Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Int Immunopharmacol ; 127: 111407, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38134594

RESUMO

Pancreatic cancer (PC) is a serious threat to human health, with most patients diagnosed at the advanced stages of the disease. Treatment with gemcitabine (GEM) leads to PC GEM resistance. In addition, cancer stem cell (CSC)-derived exosomes play an important role in cancer progression. We aimed to investigate the role and mechanism of action of PC stem cell-derived exosomes in PC drug resistance and progression. CSC-derived exosomes increased the proportion of F4/80+/CD86 + cells and levels of M2 polarization factors. miR-210 is expressed in CSC-derived exosomes. Thus, following co-culture, miR-210 was taken up by macrophages. Transfection or the addition of miR-210 mimics increased the proportion of F4/80+/CD206 + cells and levels of M2 polarization factors. Further, the miR-210 targets inhibited the levels of FGFRL1. The FGFRL1 overexpression plasmid also inhibited miR-210-mediated M2 polarization. After co-culture of THP-M2 cells with PC cells and treatment with GEM, the survival rate, migration rate, and levels of MDR, YB-1, BCRP, p-PI3K, p-AKT, and p-mTOR in PC cells increased. And THP-M2 increased the tumor volume and MDR, YB-1, BCRP, p-PI3K, p-AKT, and p-mTOR levels. Overall, miR-210 from PC stem cell-derived exosome targets and inhibits FGFRL1 to promote macrophage M2 polarization, which activates the p-PI3K/p-AKT/p-mTOR pathway and increases GEM resistance.


Assuntos
MicroRNAs , Neoplasias Pancreáticas , Humanos , Gencitabina , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Macrófagos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo
2.
Ophthalmic Res ; 66(1): 913-920, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37062273

RESUMO

INTRODUCTION: Vascular endothelial cell injury and angiogenesis induced by hyperglycemia are the main pathological basis of vascular complications in diabetes mellitus. Our study aimed to investigate the role and mechanism of miR-210-3p in high glucose (HG)-induced angiogenesis. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with HG to mimic the pathological process of hyperglycemia. HUVECs were divided into the control group, HG group, HG+inhibitor-NC group, and HG+miR-210-3p inhibitor group. Proliferation and migration were tested by wound healing assay, tube formation, and Transwell assay. Quantitation real-time PCR and Western blots were performed to determine the expression of miR-210-3p and relative proteins, respectively. RESULTS: The level of miR-210-3p significantly increased in HUVECs treated by HG. The knockdown of miR-210-3p attenuated the tube formation, proliferation, and migration of cultured HUVECs in vitro to inhibit angiogenesis by increasing the expression of fibroblast growth factor receptor-like 1 (FGFRL1) and then attenuating the phosphorylation of signal transducer and activator of transcription 3 (STAT3), extracellular regulated protein kinases, and protein kinase B (Akt). CONCLUSION: Our study revealed that miR-210-3p might be a promising target for treating diabetic-associated vascular injury.


Assuntos
Diabetes Mellitus , Hiperglicemia , MicroRNAs , Humanos , Regulação para Baixo , MicroRNAs/genética , Angiogênese , Células Endoteliais da Veia Umbilical Humana , Diabetes Mellitus/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Glucose/toxicidade , Proliferação de Células , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo
3.
Asian Pac J Cancer Prev ; 24(4): 1331-1341, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37116156

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , MicroRNAs , Humanos , Proliferação de Células , Neoplasias Esofágicas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinoma de Células Escamosas/patologia , Regulação para Cima , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo
4.
PLoS One ; 17(8): e0273237, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35980984

RESUMO

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.


Assuntos
Hipertensão , Osteoporose , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Hipertensão/genética , Osteoporose/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(3): 251-256, 2022 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-35365991

RESUMO

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.


Assuntos
Neoplasias do Colo , Receptores de Fatores de Crescimento de Fibroblastos , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Mamíferos , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética
7.
Am J Med Genet A ; 185(3): 836-840, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33443296

RESUMO

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.


Assuntos
Deleção Cromossômica , Haploinsuficiência , Hérnias Diafragmáticas Congênitas/patologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Feminino , Hérnias Diafragmáticas Congênitas/etiologia , Humanos , Recém-Nascido , Masculino , Prognóstico
8.
Biomolecules ; 10(10)2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33019532

RESUMO

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.


Assuntos
Fator 8 de Crescimento de Fibroblasto/genética , Domínios de Imunoglobulina/genética , Rim/crescimento & desenvolvimento , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Animais , Transição Epitelial-Mesenquimal/genética , Humanos , Rim/metabolismo , Ligantes , Camundongos , Camundongos Knockout , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Ressonância de Plasmônio de Superfície
9.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 38(5): 558-565, 2020 Oct 01.
Artigo em Chinês | MEDLINE | ID: mdl-33085242

RESUMO

OBJECTIVE: This study aims to investigate the expression of fibroblast growth factor receptor like 1 (FGFRL1) in oral squamous cell carcinoma (OSCC) and reveals its association with tumor cell proliferation and migration. METHODS: Western blot was performed to detect the expression of FGFRL1 protein in OSCC tissues, adjacent normal tissues, OSCC cell lines and normal epithelial cells. After knocking down of FGFRL1 in HN4 cells, CCK-8 and Ki67 assays were performed to detect cell proliferation, wounding healing assay and transwell were performed to detect cell-migration. Western blot was used to detect the expression of protein related to epithelial-mesenchymal transition (EMT). RESULTS: The expression of FGFRL1 in OSCC tissues was higher than that in adjacent nontumor tissues, respectively (t=2.820, P=0.047 8). Moreover, the expression of FGFRL1 in OSCC cells was higher than that in HOK cells. Quantitative real-time polymerase chain reaction (qRT-PCR) showed that FGFRL1 expression of FGFRL1 RNA in HOK cells was lower than that in OSCC cells. HN4 cells transfected with FGFRL1 siRNA were included in the experimental group, whereas HN4 cells treated with NC siRNA were included in the control group. CCK-8 experiment showed no significant difference between the experimental and control groups with regard to proliferation ability at 48 h (P=0.478 1) and 72 h (P=0.334 2). Migration experiment showed that the wound healing areas in the experimental group after 12 h (P=0.022 8), 24 h (P=0.005 1), and 36 h (P=0.009 5)were smaller than that in the control group. Transwell invasion assay showed that the number of invaded cells in the experimental group after 16 h (P=0.008 7) and 24 h (P=0.008 6) were lower than that in the control group. Knocking-down FGFRL1 up-regulated the expression of E-cadherin and down-regulated the expression of N-cadherin and Vimentin in HN4 cells. CONCLUSIONS: FGFRL1 expression in the OSCC tissues was significantly higher than that in the adjacent nontumor tissues. FGFRL1 expression in the OSCC cells was significantly higher than that in the HOK cells, and FGFRL1 had no effect on cell proliferation but promoted tumor cell migration and EMT.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Bucais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos
10.
J Cell Mol Med ; 24(11): 6324-6339, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32396269

RESUMO

Lung cancer has the highest mortality rate among human cancers, and the majority of deaths can be attributed to metastatic spread. Lung cancer stem cells (CSCs) are a component of the tumour microenvironment that contributes to this process. Exosomes are small membrane vesicles secreted by all types of cells that mediate cell interactions, including cancer metastasis. Here, we show that lung CSC-derived exosomes promote the migration and invasion of lung cancer cells, up-regulate expression levels of N-cadherin, vimentin, MMP-9 and MMP-1, and down-regulate E-cadherin expression. Moreover, we verified that these exosomes contribute to a pro-metastatic phenotype in lung cancer cells via miR-210-3p transfer. The results of bioinformatics analysis and dual-luciferase reporter assays further indicated that miR-210-3p may bind to fibroblast growth factor receptor-like 1 (FGFRL1); silencing FGFRL1 enhanced the metastatic ability of lung cancer cells, whereas overexpressing FGFRL1 suppressed metastasis. Taken together, our results provide new insights into a potential molecular mechanism whereby lung CSC-derived exosomal miR-210-3p targets FGFRL1 to promote lung cancer metastasis. FGFRL1 may be a promising therapeutic target in lung cancer.


Assuntos
Exossomos/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Endocitose , Exossomos/ultraestrutura , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , MicroRNAs/genética , Fenótipo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
11.
J Recept Signal Transduct Res ; 40(3): 257-263, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32098557

RESUMO

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.


Assuntos
Adenocarcinoma/tratamento farmacológico , Terapia de Alvo Molecular , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Neoplasias Retais/tratamento farmacológico , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Neoplasias Retais/patologia , Ensaio Tumoral de Célula-Tronco
12.
J Cell Mol Med ; 24(3): 2123-2134, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31957179

RESUMO

Fibroblast growth factor receptor-like 1 (FGFRL1), a member of the FGFR family, has been demonstrated to play important roles in various cancers. However, the role of FGFRL1 in small-cell lung cancer (SCLC) remains unclear. Our study aimed to investigate the role of FGFRL1 in chemoresistance of SCLC and elucidate the possible molecular mechanism. We found that FGFRL1 levels are significantly up-regulated in multidrug-resistant SCLC cells (H69AR and H446DDP) compared with the sensitive parental cells (H69 and H446). In addition, clinical samples showed that FGFRL1 was overexpressed in SCLC tissues, and high FGFRL1 expression was associated with the clinical stage, chemotherapy response and survival time of SCLC patients. Knockdown of FGFRL1 in chemoresistant SCLC cells increased chemosensitivity by increasing cell apoptosis and cell cycle arrest, whereas overexpression of FGFRL1 in chemosensitive SCLC cells produced the opposite results. Mechanistic investigations showed that FGFRL1 interacts with ENO1, and FGFRL1 was found to regulate the expression of ENO1 and its downstream signalling pathway (the PI3K/Akt pathway) in SCLC cells. In brief, our study demonstrated that FGFRL1 modulates chemoresistance of SCLC by regulating the ENO1-PI3K/Akt pathway. FGFRL1 may be a predictor and a potential therapeutic target for chemoresistance in SCLC.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Pulmonares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfopiruvato Hidratase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Carcinoma de Pequenas Células do Pulmão/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico
13.
Biosci Rep ; 40(1)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31919528

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Longo não Codificante/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais , Regulação para Cima
14.
Dev Biol ; 461(1): 43-54, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31923383

RESUMO

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.


Assuntos
Diafragma/anormalidades , Rim/embriologia , Sistema Musculoesquelético/embriologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese/genética , Organogênese/fisiologia , Domínios Proteicos/genética
15.
Genes (Basel) ; 10(12)2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31835491

RESUMO

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.


Assuntos
Escamas de Animais/fisiologia , Escamas de Animais/efeitos da radiação , Takifugu/genética , Adaptação Biológica/genética , Animais , Mapeamento Cromossômico , Peixes/genética , Fenótipo , Filogenia , Locos de Características Quantitativas/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo
16.
Gene ; 679: 320-327, 2018 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-30217759

RESUMO

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.


Assuntos
Povo Asiático/genética , Hipertensão/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Quinases da Família src/genética , Adolescente , Proteína Tirosina Quinase CSK , Criança , China , Estudos Transversais , Epistasia Genética , Feminino , Estudos de Associação Genética , Humanos , Masculino
17.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30217817

RESUMO

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.


Assuntos
Citocinas/imunologia , Diabetes Mellitus/genética , Células Secretoras de Insulina/imunologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Animais , Diabetes Mellitus/imunologia , Dimerização , Feminino , Fator 2 de Crescimento de Fibroblastos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/imunologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/imunologia , Regulação para Cima
18.
J Immunol Res ; 2018: 7438608, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29675438

RESUMO

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.


Assuntos
Carcinogênese , Neoplasias Ovarianas/metabolismo , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Estudos de Coortes , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Ouriços/metabolismo , Xenoenxertos , Humanos , Camundongos , Análise em Microsséries , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais
19.
Mol Cell Endocrinol ; 461: 194-204, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-28923346

RESUMO

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.


Assuntos
Adipócitos/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Perfilação da Expressão Gênica , Inativação Gênica , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética
20.
Taiwan J Obstet Gynecol ; 56(6): 821-826, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29241927

RESUMO

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.


Assuntos
Amniocentese/métodos , Transtornos Cromossômicos/diagnóstico , Fenda Labial/diagnóstico , Fissura Palatina/diagnóstico , Anormalidades Craniofaciais/diagnóstico , Ectromelia/diagnóstico , Hipertelorismo/diagnóstico , Proteínas Associadas aos Microtúbulos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Síndrome de Wolf-Hirschhorn/diagnóstico , Adulto , Transtornos Cromossômicos/embriologia , Transtornos Cromossômicos/genética , Cromossomos Humanos Par 4 , Fenda Labial/embriologia , Fenda Labial/genética , Fissura Palatina/embriologia , Fissura Palatina/genética , Hibridização Genômica Comparativa , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/genética , Análise Citogenética , Ectromelia/embriologia , Ectromelia/genética , Feminino , Humanos , Hipertelorismo/embriologia , Hipertelorismo/genética , Hibridização in Situ Fluorescente , Gravidez , Síndrome de Wolf-Hirschhorn/embriologia , Síndrome de Wolf-Hirschhorn/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...