Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Asian Pac J Cancer Prev ; 24(4): 1331-1341, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37116156

ABSTRACT

BACKGROUND: Fibroblast growth factor receptors are growth factor receptor tyrosine kinases, exerting their roles in embryogenesis, tissue homeostasis, and development of cancer. However, little is known about the expression and function of FGFRL1 in esophageal cancer (EC). METHODS: We systematically evaluated the expression of FGFRL1 in TCGA and GETex datasets followed by expression analysis in EC cell lines and clinical specimens using immunofluorescence (IF) and immunohistochemistry (IHC) respectively. RESULTS: GEPIA analysis on TCGA and GETex datasets identified significant upregulation of FGFRL1 in EC patients (n=182) compared to normal controls (n=286, p<0.05). IHC analysis showed significantly higher FGFRL1 expression in EC tissues as compared to the distant matched non-malignant tissues (p<0.001).  Immunoflourescence in EC cells suggested increased expression of FGFRL1 from WDSCC (KYSE30) to MDSCC (KYSE140) and finally to PDSCC (KYSE410). In-silico tools predicted miR-107 as most significant miRNA regulating FGFRL1 expression. qRT-PCR revealed miR-107 expression to be significantly and inversely correlated with FGFRL1 expression in 73% (22/30) EC tissues (p=0.015) and over-expression of miR-107 resulted in significantly decreased expression of FGFRL1 at mRNA (fold change=0.11, p=0.0016) as well as protein level in miR-107 versus NC treated cells. Luciferase reporter assay using FGFRL1-3'UTR further confirmed it to be a direct target of miR-107. CONCLUSION: Our results herein document clinical as well as functional relevance of FGFRL1 in EC and its regulation by miR-107.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , MicroRNAs , Humans , Cell Proliferation , Esophageal Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinoma, Squamous Cell/pathology , Up-Regulation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptor, Fibroblast Growth Factor, Type 5/metabolism
2.
PLoS One ; 17(8): e0273237, 2022.
Article in English | MEDLINE | ID: mdl-35980984

ABSTRACT

Hypertension and osteoporosis are two major disorders, which interact with each other. Specific genetic signals involving the fibroblast growth factor receptor-like 1 (FGFRL1) gene are related to high blood pressure and bone growth in giraffes. FGFRL1 is associated with cardiovascular system and bone formation. We performed an association study to investigate the role of FGFRL1 in hypertension, osteoporosis, and height determination in humans. In addition, we identified three kinds of phenotypes in fibroblast growth factor (FGF) genes and examined their association with the FGFRL1 gene. We identified 42 SNPs in the FGFRL1 gene associated with each trait. We then analyzed the potential functional annotation of each SNP. The FGFRL1 gene was found to be associated with height, hypertension, and osteoporosis, consistent with the results of a previous study. In addition, the FGF2, FGF4, FGF10, FGF18, and FGF22 genes were found to interact with the FGFRL1 gene. Our study suggests that both FGFRL1 and FGFRL1-related genes may determine the height and the prevalence of osteoporosis and hypertension in the Korean population.


Subject(s)
Hypertension , Osteoporosis , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Hypertension/genetics , Osteoporosis/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Receptors, Fibroblast Growth Factor/metabolism
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(3): 251-256, 2022 Mar.
Article in Chinese | MEDLINE | ID: mdl-35365991

ABSTRACT

Objective To investigate the effect of fibroblast growth factor receptor like 1 (FGFRL1) overexpression on the biological behavior of HCT116 human colon cancer cell line. Methods A recombinant plasmid, named as pcDNA3.1-FGFRL1 which expresses FGFRL1 in mammal cells, was constructed. After a transfection of HCT116 cells with pcDNA3.1-FGFRL1, the stable expression cell line was obtained via continual selection with G418, and FGFRL1 expression was analyzed by real time quantitative PCR and Western blotting. In the following experiment, cells were divided into three groups: the blank group (untreated HCT116 cells), the negative group (empty vector stably transfected cells) and the experience group (pcDNA3.1-FGFRL1 stably transfected cells). Cell proliferation was detected by CCK-8 assay. Cell migration ability was analyzed with TranswellTM assay and their apoptosis was evaluated by flow cytometry. Results FGFRL1 mRNA and protein levels increased significantly in FGFRL1 overexpression group. After the overexpression of FGFRL1, proliferation and migration of HCT116 cells dropped significantly, while their apoptosis increased significantly. Conclusion Overexpression of FGFRL1 inhibits the proliferation and migration of colon cancer HCT116 cells and promotes their apoptosis.


Subject(s)
Colonic Neoplasms , Receptors, Fibroblast Growth Factor , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mammals , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptors, Fibroblast Growth Factor/genetics
5.
Am J Med Genet A ; 185(3): 836-840, 2021 03.
Article in English | MEDLINE | ID: mdl-33443296

ABSTRACT

Fibroblast growth factor receptor-like 1 (FGFRL1) encodes a transmembrane protein that is related to fibroblast growth factor receptors but lacks an intercellular tyrosine kinase domain. in vitro studies suggest that FGFRL1 inhibits cell proliferation and promotes cell differentiation and cell adhesion. Mice that lack FGFRL1 die shortly after birth from respiratory distress and have abnormally thin diaphragms whose muscular hypoplasia allows the liver to protrude into the thoracic cavity. Haploinsufficiency of FGFRL1 has been hypothesized to contribute to the development of congenital diaphragmatic hernia (CDH) associated with Wolf-Hirschhorn syndrome. However, data from both humans and mice suggest that disruption of one copy of FGFRL1 alone is insufficient to cause diaphragm defects. Here we report a female fetus with CDH whose 4p16.3 deletion allows us to refine the Wolf-Hirschhorn syndrome CDH critical region to an approximately 1.9 Mb region that contains FGFRL1. We also report a male infant with isolated left-sided diaphragm agenesis who carried compound heterozygous missense variants in FGFRL1. These cases provide additional evidence that deleterious FGFRL1 variants may contribute to the development of CDH in humans.


Subject(s)
Chromosome Deletion , Haploinsufficiency , Hernias, Diaphragmatic, Congenital/pathology , Receptor, Fibroblast Growth Factor, Type 5/genetics , Female , Hernias, Diaphragmatic, Congenital/etiology , Humans , Infant, Newborn , Male , Prognosis
6.
Biomolecules ; 10(10)2020 10 01.
Article in English | MEDLINE | ID: mdl-33019532

ABSTRACT

In mammals, the novel protein fibroblast growth factor receptor-like 1 (FGFRL1) is involved in the development of metanephric kidneys. It appears that this receptor controls a crucial transition of the induced metanephric mesenchyme to epithelial renal vesicles, which further develop into functional nephrons. FGFRL1 knockout mice lack metanephric kidneys and do not express any fibroblast growth factor (FGF) 8 in the metanephric mesenchyme, suggesting that FGFRL1 and FGF8 play a decisive role during kidney formation. FGFRL1 consists of three extracellular immunoglobulin (Ig) domains (Ig1-Ig2-Ig3), a transmembrane domain and a short intracellular domain. We have prepared the extracellular domain (Ig123), the three individual Ig domains (Ig1, Ig2, Ig3) as well as all combinations containing two Ig domains (Ig12, Ig23, Ig13) in recombinant form in human cells. All polypeptides that contain the Ig2 domain (Ig123, Ig12, Ig23, Ig2) were found to interact with FGF8 with very high affinity, whereas all constructs that lack the Ig2 domain (Ig1, Ig3, Ig13) poorly interacted with FGF8 as shown by ELISA and surface plasmon resonance. It is therefore likely that FGFRL1 represents a physiological receptor for FGF8 in the kidney and that the ligand primarily binds to the Ig2 domain of the receptor. With Biacore experiments, we also measured the affinity of FGF8 for the different constructs. All constructs containing the Ig2 domain showed a rapid association and a slow dissociation phase, from which a KD of 2-3 × 10-9 M was calculated. Our data support the hypothesis that binding of FGF8 to FGFRL1 could play an important role in driving the formation of nephrons in the developing kidney.


Subject(s)
Fibroblast Growth Factor 8/genetics , Immunoglobulin Domains/genetics , Kidney/growth & development , Receptor, Fibroblast Growth Factor, Type 5/genetics , Animals , Epithelial-Mesenchymal Transition/genetics , Humans , Kidney/metabolism , Ligands , Mice , Mice, Knockout , Nephrons/growth & development , Nephrons/metabolism , Surface Plasmon Resonance
7.
J Recept Signal Transduct Res ; 40(3): 257-263, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32098557

ABSTRACT

Fibroblast growth factor receptor-like-1 (FGFRL1) is important to cell motility and links with tumorigenic potential in various types of cancers. To investigate the biological function and underlying mechanism of FGFRL1 in rectal adenocarcinoma, we conducted this study. TCGA and Oncomine databases were used to analyze FGFRL1 expression and its association with clinical characteristics or overall survival (OS) in rectal adenocarcinoma patients. siRNA strategy was implemented to knockdown FGFRL1 expression in rectal adenocarcinoma cells. CCK8, colony formation, wound healing, and transwell assays were implemented to measure cell behaviors. qRT-PCR and western blot were utilized to identify mRNA and protein expression levels. FGFRL1 was significantly increased in rectal adenocarcinoma tissue samples, either colon or rectum. High-regulation of FGFRL1 expression induced poorer outcome of rectal adenocarcinoma patients. Downregulation of FGFRL1 inhibited the proliferation, colony formation, migration, and invasion of SW837 cells. The MAPK pathway-related proteins, phosphorylation of MEK and ERK, were also decreased after si-FGFRL1 transfection. These findings demonstrated that FGFRL1, acting as a potential inducator, may promote the progression of rectal adenocarcinoma via activating the MAPK signaling pathway.


Subject(s)
Adenocarcinoma/drug therapy , Molecular Targeted Therapy , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Rectal Neoplasms/drug therapy , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Receptor, Fibroblast Growth Factor, Type 5/genetics , Rectal Neoplasms/pathology , Tumor Stem Cell Assay
8.
Dev Biol ; 461(1): 43-54, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31923383

ABSTRACT

FgfrL1 is a novel growth factor receptor that is primarily expressed in musculoskeletal tissues and the kidney. FgfrL1-deficient mice have a malformed diaphragm and no kidneys. Such animals die immediately after birth because they are not able to inflate their lungs. The FgfrL1 molecule is composed of three extracellular Ig domains, a transmembrane helix and a short intracellular domain. To investigate the contribution of each of these domains to the function of the novel receptor, we generated mice with deletions of the individual domains. Mice lacking the intracellular domain are viable and phenotypically normal. Mice lacking the first (N-terminal) Ig domain are also viable and normal, but have a reduced life span. Mice lacking the Ig2 or the Ig3 domain are born alive, but die within 24 â€‹h after birth. Ig2-deficient animals exhibit substantially smaller kidneys than wild-type littermates and contain a lower number of glomeruli. Ig3-deficient mice completely lack metanephric kidneys. Interestingly, both the Ig2 and the Ig3-deficient animals show only minor alterations in the diaphragm, which still enables them to inflate their lungs after birth. Our results demonstrate that the principal function of the FgfrL1 receptor is to control the growth of the metanephric kidneys by regulating nephrogenesis. It appears that this function is primarily accomplished by the Ig3 domain with some contribution of the Ig2 domain. It is conceivable that the two domains interact with an Fgf ligand and another molecule from the surface of neighboring cells to induce condensation of the metanephric mesenchyme to renal epithelia and glomeruli.


Subject(s)
Diaphragm/abnormalities , Kidney/embryology , Musculoskeletal System/embryology , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/genetics , Organogenesis/physiology , Protein Domains/genetics
9.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31919528

ABSTRACT

Long non-coding RNA (lncRNA) FYVE, RhoGEF and PH domain containing 5 antisense RNA 1 (FGD5-AS1) has been reported as an oncogene in colorectal cancer, promoting its tumorgenesis. The present paper focused on searching the potential function of FGD5-AS1 in non-small cell lung carcinoma (NSCLC). There are connections between the expression of lncRNA FGD5-AS1 and human NSCLC tumor growth and progression. Also, the relationships between FGD5-AS1, hsa-miR-107 and mRNA fibroblast growth factor receptor like 1 (FGFRL1) are going to test their interaction in NSCLC cell lines, which may cause a series of biological behaviors of NSCLC cells. qRT-PCR analysis was conducted to test the expression of RNAs in different situation. CCK-8 experiment and clone formation assay were performed to assess proliferation of NSCLC cells. Also, connection between FGD5-AS1 and hsa-miR-107 were investigated by luciferase reporter assay and RNA pull-down assay. Rescue experiments were performed to verify the modulating relationship between FGD5-AS1, hsa-miR-107 and FGFRL1. High-level expression of FGD5-AS1 was found in NSCLC. FGD5-AS1 may promote the proliferation of NSCLC cells. Also, the combination between hsa-miR-107, FGD5-AS1 and NSCLC have been proved, which means they can play an interaction function in NSCLC cells. Thence, we concluded that lncRNA FGD5-AS1 promotes non-small cell lung cancer cell proliferation through sponging hsa-miR-107 to up-regulate FGFRL1.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , MicroRNAs/genetics , Middle Aged , RNA, Long Noncoding/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Signal Transduction , Up-Regulation
10.
Genes (Basel) ; 10(12)2019 12 10.
Article in English | MEDLINE | ID: mdl-31835491

ABSTRACT

Rapid radiation associated with phenotypic divergence and convergence provides an opportunity to study the genetic mechanisms of evolution. Here we investigate the genus Takifugu that has undergone explosive radiation relatively recently and contains a subset of closely-related species with a scale-loss phenotype. By using observations during development and genetic mapping approaches, we show that the scale-loss phenotype of two Takifugu species, T. pardalis Temminck & Schlegel and T. snyderi Abe, is largely controlled by an overlapping genomic segment (QTL). A search for candidate genes underlying the scale-loss phenotype revealed that the QTL region contains no known genes responsible for the evolution of scale-loss phenotype in other fishes. These results suggest that the genes used for the scale-loss phenotypes in the two Takifugu are likely the same, but the genes used for the similar phenotype in Takifugu and distantly related fishes are not the same. Meanwhile, Fgfrl1, a gene predicted to function in a pathway known to regulate bone/scale development was identified in the QTL region. Since Fgfr1a1, another memebr of the Fgf signaling pathway, has been implicated in scale loss/scale shape in fish distantly related to Takifugu, our results suggest that the convergence of the scale-loss phenotype may be constrained by signaling modules with conserved roles in scale development.


Subject(s)
Animal Scales/physiology , Animal Scales/radiation effects , Takifugu/genetics , Adaptation, Biological/genetics , Animals , Chromosome Mapping , Fishes/genetics , Phenotype , Phylogeny , Quantitative Trait Loci/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptor, Fibroblast Growth Factor, Type 5/metabolism
11.
Gene ; 679: 320-327, 2018 Dec 30.
Article in English | MEDLINE | ID: mdl-30217759

ABSTRACT

Obesity is a major risk for hypertension. However, the associations between hypertension susceptibility loci and the risk of obesity as well as the effects of gene-gene interactions are unclear, especially in the Chinese children population. Six single nucleotide polymorphisms (SNPs) (ATP2B1 rs17249754, CSK rs1378942, MTHFR rs1801133, CYP17A1 rs1004467, STK39 rs3754777, FGF5 rs16998073) were genotyped for 3503 Chinese children, aged 6-18 years. Of them, 758 obese cases and 2745 controls were identified based on the International Obesity Task Force age- and sex-specific BMI references. Among the six SNPs, three were associated with obesity risk (CSK rs1378942: odds ratio (OR) = 1.20, 95% confidence interval (CI) 1.01-1.43, P = 0.042; MTHFR rs1801133: OR = 1.19, 95% CI 1.05-1.34, P = 0.006; FGF5 rs16998073: OR = 1.14, 95% CI 1.00-1.29, P = 0.047). The genetic risk score (GRS), based on these three SNPs (CSK rs1378942, MTHFR rs1801133, FGF5 rs16998073), showed a positive association with risk of obesity (OR = 1.18, 95% CI 1.09-1.28, P = 7.60 × 10-5). The same association signals were also detected in the subgroups of puberty and inactivity. In addition, interaction analyses among these loci implied a potential gene-gene interaction between MTHFR and FGF5. These findings show a significant association of hypertension susceptibility loci in Chinese children, suggesting a likely influence of genetic and environmental factors on the risk of obesity.


Subject(s)
Asian People/genetics , Hypertension/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Obesity/genetics , Polymorphism, Single Nucleotide , Receptor, Fibroblast Growth Factor, Type 5/genetics , src-Family Kinases/genetics , Adolescent , CSK Tyrosine-Protein Kinase , Child , China , Cross-Sectional Studies , Epistasis, Genetic , Female , Genetic Association Studies , Humans , Male
12.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217817

ABSTRACT

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Subject(s)
Cytokines/immunology , Diabetes Mellitus/genetics , Insulin-Secreting Cells/immunology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Animals , Diabetes Mellitus/immunology , Dimerization , Female , Fibroblast Growth Factor 2/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 5/chemistry , Receptor, Fibroblast Growth Factor, Type 5/immunology , Up-Regulation
13.
J Immunol Res ; 2018: 7438608, 2018.
Article in English | MEDLINE | ID: mdl-29675438

ABSTRACT

Fibroblast growth factor receptor-like-1 (FGFRL1) has been identified as the fifth fibroblast growth factor receptor. So far, little is known about its biological functions, particularly in cancer development. Here, for the first time, we demonstrated the roles of FGFRL1 in ovarian carcinoma (OC). An array and existing databases were used to investigate the expression profile of FGFRL1 and the relationship between FGFRL1 expression and clinicopathological parameters. FGFRL1 was significantly upregulated in OC patients, and high FGFRL1 expression was correlated with poor prognosis. In vitro cell proliferation, apoptosis and migration assays, and in vivo subcutaneous xenograft tumor models were used to determine the role of FGFRL1. Loss of function of FGFRL1 significantly influenced cell proliferation, apoptosis, and migration of OC cells in vitro and tumor growth in vivo. Chromatin immunoprecipitation PCR analysis and microarray hybridization were performed to uncover the mechanism. FGFRL1 expression could be induced by hypoxia through hypoxia-inducible factor 1α, which directly binds to the promoter elements of FGFRL1. FGFRL1 promoted tumor progression by crosstalk with Hedgehog (Hh) signaling. Taken together, FGFRL1 is a potential predictor and plays an important role in tumor growth and Hh signaling which could serve as potential therapeutic targets for the treatment of OC.


Subject(s)
Carcinogenesis , Ovarian Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cohort Studies , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Hedgehogs/metabolism , Heterografts , Humans , Mice , Microarray Analysis , Neoplasm Transplantation , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Signal Transduction
14.
Mol Cell Endocrinol ; 461: 194-204, 2018 02 05.
Article in English | MEDLINE | ID: mdl-28923346

ABSTRACT

Fibroblast growth factors (FGF) and their receptors (FGFRs) regulate many developmental processes including differentiation of mesenchymal stromal cells (MSC). We developed two MSC lines capable of differentiating to osteoblasts and adipocytes and studied the role of FGFRs in this process. We identified FGFR2 and fibroblast growth factor receptor like-1 (FGFRL1) as possible actors in MSC differentiation with gene microarray and qRT-PCR. FGFR2 and FGFRL1 mRNA expression strongly increased during MSC differentiation to osteoblasts. FGF2 treatment, resulting in downregulation of FGFR2, or silencing FGFR2 expression with siRNAs inhibited osteoblast differentiation. During adipocyte differentiation expression of FGFR1 and FGFRL1 increased and was down-regulated by FGF2. FGFR1 knockdown inhibited adipocyte differentiation. Silencing FGFR2 and FGFR1 in MSCs was associated with decreased FGFRL1 expression in osteoblasts and adipocytes, respectively. Our results suggest that FGFR1 and FGFR2 regulate FGFRL1 expression. FGFRL1 may mediate or modulate FGFR regulation of MSC differentiation together with FGFR2 in osteoblastic and FGFR1 in adipocytic lineage.


Subject(s)
Adipocytes/cytology , Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Down-Regulation/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Expression Profiling , Gene Silencing , Male , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptors, Fibroblast Growth Factor/genetics
15.
Taiwan J Obstet Gynecol ; 56(6): 821-826, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29241927

ABSTRACT

OBJECTIVE: We present prenatal diagnosis of a 4p16.3 interstitial microdeletion associated with bilateral cleft lip and palate and short long bones on prenatal ultrasound, and we discuss the genotype-phenotype correlation. MATERIALS AND METHODS: A 32-year-old woman underwent amniocentesis at 22 weeks of gestation because of bilateral cleft lip and palate and short limbs on prenatal ultrasound. Conventional cytogenetic analysis was performed on cultured amniocytes and parental bloods. Oligonucleotide array comparative genomic hybridization (aCGH) was performed on the DNAs extracted from uncultured amniocytes, parental bloods and umbilical cord. Metaphase fluorescence in situ hybridization (FISH) was performed on cultured amniocytes. RESULTS: Amniocentesis revealed a karyotype of 46,XY. The parental karyotypes were normal. aCGH analysis on uncultured amniocytes revealed a 1.66-Mb interstitial microdeletion at 4p16.3 encompassing 23 Online Mendelian Inheritance of in Man (OMIM) genes including FGFRL1 and TACC3. The parents did not have such a deletion. The pregnancy was subsequently terminated, and a malformed fetus was delivered with typical Wolf-Hirschhorn syndrome (WHS) facial appearance and bilateral cleft lip and palate. aCGH analysis of the umbilical cord confirmed the prenatal diagnosis with a result of arr 4p16.3 (72,447-1,742,649) × 1.0 [GRCh37 (hg19)]. Metaphase FISH analysis of cultured amniocytes confirmed a 4p16.3 microdeletion. CONCLUSION: Haploinsufficiency of FGFRL1 and TACC3 at 4p16.3 can be associated with bilateral cleft lip and palate of WHS facial dysmorphism and short long bones. Prenatal diagnosis of facial cleft with short long bones should raise a suspicion of chromosome microdeletion syndromes.


Subject(s)
Amniocentesis/methods , Chromosome Disorders/diagnosis , Cleft Lip/diagnosis , Cleft Palate/diagnosis , Craniofacial Abnormalities/diagnosis , Ectromelia/diagnosis , Hypertelorism/diagnosis , Microtubule-Associated Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Wolf-Hirschhorn Syndrome/diagnosis , Adult , Chromosome Disorders/embryology , Chromosome Disorders/genetics , Chromosomes, Human, Pair 4 , Cleft Lip/embryology , Cleft Lip/genetics , Cleft Palate/embryology , Cleft Palate/genetics , Comparative Genomic Hybridization , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/genetics , Cytogenetic Analysis , Ectromelia/embryology , Ectromelia/genetics , Female , Humans , Hypertelorism/embryology , Hypertelorism/genetics , In Situ Hybridization, Fluorescence , Pregnancy , Wolf-Hirschhorn Syndrome/embryology , Wolf-Hirschhorn Syndrome/genetics
16.
Arch Biochem Biophys ; 625-626: 54-64, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28596102

ABSTRACT

FGFRL1 is a transmembrane receptor that can induce the fusion of CHO cells to multinucleated syncytia. This cell fusion activity has been attributed to the extracellular Ig3 domain of the receptor. We investigated how the fusogenic activity evolved during the evolution of animals. We found that the Ig3 domain from humans, mice, chicken and fish stimulates fusion of CHO cells, while the Ig3 domain from lancelet and sea urchin does not. It is therefore conceivable that the fusogenic activity of FGFRL1 developed during the evolution of vertebrates. Bony fish contain two copies of the FGFRL1 gene because they have undergone a whole-genome duplication. One of the corresponding proteins (FGFRL1a) induces cell-cell fusion, while the other (FGFRL1b) does not. Analysis of chimeric constructs and in vitro mutagenesis suggested that FGFRL1b has lost its fusogenic activity after duplication. A rescue experiment supported this conclusion. When four amino acids were changed, the Ig3 domain of FGFRL1b was converted into an active, fusogenic protein comparable to FGFRL1a. The four amino acids are located in a hydrophobic pocket of the Ig3 domain. It is likely that this hydrophobic pocket interacts with a target molecule on the membrane of adjacent cells to induce cell-cell fusion.


Subject(s)
Giant Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Fusion , Cloning, Molecular , Cricetulus , Evolution, Molecular , Giant Cells/cytology , Humans , Protein Domains , Receptor, Fibroblast Growth Factor, Type 5/chemistry , Receptor, Fibroblast Growth Factor, Type 5/genetics , Sequence Alignment
17.
Tumour Biol ; 39(5): 1010428317698370, 2017 May.
Article in English | MEDLINE | ID: mdl-28459213

ABSTRACT

Fibroblast growth factor receptors are growth factor receptor tyrosine kinases, exerting their roles in embryogenesis, tissue homeostasis, and development of breast cancer. Recent genetic studies have identified some subtypes of fibroblast growth factor receptors as strong genetic loci associated with breast cancer. In this article, we review the recent epidemiological findings and experiment results of fibroblast growth factor receptors in breast cancer. First, we summarized the structure and physiological function of fibroblast growth factor receptors in humans. Then, we discussed the common genetic variations in fibroblast growth factor receptors that affect breast cancer risk. In addition, we also introduced the potential roles of each fibroblast growth factor receptors isoform in breast cancer. Finally, we explored the potential therapeutics targeting fibroblast growth factor receptors for breast cancer. Based on the biological mechanisms of fibroblast growth factor receptors leading to the pathogenesis in breast cancer, targeting fibroblast growth factor receptors may provide new opportunities for breast cancer therapeutic strategies.


Subject(s)
Breast Neoplasms/genetics , Molecular Targeted Therapy , Receptors, Fibroblast Growth Factor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Genetic Association Studies , Humans , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction/genetics
18.
Sci Rep ; 7: 41903, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28211912

ABSTRACT

The characterization of the epigenetic changes within the obesity-related adipose tissue will provide new insights to understand this metabolic disorder, but adipose tissue is not easy to sample in population-based studies. We aimed to evaluate the capacity of circulating leukocytes to reflect the adipose tissue-specific DNA methylation status of obesity susceptibility. DNA samples isolated from subcutaneous adipose tissue and circulating leukocytes were hybridized in the Infinium HumanMethylation 450 BeadChip. Data were compared between samples from obese (n = 45) and non-obese (n = 8-10) patients by Wilcoxon-rank test, unadjusted for cell type distributions. A global hypomethylation of the differentially methylated CpG sites (DMCpGs) was observed in the obese subcutaneous adipose tissue and leukocytes. The overlap analysis yielded a number of genes mapped by the common DMCpGs that were identified to reflect the obesity state in the leukocytes. Specifically, the methylation levels of FGFRL1, NCAPH2, PNKD and SMAD3 exhibited excellent and statistically significant efficiencies in the discrimination of obesity from non-obesity status (AUC > 0.80; p < 0.05) and a great correlation between both tissues. Therefore, the current study provided new and valuable DNA methylation biomarkers of obesity-related adipose tissue pathogenesis through peripheral blood analysis, an easily accessible and minimally invasive biological material instead of adipose tissue.


Subject(s)
DNA Methylation , Leukocytes/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , CpG Islands , Female , Genome, Human , Humans , Male , Middle Aged , Muscle Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Serine Endopeptidases/genetics , Smad3 Protein/genetics
19.
Oncol Rep ; 36(5): 2553-2562, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666683

ABSTRACT

Hypoxia drives cancer to become more aggressive, particularly angiogenesis, and the corresponding mechanisms still need to be further investigated. In hepatocellular carcinoma (HCC), the master hypoxia-induced microRNA (miRNA) miR-210 is upregulated in HCC and participates in HCC progression, but its roles in hypoxia-induced HCC angiogenesis are still unknown. Moreover, the correlation between miR-210 expression and HCC clinical progression also needs elucidation. In the present study, we found that miR-210 expression was progressively increased from normal liver and adjacent non-tumor tissues, to incipient and advanced tumor tissues. In HCC patients, high miR-210 expression was significantly correlated with poor prognosis, both tumor-free survival and overall survival. Moreover, miR-210 expression in HCC was significantly positively correlated with microvascular density. Both in vitro and in vivo studies determined that miR-210 promoted HCC angiogenesis, and the corresponding mechanism was identified to be the direct targeting and inhibition of fibroblast growth factor receptor-like 1 (FGFRL1) expression. Thus, we suggest a new prognosis predictor for HCC patients, and determined the roles of hypoxic miR-210 in HCC angiogenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Receptor, Fibroblast Growth Factor, Type 5/biosynthesis , Adult , Aged , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Male , Mice , MicroRNAs/biosynthesis , Middle Aged , Neovascularization, Pathologic/pathology , Prognosis , Receptor, Fibroblast Growth Factor, Type 5/genetics , Xenograft Model Antitumor Assays
20.
Fiziol Zh (1994) ; 62(2): 12-23, 2016.
Article in English | MEDLINE | ID: mdl-29537219

ABSTRACT

Obesity and its metabolic complications are one of the most profound public health problems and result from interactions between genes and environmental. The development of obesity is tightly connected with dysregulation of intrinsic gene expression mechanisms controlling majority of metabolic processes, which are essential for regulation many physiological functions, including insulin sensitivity, cellular proliferation and angiogenesis. Our objective was to evaluate if expression of angiogenesis related genes VEGF-A, CYR61, PDGFC, FGF1, FGF2, FGFR2, FGFRL1, E2F8, BAI2, HIF1A, and EPAS1 at mRNA level in adipose tissue could participate in the development of obesity and metabolic complications. We have shown that expression level of VEGF-A, PDGFC, FGF2, and FGFRL1 genes is decreased in adipose tissue of obese men with normal glucose tolerance (NGT) versus a group of control subjects. At the same time, in this group of obese individuals a significant up-regulation of CYR61, FGF1, FGFR2, E2F8, BAI2, and HIF1A gene expressions was observed. Impaired glucose tolerance (IGT) in obese patients associates with down-regulation of CYR61 and FGFR2 mRNA and up-regulations of E2F8, FGF1, FGF2, VEGF-A and its splice variant 189 mRNA expressions in adipose tissue versus obese (NGT) individuals. Thus, our data demonstrate that the expression of almost all studied genes is affected in subcutaneous adipose tissue of obese individuals with NGT and that glucose intolerance is associated with gene-specific changes in the expression of E2F8, FGF1, FGF2, VEGF-A, CYR61 and FGFR2 mRNAs. The data presented here provides evidence that VEGF-A, CYR61, PDGFC, FGF1, FGF2, FGFR2, FGFRL1, E2F8, BAI2, and HIF1A genes are possibly involved in the development of obesity and its complications.


Subject(s)
Gene Expression Regulation , Glucose Intolerance/genetics , Glucose/metabolism , Neovascularization, Pathologic/genetics , Obesity/genetics , Subcutaneous Fat/metabolism , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Case-Control Studies , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Tolerance Test , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/metabolism , Insulin Resistance , Lymphokines/genetics , Lymphokines/metabolism , Male , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Obesity/pathology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Subcutaneous Fat/blood supply , Subcutaneous Fat/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL