused endothelial glycocalyx impairment and emphysematous changes in murine models and human specimens. Pretreatment of FGF10 attenuated the development of emphysema and the shedding of glycocalyx components induced by CSE in vivo. However, FGF10 did not attenuate the emphysema induced by endothelial-specific killing peptide CGSPGWVRC-GG-D(KLAKLAK)2. Mechanistically, FGF10 alleviated smoke-induced endothelial apoptosis and glycocalyx repair through FGFR1/ERK/SOX9/HS6ST1 signaling in vitro. FGF10 was shown to repair pulmonary glycocalyx injury and endothelial apoptosis, and attenuate smoke-induced COPD through FGFR1 signaling. CONCLUSIONS: Our results suggest that FGF10 may serve as a potential therapeutic strategy against COPD via endothelial repair and glycocalyx reconstitution.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Animals , Apoptosis/physiology , Emphysema/complications , Fibroblast Growth Factor 10 , Glycocalyx/metabolism , Glycocalyx/pathology , Humans , Mice , Pulmonary Emphysema/metabolism , Receptors, Fibroblast Growth Factor/therapeutic use , Nicotiana
11.
Clin Pediatr Endocrinol ; 31(3): 172-177, 2022.
Article in English | MEDLINE | ID: mdl-35928375

ABSTRACT

Recent studies have indicated that heterozygous loss-of-function variants in fibroblast growth factor receptor 1 (FGFR1) are involved in the development of congenital hypogonadotropic hypogonadism and combined pituitary hormone deficiency (CPHD). We encountered a Japanese boy with short stature and pubertal failure. Endocrine studies showed GH, TSH, and LH/FSH deficiencies, and brain magnetic resonance imaging delineated hypoplastic anterior pituitary and ectopic posterior pituitary. The patient was treated with GH, l-thyroxine, and hCG/rFSH. Next-generation sequencing panel for pituitary dysfunction identified a probably weak disease-associated heterozygous missense variant in FGFR1 (NM_023110.3:c.176A>T:p.(Asp59Val)), together with a probably non-deleterious heterozygous missense variant in KISS1R (NM_032551.5:c.769G>C:p.(Val257Leu)). We also review six previously reported CHPD patients with probably deleterious FGFR1 variants. The data, in conjunction with the previously reported cases, argue for the relevance of FGFR1 variants to the development of CPHD.

12.
Biochem Biophys Res Commun ; 619: 84-89, 2022 09 03.
Article in English | MEDLINE | ID: mdl-35749940

ABSTRACT

Fibroblast growth factor 21 (FGF21) is regulated by peroxisome proliferator activated receptor α (PPARα) in the liver. FGF21 regulates lipid metabolism via fibroblast growth factor receptor 1 (FGFR1). FGF21 protect against alcoholic fatty liver (AFL), however, FGF21 does not exert protective effect through liver FGFR1. We have previously shown that PPARα agonist WY-14,643 induces FGF21 and adipose atrophy but fails to protect against chronic ethanol-induced AFL in mice lacking adipose FGFR1. In this study we tested the direct role of the FGF21 in regulation of adipose tissue mass and ethanol induced-hepatic triglyceride (TG) accumulation in normal control (fgfr1fl/fl) mice and in adipose FGFR1 knockout mice (fgfr1adipoQ-cre). First, we tested whether WY-14,643 effects on adipose atrophy and AFL can be recapitulated in binge alcohol model. As in chronic model, adipose tissue mass and serum free fatty acid (FFA) were decreased by WY-14,643 in the fgfr1adipoQ-cre mice but not in the fgfr1fl/fl mice. However, in contrast to the chronic model, binge ethanol-induced AFL was blunted by WY-14,643 to a greater extent in the fgfr1adipoQ-cre mice than in the fgfr1fl/fl mice. Similarly, circulating FGF21 was elevated by binge ethanol to a greater extent in the fgfr1adipoQ-cre mice than in the fgfr1fl/fl mice on top of WY-14,643 treatment. Accordingly, we tested the involvement of the FGF21 in adipose atrophy and AFL. Consistent with FGFR1-dependent effects of WY-14,643 on adipose atrophy and AFL, recombinant mouse FGF21 (rFGF21) injection induced adipose atrophy, blunted AFL and serum TG elevation to a greater extent in the fgfr1adipoQ-cre mice than in the fgfr1fl/fl mice. These results indicated the consistency of adipose FGFR1 dependent effect of WY-14,643 and FGF21 in PPARα-mediated regulation of adipose tissue mass and fat mobilization from adipose tissues to the liver, suggesting that adipose tissues crosstalk with liver through an interaction between liver PPARα-FGF21 and adipose FGFR1 to maintain adipose tissue mass.


Subject(s)
Fatty Liver, Alcoholic , PPAR alpha , Adipose Tissue/metabolism , Animals , Atrophy , Ethanol/pharmacology , Fatty Liver, Alcoholic/metabolism , Fibroblast Growth Factors/metabolism , Liver/metabolism , Mice , Mice, Knockout , PPAR alpha/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
13.
Bioengineered ; 13(1): 1530-1540, 2022 01.
Article in English | MEDLINE | ID: mdl-35006038

ABSTRACT

Psoriasis is a common chronic immune-mediated disease that often has a serious negative impact on the physical and mental health of patients. Dihydroartemisinin (DHA) is a drug with anti-fibrotic and anti-inflammatory effects that may be involved in the autoimmune regulation of immune diseases. However, the effects of DHA on psoriasis have not been reported comprehensively. Therefore, the aim of this study was to investigate the effect of DHA on abnormal proliferation and inflammation of epidermal keratinocyte cells in psoriasis and its mechanism of action. IL-17A-induced human epidermal keratin-forming cells (HaCaT) were used as a model. And after induction exposure to different concentrations of DHA, CCK-8, EDU staining, wound healing and Western blotting were performed to assess cell viability, proliferation, migration, differentiation and inflammatory factors, respectively. Subsequently, agonists of fibroblast growth factor receptor 1 (FGFR1) were added and the above experiments were repeated. The results showed that DHA obviously inhibited IL-17A-induced hyperproliferation, migration and expression of inflammatory factors in HaCaT cells. Furthermore, FGFR1 was highly expressed in IL-17A-induced HaCaT cells, and DHA inhibited its expression. However, the inhibitory effect of DHA on IL-17A-induced HaCaT cells was reversed after the addition of FGFR1 agonist. In conclusion, DHA could inhibit IL-17A-induced hyperproliferation and inflammation of keratinocytes by targeting FGFR1, which also provided a new target for the treatment of psoriasis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Artemisinins/pharmacology , Interleukin-17/adverse effects , Keratinocytes/cytology , Psoriasis/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Models, Biological , Psoriasis/drug therapy , Up-Regulation/drug effects
14.
Indian J Pathol Microbiol ; 64(2): 254-260, 2021.
Article in English | MEDLINE | ID: mdl-33851617

ABSTRACT

CONTEXT: Breast cancer is the most common cause of cancer mortality among women worldwide. It is a heterogeneous disease partly responsible for treatment failure in luminal B patients. Deregulation of fibroblast growth factor signaling has been found and its therapeutic/prognostic value is explored. AIMS: Most of the research has studied the FGFR1 gene while our study explored its protein expression by immunohistochemestry and examined the association with clinicopathological features, different molecular subtypes and survival. SUBJECTS AND METHODS: Formalin-fixed and paraffin-embedded samples of invasive breast carcinomas were used to analyze FGFR1 expression. FGFR 1 was scored by percentage and intensity of cell cytoplasm staining, correlations were investigated and survival curves were constructed. STATISTICAL ANALYSIS USED: Chi-square test was used to assess the relationship between the marker expression and the clinicopathological characteristics. Overall specific survival curves were estimated using the Kaplan-Meier method and statistical significance was assessed using the log-rank test. RESULTS: FGFR1 was associated at different staining threshold cut-offs with tumor size (P = 0.002), infiltrating lymph node (P = 0.022), distant metastasis (P = 0.003), positive estrogen receptor (P = 0.000), HER2 overexpression (P = 0.044) and luminal phenotypes (P = 0.026). The results also emphasize FGFR1 correlation expression with distant metastasis in luminal B tumors (P = 0.035) but not with luminal A and with overexpressed HER2 protein in both luminal tumors. FGFR1 expression affect luminal B patients survival with poor outcome. CONCLUSIONS: FGFR1 expression may serve as a prognostic and predictive factor in luminal breast cancers, it can also be considered as a potential therapeutic target in luminal B cases.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Breast Neoplasms/mortality , Cell Proliferation/physiology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Middle Aged , Prognosis , Receptor, Fibroblast Growth Factor, Type 1/genetics
15.
Methods Cell Biol ; 162: 171-203, 2021.
Article in English | MEDLINE | ID: mdl-33707012

ABSTRACT

The application of both fluorescence and electron microscopy results in a powerful combination of imaging modalities called "correlative light and electron microscopy" (CLEM). Whereas conventional transmission electron microscopy (TEM) tomography is only able to image sections up to a thickness of ~300nm, scanning transmission electron microscopy (STEM) tomography at 200kV allows the analysis of sections up to a thickness of 900nm in three dimensions. In the current study we have successfully integrated STEM tomography into CLEM as demonstrated for human retinal pigment epithelial 1 (RPE1) cells expressing various fluorescent fusion proteins which were high-pressure frozen and then embedded in Lowicryl HM20. Fluorescently labeled gold nanoparticles were applied onto resin sections and imaged by fluorescence and electron microscopy. STEM tomograms were recorded at regions of interest, and overlays were generated using the eC-CLEM software package. Through the nuclear staining of living cells, the use of fluorescently labeled gold fiducials for the generation of overlays, and the integration of STEM tomography we have markedly extended the application of the Kukulski protocol (Kukulski et al., 2011, 2012). Various fluorescently tagged proteins localizing to different cellular organelles could be assigned to their ultrastructural compartments. By combining STEM tomography with on-section CLEM, fluorescently tagged proteins can be localized in three-dimensional ultrastructural environments with a volume of at least 2.7×2.7×0.5µm.


Subject(s)
Electron Microscope Tomography , Metal Nanoparticles , Gold , Humans , Microscopy, Electron , Microscopy, Fluorescence
16.
BMC Cancer ; 20(1): 348, 2020 Apr 18.
Article in English | MEDLINE | ID: mdl-32326908

ABSTRACT

BACKGROUND: The gene encoding fibroblast growth factor receptor 1 (FGFR1) is emerging as a therapeutic and prognostic biomarker in various cancer types, including head and neck squamous cell carcinoma (SCC). Here, we investigated the clinicopathologic implication of FGFR1 gene amplification and protein overexpression in hypopharyngeal and laryngeal SCC. METHODS: Fluorescence in situ hybridization and immunohistochemistry were performed to determine FGFR1 gene amplification and protein overexpression in 209 surgically resected cases. RESULTS: FGFR1 amplification observed in 8 (8/66, 12.1%; 6 hypopharynx and 2 larynx) patients and high FGFR1 expression in 21 (21/199, 10.6%) patients significantly correlated with lymph node metastasis and advanced pathological stages. FGFR1 amplification was also associated with worse disease-free survival in multivariate analysis (hazard ratio = 4.527, P = 0.032). High FGFR1 expression was more frequently observed, consistent with the worsening of the degree of histologic differentiation. CONCLUSIONS: FGFR1 amplification may serve as an independent prognostic factor for disease-free survival in hypopharyngeal and laryngeal SCC. Aberrant FGFR signaling caused by FGFR1 gene amplification or protein overexpression may play a crucial role in the malignant evolution and progression of hypopharyngeal and laryngeal SCC, and offer novel therapeutic opportunities in patients with hypopharyngeal and laryngeal SCC that usually lack specific therapeutic targets.


Subject(s)
Biomarkers, Tumor/analysis , Gene Amplification , Hypopharyngeal Neoplasms/pathology , Laryngeal Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Gene Dosage , Humans , Hypopharyngeal Neoplasms/genetics , Hypopharyngeal Neoplasms/metabolism , Hypopharyngeal Neoplasms/surgery , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/surgery , Laryngectomy/mortality , Male , Middle Aged , Pharyngectomy/mortality , Prognosis , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/surgery , Survival Rate
17.
Aging (Albany NY) ; 12(6): 5183-5194, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32202509

ABSTRACT

BACKGROUND: Most recently, long non-coding RNAs (lncRNAs) emerge as crucial modulators in many biological processes, such as embryonic development, cell growth, and tumorigenesis. However, the correlations between lncRNAs and colorectal cancer (CRC) cell proliferation, metastasis, and gemcitabine resistance are not well understood. RESULTS: The expression of AGAP2-AS1 was overexpressed in CRC tissues and negatively correlated with the survival of patients with CRC. AGAP2-AS1 promoted CRC cell proliferation and inhibited apoptosis. Moreover, AGAP2-AS1 enhanced the chemoresistance of CRC cells to gemcitabine. In addition, AGAP2-AS1 enhanced the migration and invasion of CRC cells. Mechanistic studies showed that AGAP2-AS1 regulated fibroblast growth factor receptor 1 (FGFR1) expression by sponging miR-497 in CRC progression. CONCLUSION: We identified an oncogenic role of AGAP2-AS1 in the development and progression of CRC. METHODS: qRT-PCR was used to measure the expression of AGAP2 Antisense RNA 1 (AGAP2-AS1) in 116 cases of CRC and adjacent normal tissues. Luciferase reporter assays was used to detect the interaction between AGAP2-AS1 and miR-497. The xenograft tumor experiment was used to study the in vivo function of AGAP2-AS1.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cell Movement/genetics , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Apoptosis/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Oncogenes , Prognosis , Up-Regulation , Gemcitabine
18.
Ann Transl Med ; 7(22): 669, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31930070

ABSTRACT

BACKGROUND: Previous studies about the prognostic and clinicopathological significance of fibroblast growth factor receptor 1 (FGFR1) amplification in resected esophageal squamous cell carcinoma (ESCC) are controversial. Therefore, the aim of the current meta-analysis was to determine the association of FGFR1 amplification with prognosis and clinicopathological characteristics of resected ESCC patients. METHODS: The PubMed, EMBASE, Web of Science, The Cochrane Library, CNKI, Wanfang, VIP and SinoMed databases were searched systematically from the establishment date of databases to April 1, 2019 to identify related studies. The correlations of FGFR1 amplification of prognosis and clinicopathological characteristics in ESCC were assessed by the combined hazard ratio (HR) with 95% confidence interval (CI) and combined odds ratio (OR) with 95% CI, respectively. All statistical analyses were performed by the Stata 12.0 software. RESULTS: A total of nine retrospective studies involving 2,326 patients who received the surgery were included into the current meta-analysis. The results indicated that FGFR1 amplification was significantly correlated with worse overall survival (OS) (HR =1.50, 95% CI: 1.25-1.81, P<0.001), disease-free survival (DFS) (HR =1.58, 95% CI: 1.27-1.96, P<0.001), lymph node metastasis (OR =1.45, 95% CI: 1.13-1.86, P=0.004), higher TNM stage (OR =1.33, 95% CI: 1.03-1.72, P=0.027) and poorer differentiation (OR =1.10, 95% CI: 1.07-1.13, P<0.001). CONCLUSIONS: The current meta-analysis strongly demonstrates that FGFR1 amplification is an independent prognostic risk factor for resected ESCC patients and more prevalent among patients with advanced tumor stage and poorer differentiation.

19.
Dev Biol ; 425(2): 208-222, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28389228

ABSTRACT

How the fetal-placental arterial connection is made and positioned relative to the embryonic body axis, thereby ensuring efficient and directed blood flow to and from the mother during gestation, is not known. Here we use a combination of genetics, timed pharmacological inhibition in living mouse embryos, and three-dimensional modeling to link two novel architectural features that, at present, have no status in embryological atlases. The allantoic core domain (ACD) is the extraembryonic extension of the primitive streak into the allantois, or pre-umbilical tissue; the vessel of confluence (VOC), situated adjacent to the ACD, is an extraembryonic vessel that marks the site of fetal-placental arterial union. We show that genesis of the fetal-placental connection involves the ACD and VOC in a series of steps, each one dependent upon the last. In the first, Brachyury (T) ensures adequate extension of the primitive streak into the allantois, which in turn designates the allantoic-yolk sac junction. Next, the streak-derived ACD organizes allantoic angioblasts to the axial junction; upon signaling from Fibroblast Growth Factor Receptor-1 (FGFR1), these endothelialize and branch, forming a sprouting VOC that unites the umbilical and omphalomesenteric arteries with the fetal dorsal aortae. Arterial union is followed by the appearance of the medial umbilical roots within the VOC, which in turn designate the correct axial placement of the lateral umbilical roots/common iliac arteries. In addition, we show that the ACD and VOC are conserved across Placentalia, including humans, underscoring their fundamental importance in mammalian biology. We conclude that T is required for correct axial positioning of the VOC via the primitive streak/ACD, while FGFR1, through its role in endothelialization and branching, further patterns it. Together, these genetic, molecular and structural elements safeguard the fetus against adverse outcomes that can result from vascular mispatterning of the fetal-placental arterial connection.


Subject(s)
Arteries/embryology , Fetal Proteins/metabolism , Fetus/embryology , Gastrula/blood supply , Gastrula/metabolism , Morphogenesis , Placenta/embryology , T-Box Domain Proteins/metabolism , Allantois/embryology , Allantois/metabolism , Animals , Arteries/metabolism , Endothelium, Vascular/metabolism , Female , Fetus/metabolism , Gastrula/embryology , Mice , Models, Biological , Placenta/metabolism , Pregnancy , Primitive Streak/embryology , Primitive Streak/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Umbilical Arteries/embryology , Umbilical Arteries/metabolism , Vascular Remodeling , Yolk Sac/metabolism
20.
Cancer ; 122(19): 3024-31, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27315356

ABSTRACT

BACKGROUND: Fibroblast growth factor receptor 1 (FGFR1) amplification is a potential driving oncogene in squamous cell cancer (SCC) of the lung. The current phase 2 study evaluated the efficacy and tolerability of dovitinib, an FGFR inhibitor, in patients with advanced SCC of the lung. METHODS: Patients with pretreated advanced SCC of the lung whose tumors demonstrated FGFR1 amplification of > 5 copies by fluorescence in situ hybridization were enrolled. Dovitinib at a dose of 500 mg was administered orally, once daily, on days 1 to 5 of every week, followed by 2 days off. The primary endpoint was overall response. Secondary endpoints were progression-free survival, overall survival, and toxicity. RESULTS: All 26 patients were men with a median age of 68 years (range, 52-80 years). The majority of patients were ever-smokers. The median duration of dovitinib administration (28 days per cycle) was 2.5 months (range, 0.7-8.6 months). The overall response rate was 11.5% (95% confidence interval [95% CI], 0.8%-23.8%) and the disease control rate was 50% (95% CI, 30.8%-69.2%), with 3 patients achieving partial responses. Response durations for the patients with partial responses were ≥4.5 months, ≥ 5.1 months, and 6.1 months, respectively. After a median follow-up of 15.7 months (range, 1.2-25.6 months), the median overall survival was 5.0 months (95% CI, 3.6-6.4 months) and the median progression-free survival was 2.9 months (95% CI, 1.5-4.3 months). The most common grade 3 or higher adverse events were fatigue (19.2%), anorexia (11.5%), and hyponatremia (11.5%) (event severity was graded based on National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). CONCLUSIONS: Treatment with dovitinib demonstrated modest efficacy in patients with advanced SCC with FGFR1 amplification. Further studies to evaluate other biomarkers correlated with the efficacy of dovitinib in patients with SCC are warranted. Cancer 2016;122:3024-3031. © 2016 American Cancer Society.


Subject(s)
Benzimidazoles/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Gene Amplification , Lung Neoplasms/drug therapy , Quinolones/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/genetics , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Female , Follow-Up Studies , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL