Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters

Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 634: 152-158, 2022 12 17.
Article in English | MEDLINE | ID: mdl-36244113

ABSTRACT

Acute lung injury (ALI), with high morbidity and mortality, is mainly resulted by infectious or non-infectious inflammatory stimulators, and it will further evolve into acute respiratory distress syndrome if not controlled. Fibroblast growth factors (FGFs) consist of more than 23 kinds of members, which are involved in various pathophysiological processes of body. However, the effect of FGF5, one member of FGFs, is still not certain in lipopolysaccharide (LPS)-induced ALI. In this study, we explored the possible impacts of FGF5 in LPS-induced ALI and primarily focused on endothelial cell, which was one of the most vulnerable cells in septic ALI. In the mouse group of FGF5 overexpression, LPS-induced lung injuries were mitigated, as well as the pyroptosis levels of pulmonary vascular endothelial cells. Additionally, in vitro human umbilical vein endothelial cells (HUVECs), our results showed that the level of cell pyroptosis was ameliorated with FGF5 overexpression, and AKT signal was activated with the overexpression of FGF5, whereas after administration of MK2206, an inhibitor of AKT signal, the protection of FGF5 was inhibited. Therefore, these results suggested that FGF5 exerted protective effects in endothelial cells exposed to LPS, and this protection of FGF5 could be attributed to activated AKT signal.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Mice , Humans , Animals , Lipopolysaccharides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Mice, Inbred C57BL , Acute Lung Injury/metabolism , Signal Transduction , Lung/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Fibroblast Growth Factor 5/pharmacology
2.
Biochem Biophys Res Commun ; 636(Pt 2): 104-112, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36368152

ABSTRACT

Sepsis accompanied by myocardial injury is an important cause of multiple organ dysfunction, and its underlying molecular mechanism is not fully clear. Although diverse effects of fibroblast growth factor (FGF) in heart have been discovered till now, the specific role of FGF5 in heart remains unclear. Therefore, our study aims to explore the possible impacts of FGF5 on sepsis-induced cardiac injury. Sepsis-induced cardiac injury was established through administration of lipopolysaccharide (LPS). The expression level of FGF5 in sepsis heart was decreased, and injection of FGF5-overexpressing adenovirus attenuated cardiac injury reflected by echocardiographic and pathological findings. Besides, FGF5 overexpression, not only in vivo heart but also in vitro cardiomyocytes, reduced the levels of oxidative stress and pyroptosis resulted from LPS. In addition, overexpression of FGF5 reduced LPS-activated the levels of phosphorylated CaMKII (p-CaMKII), p-NFκB, NLRP3, caspase-1, IL-1ß and IL-18. Furthermore, KN93, the inhibitor of CaMKII, exerted the similarly protective effects on LPS-induced pyroptosis. In summary, our study implied the beneficial effects of FGF5 on LPS-induced cardiac injury, which was at least partially attributed to the inhibition of CaMKII-mediated pyroptotic signaling.


Subject(s)
Pyroptosis , Sepsis , Humans , Myocytes, Cardiac/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Sepsis/metabolism , Fibroblast Growth Factor 5/metabolism , Fibroblast Growth Factor 5/pharmacology
3.
Anim Biotechnol ; 30(4): 279-286, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30798699

ABSTRACT

The relationship between PLP2 gene and cashmere fiber quality of Liaoning cashmere goat was investigated. The sheep fibroblast cells were treated with exogenous cytokines and melatonin, independently, and RNA interference, RT-PCR and in situ hybridization were utilized for investigating the PLP2 gene regulation mechanism underlying the Liaoning cashmere growth. The results showed that the expression of PLP2 gene in the prosperous and degenerative stage is higher than that of the primary follicle, indicating that the PLP2 gene promotes the secondary follicle, wherein the gene is expressed only in the inner root sheath, suggesting its correlation to hair loss. The results of RT-PCR showed that the trend of FGF5 expression in PLP2 gene was positively regulated. The influence of MT on the expression of PLP2 gene was negatively regulated, and the inhibition was gradually enhanced with the passage of time. Studies have confirmed that the Noggin gene is an inhibitor of the BMP signaling pathway. After the noggin gene interferes with the lentivirus infection, the expression of the PLP2 gene is downregulated. Therefore, the PLP2 gene, along with the other suppressor genes including the noggin gene, might affect the development of hair follicles by inhibiting the BMP(Bone morphogenetic proteins)pathway.


Subject(s)
Goats/growth & development , Hair Follicle/growth & development , Membrane Proteins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fibroblast Growth Factor 5/pharmacology , Fibroblasts/metabolism , Fibroblasts/virology , Goats/genetics , Goats/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Insulin-Like Growth Factor I/pharmacology , Lipid Metabolism , Melatonin/pharmacology , Membrane Proteins/genetics , Organ Specificity , RNA Interference , Sheep/metabolism , Sheep/virology , Time Factors
4.
Chin Med J (Engl) ; 135(23): 2859-2868, 2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36728504

ABSTRACT

BACKGROUND: Sepsis is a systemic inflammatory syndrome induced by several infectious agents. Multiple organs are affected by sepsis, including the liver, which plays an important role in metabolism and immune homeostasis. Fibroblast growth factors (FGFs) participate in several biological processes, although the role of FGF5 in sepsis is unclear. METHODS: In this study, lipopolysaccharide (LPS) was administrated to mice to establish a sepsis-induced liver injury. A similar in vitro study was conducted using L-02 hepatocytes. Western blot and immunohistochemistry staining were performed to evaluate the FGF5 expression level in liver tissues and cells. Inflammatory cell infiltrations, cleaved-caspase-3 expressions, reactive oxygen species and levels of inflammatory cytokines were detected by immunofluorescence, dihydroethidium staining, and reverse transcription quantitative polymerase chain reaction analysis, respectively. Flow cytometry was used to detect the apoptosis level of cells. In addition, ribonucleic acid (RNA)-sequencing was applied to explore the possible mechanism by which FGF5 exerted effects. RESULTS: LPS administration caused FGF5 down-regulation in the mouse liver as well as in L-02 hepatocytes. Additionally, with FGF5 overexpression, liver injury and the level of hepatocyte apoptosis were ameliorated. Further, RNA sequencing performed in hepatocytes revealed the phosphoinositide-3-kinase/protein kinase B (PI3K/AKT) pathway as a possible pathway regulated by FGF5 . This was supported using an inhibitor of the PI3K/AKT pathway, which abrogated the protective effect of FGF5 in LPS-induced hepatocyte injury. CONCLUSION: The anti-apoptotic effect of FGF5 on hepatocytes suffering from LPS has been demonstrated and was dependent on the activation of the PI3K/AKT signaling pathway.


Subject(s)
Apoptosis , Fibroblast Growth Factor 5 , Hepatocytes , Sepsis , Animals , Mice , Fibroblast Growth Factor 5/metabolism , Fibroblast Growth Factor 5/pharmacology , Lipopolysaccharides , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/metabolism
5.
Pancreas ; 37(2): 159-64, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18665077

ABSTRACT

OBJECTIVES: Fibroblast growth factor receptor 1 (FGFR1) isoform IIIc enhances and FGFR1-IIIb inhibits pancreatic cancer cell growth. Nothing is presently known about the expression and regulation of human FGFR1-III isoforms. The aim of this study was to identify regulators modulating the specific expression of human FGFR1-IIIb and FGFR1-IIIc. METHODS: Parental cells, cells overexpressing FGFR1-III isoforms, and cells harboring a tetracycline-inducible cyclin D1 antisense expression vector system were used as model systems. RESULTS: FGFR1-IIIb and -IIIc were coexpressed in human pancreatic cancer cells, with FGFR1-IIIc being the predominant isoform. FGFR1-IIIb mRNA expression decreased at higher cell density, whereas FGFR1-IIIc expression remained constant. Insulinlike growth factor I and epidermal growth factor induced expression of FGFR1-IIIc without altering FGFR1-IIIb. In contrast, fibroblast growth factor (FGF)1, FGF2, and FGF5 induced FGFR1-IIIc and reduced the expression of FGFR1-IIIb. Overexpression of one isoform did not alter the expression of the corresponding FGFR1-III isoform. Inhibition of cyclin D1, known to be induced by insulinlike growth factor I, epidermal growth factor, and FGF2, resulted in an inhibition of FGFR1-IIIc expression, whereas FGFR1-IIIb expression was enhanced. CONCLUSIONS: This study demonstrated for the first time that FGFR1-IIIb and FGFR1-IIIc are coexpressed and that the FGFR1-III isoformsare differentially regulated by growth factors and cyclin D1.


Subject(s)
Cyclin D1/metabolism , Growth Substances/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Alternative Splicing/drug effects , Base Sequence , Cell Line, Tumor , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , DNA Primers/genetics , Exons , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 5/metabolism , Fibroblast Growth Factor 5/pharmacology , Growth Substances/pharmacology , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Protein Isoforms/genetics
6.
Oncogene ; 27(30): 4180-90, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18362893

ABSTRACT

Fibroblast growth factor 5 (FGF5) is widely expressed in embryonic but scarcely in adult tissues. Here we report simultaneous overexpression of FGF5 and its predominant high-affinity receptor (FGFR1 IIIc) in astrocytic brain tumour specimens (N=49) and cell cultures (N=49). The levels of both ligand and receptor increased with enhanced malignancy in vivo and in vitro. Furthermore, secreted FGF5 protein was generally present in the supernatants of glioblastoma (GBM) cells. siRNA-mediated FGF5 downmodulation reduced moderately but significantly GBM cell proliferation while recombinant FGF5 (rFGF5) increased this parameter preferentially in cell lines with low endogenous expression levels. Apoptosis induction by prolonged serum starvation was significantly prevented by rFGF5. Moreover, tumour cell migration was distinctly stimulated by rFGF5 but attenuated by FGF5 siRNA. Blockade of FGFR1-mediated signals by pharmacological FGFR inhibitors or a dominant-negative FGFR1 IIIc protein inhibited GBM cell proliferation and/or induced apoptotic cell death. Moreover, rFGF5 and supernatants of highly FGF5-positive GBM cell lines specifically stimulated proliferation, migration and tube formation of human umbilical vein endothelial cells. In summary, we demonstrate for the first time that FGF5 contributes to the malignant progression of human astrocytic brain tumours by both autocrine and paracrine effects.


Subject(s)
Autocrine Communication/physiology , Brain Neoplasms/genetics , Fibroblast Growth Factor 5/physiology , Glioblastoma/genetics , Oncogenes , Paracrine Communication/physiology , Autocrine Communication/drug effects , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Disease Progression , Fibroblast Growth Factor 5/genetics , Fibroblast Growth Factor 5/pharmacology , Genes, Dominant/physiology , Humans , Mutant Proteins/genetics , Mutant Proteins/physiology , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/genetics , Oncogenes/physiology , Paracrine Communication/drug effects , Recombinant Proteins/pharmacology , Transfection , Tumor Cells, Cultured
7.
RNA ; 12(5): 851-61, 2006 May.
Article in English | MEDLINE | ID: mdl-16540693

ABSTRACT

The scanning model of RNA translation proposes that highly stable secondary structures within mRNAs can inhibit translation, while structures of lower thermal stability also affect translation if close enough to the 5' methyl G cap. However, only fragmentary information is available about the dependence of translation efficiency in live mammalian cells on the thermodynamic stability, location, and GC content of RNA structures in the 5'-untranslated region. We devised a two-color fluorescence assay for translation efficiency in single live cells and compared a wide range of hairpins with predicted thermal stabilities ranging from -10 to -50 kcal/mol and 5' G cap-to-hairpin distances of 1-46 bases. Translation efficiency decreased abruptly as hairpin stabilities increased from deltaG = -25 to -35 kcal/mol. Shifting a hairpin as little as nine bases relative to the 5' cap could modulate translation more than 50-fold. Increasing GC content diminished translation efficiency when predicted thermal stability and cap-to-hairpin distances were held constant. We additionally found naturally occurring 5'-untranslated regions affected translation differently in live cells compared with translation in in vitro lysates. Our study will assist scientists in designing experiments that deliberately modulate mammalian translation with designed 5' UTRs.


Subject(s)
RNA Caps/metabolism , Animals , Base Sequence , CHO Cells , COS Cells , Carrier Proteins/pharmacology , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cytomegalovirus/genetics , DNA Repair Enzymes , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 5/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , In Vitro Techniques , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Caps/chemistry , RNA Caps/genetics , RNA, Viral/genetics , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL