Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Int J Biol Sci ; 17(7): 1795-1807, 2021.
Article in English | MEDLINE | ID: mdl-33994863

ABSTRACT

Emerging evidence has suggested the functions of exosomes in allergic diseases including asthma. By using a mouse model with asthma induced by ovalbumin (OVA), we explored the roles of M2 macrophage-derived exosomes (M2Φ-Exos) in asthma progression. M2Φ-Exos significantly alleviated OVA-induced fibrosis and inflammatory responses in mouse lung tissues, as well as inhibited abnormal proliferation, invasion, and fibrosis-related protein production in platelet derived growth factor (PDGF-BB) treated primary mouse airway smooth muscle cells (ASMCs). The OVA administration in mice or the PDGF-BB treatment in ASMCs reduced the expression of miR-370, which was detected in M2Φ-Exos by miRNA sequencing. However, treating the mice or ASMCs with M2Φ-Exos reversed the inhibitory effect of OVA or PDGF-BB on miR-370 expression. We identified that the target of miR-370 was fibroblast growth factor 1 (FGF1). Downregulation of miR-370 by Lv-miR-370 inhibitor or overexpression of FGF1 by Lv-FGF1 blocked the protective roles of M2Φ-Exos in asthma-like mouse and cell models. M2Φ-Exos were found to inactivate the MAPK signaling pathway, which was recovered by miR-370 inhibition or FGF1 overexpression. Collectively, we conclude that M2Φ-Exos carry miR-370 to alleviate asthma progression through downregulating FGF1 expression and the MAPK/STAT1 signaling pathway. Our study may offer a novel insight into asthma treatment.


Subject(s)
Asthma/genetics , Fibroblast Growth Factor 1/genetics , Gene Expression Regulation , Macrophages, Alveolar/metabolism , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , STAT1 Transcription Factor/genetics , Animals , Asthma/metabolism , Asthma/pathology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Down-Regulation , Exosomes/genetics , Exosomes/metabolism , Fibroblast Growth Factor 1/biosynthesis , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , STAT1 Transcription Factor/biosynthesis , Signal Transduction
2.
JCI Insight ; 5(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32493839

ABSTRACT

The severity of Duchenne muscular dystrophy (DMD), an incurable disease caused by the lack of dystrophin, might be modulated by different factors, including miRNAs. Among them, miR-378 is considered of high importance for muscle biology, but intriguingly, its role in DMD and its murine model (mdx mice) has not been thoroughly addressed so far. Here, we demonstrate that dystrophic mice additionally globally lacking miR-378 (double-KO [dKO] animals) exhibited better physical performance and improved absolute muscle force compared with mdx mice. Accordingly, markers of muscle damage in serum were significantly decreased in dKO mice, accompanied by diminished inflammation, fibrosis, and reduced abundance of regenerating fibers within muscles. The lack of miR-378 also normalized the aggravated fusion of dystrophin-deficient muscle satellite cells (mSCs). RNA sequencing of gastrocnemius muscle transcriptome revealed fibroblast growth factor 1 (Fgf1) as one of the most significantly downregulated genes in mice devoid of miR-378, indicating FGF1 as one of the mediators of changes driven by the lack of miR-378. In conclusion, we suggest that targeting miR-378 has the potential to ameliorate DMD pathology.


Subject(s)
MicroRNAs/genetics , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle , Animals , Down-Regulation , Fibroblast Growth Factor 1/biosynthesis , Fibroblast Growth Factor 1/genetics , Mice , Mice, Inbred mdx , Mice, Knockout , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology
3.
Appl Biochem Biotechnol ; 191(4): 1562-1579, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32166590

ABSTRACT

Human acidic fibroblast growth factor (haFGF) is a multifunctional protein involved in regulating a wide range of cellular processes. As a potent therapeutic agent, it is highly desirable to produce recombinant haFGF (r-haFGF) at low cost. However, the complex structure and formation of aggregation confines its high-level soluble expression and functional form. Herein, to produce r-haFGF efficiently in E. coli, we devised a novel soluble expression and cost-effective purification approach based on fusion with Scl2-M (a novel modified collagen-like protein) for the first time. By using this strategy, more than 95% of the Scl2-M-haFGF fusion protein was highly expressed in soluble form and the expression level of targeted fusion protein in shake flasks and 5-L fermenter was 0.42 g/L and 2.28 g/L, respectively. Subsequently, the recombinant Scl2-M-haFGF was readily purified through a facile process of acid precipitation and subjected to enterokinase (EK) cleavage. After Scl2-M cleavage, tag-free r-haFGF was further purified using ion-exchange chromatography. The recovery rate of the whole purification process attained 34.2%. Furthermore, the resulting high-purity (96.0%) r-haFGF was prepared by freeze-drying as a final product, and its bioactivity was confirmed to potentiate the proliferation of L929 and BALB-3T3 fibroblasts. Overall, our developed method has the potential for the massive production of the r-haFGF in the future.


Subject(s)
Bacterial Proteins/metabolism , Collagen/metabolism , Escherichia coli/metabolism , Fermentation , Fibroblast Growth Factor 1/biosynthesis , 3T3 Cells , Animals , Chromatography, Ion Exchange , Codon , Enteropeptidase/metabolism , Humans , Hydrogen-Ion Concentration , Industrial Microbiology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/biosynthesis , Reproducibility of Results , Temperature
4.
Tumour Biol ; 39(7): 1010428317712592, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28718374

ABSTRACT

Ovarian cancer is the most lethal gynecologic malignancy, due to its high propensity for metastasis. Cancer-associated fibroblasts, as the dominant component of tumor microenvironment, are crucial for tumor progression. However, the mechanisms underlying the regulation of ovarian cancer cells by cancer-associated fibroblasts remain little known. Here, we first isolated cancer-associated fibroblasts from patients' ovarian tissues and found that cancer-associated fibroblasts promoted SKOV3 cells' proliferation, migration, and invasion. Fibroblast growth factor-1 was identified as a highly increased factor in cancer-associated fibroblasts compared with normal fibroblasts by quantitative reverse transcription polymerase chain reaction (~4.6-fold, p < 0.01) and ELISA assays (~4-fold, p < 0.01). High expression of fibroblast growth factor-1 in cancer-associated fibroblasts either naturally or through gene recombination led to phosphorylation of fibroblast growth factor receptor 4 in SKOV3 cells, which is followed by the activation of mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway and epithelial-to-mesenchymal transition-associated gene Snail1 and MMP3 expression. Moreover, treatment of SKOV3 cell with fibroblast growth factor receptor inhibitor PD173074 terminated cellular proliferation, migration, and invasion, reduced the phosphorylation level of fibroblast growth factor receptor 4, and suppressed the activation of mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway. In addition, the expression level of Snail1 and MMP3 was reduced, while the expression level of E-cadherin increased. These observations suggest a crucial role for cancer-associated fibroblasts and fibroblast growth factor-1/fibroblast growth factor receptor 4 signaling in the progression of ovarian cancer. Therefore, this fibroblast growth factor-1/fibroblast growth factor receptor 4 axis may become a potential target for the treatment of ovarian cancer.


Subject(s)
Fibroblast Growth Factor 1/genetics , Matrix Metalloproteinase 3/genetics , Ovarian Neoplasms/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Snail Family Transcription Factors/genetics , Adult , Aged , Animals , Cadherins/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Fibroblast Growth Factor 1/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Humans , Matrix Metalloproteinase 3/biosynthesis , Middle Aged , Neoplasm Invasiveness/genetics , Ovarian Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 4/biosynthesis , Signal Transduction , Snail Family Transcription Factors/biosynthesis
5.
Mol Med Rep ; 16(2): 1643-1652, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28656229

ABSTRACT

Podoplanin and fibroblast growth factor (FGF) 1 have been detected more frequently in lung squamous cell carcinoma (SQCC) compared with lung adenocarcinoma. Furthermore, it has been previous demonstrated that FGF1 is located on the edge of tumor nests in certain lung SQCC sections, which resembles the characteristic expression pattern of podoplanin. Podoplanin and FGF1 have roles in lymphangiogenesis and angiogenesis. Based on their consistently specific expression in lung SQCC and similar localization patterns, the present study aimed to investigate whether the expression of podoplanin in tumor cells is correlated with FGF1 expression in lung SQCC and whether their co­expression has clinicopathological significance, particularly for lymphangiogenesis/angiogenesis. The correlation between podoplanin and FGF1 expression in tumor cells of 82 lung SQCC cases was investigated by immunohistochemical staining and the association between the co­expression of podoplanin and FGF1, and clinicopathological factors such as microvessel density (MVD), was examined in these samples. In addition, the prognostic value of co­expression of podoplanin and FGF1 in tumor cells was determined, and the regulation of FGF1 expression and angiogenesis by podoplanin was examined in vitro in a human lung SQCC cell line. Immunohistochemical analysis demonstrated that there was a significant correlation between podoplanin and FGF1 expression in lung SQCC tumor cells (R=0.591; P<0.0001). Co­expression of podoplanin and FGF1 was significantly associated with larger primary tumor size, advanced TNM stage and higher intratumoral MVD. Survival analysis demonstrated that cases with podoplanin and FGF1 double­positive staining had a significantly lower survival rate compared with cases with double­negative staining. In vitro experiments revealed that podoplanin regulated FGF1 expression and affected tube formation of human umbilical vein endothelial cells. Combined, the results demonstrated that podoplanin was co­expressed with FGF1 in lung SQCC and this co­expression was correlated with poor prognosis.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Squamous Cell/metabolism , Fibroblast Growth Factor 1/biosynthesis , Lung Neoplasms/metabolism , Membrane Glycoproteins/biosynthesis , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Endothelial Cells/pathology , Female , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Gene Silencing , Humans , Lung Neoplasms/pathology , Lymphangiogenesis , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Middle Aged , Prognosis , Survival Rate
6.
Mol Cell Biol ; 37(18)2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28652266

ABSTRACT

Long noncoding RNAs play a pivotal role in tumor progression, but their role in cancer cells in the nutrient-starved tumor microenvironment remains unknown. Here, we show that a nutrient starvation-responsive long noncoding RNA, JHDM1D antisense 1 (JHDM1D-AS1), promotes tumorigenesis by regulating angiogenesis in response to nutrient starvation. Expression of JHDM1D-AS1 was increased in cancer cells. In addition, expression of JHDM1D-AS1 was increased in clinical tumor samples compared to that in normal tissue. Stable expression of JHDM1D-AS1 in human pancreatic cancer (PANC-1 and AsPC-1) cells promoted cell growth in vitro Remarkably, these JHDM1D-AS1-expressing cells showed a significant increase in tumor growth in vivo that was associated with increased formation of CD31+ blood vessels and elevated infiltration of CD11b+ macrophage lineage cells into tumor tissues. Genome-wide analysis of tumor xenografts revealed that expression of genes for tumor-derived angiogenic factors such as hHGF and hFGF1 concomitant with host-derived inflammation-responsive genes such as mMmp3, mMmp9, mS100a8, and mS100a9 was increased in tumor xenografts of JHDM1D-AS1-expressing pancreatic cancer cells, leading to a poor prognosis. Our results provide evidence that increased JHDM1D-AS1 expression under nutrient starvation accelerates tumor growth by upregulating angiogenesis, thus laying the foundation for improved therapeutic strategies.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Starvation/genetics , Animals , Calgranulin A/biosynthesis , Calgranulin B/biosynthesis , Cell Line, Tumor , Cell Proliferation , Fibroblast Growth Factor 1/biosynthesis , Gene Expression Profiling , Hepatocyte Growth Factor/biosynthesis , Humans , Matrix Metalloproteinase 3/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms/genetics , RNA Interference , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , RNA, Small Interfering/genetics , Transplantation, Heterologous , Tumor Microenvironment/physiology
7.
Tumour Biol ; 39(5): 1010428317705512, 2017 May.
Article in English | MEDLINE | ID: mdl-28475003

ABSTRACT

Most primarily cultured laryngeal squamous cell carcinoma cells are difficult to propagate in vitro and have a low survival rate. However, in our previous work to establish a laryngeal squamous cell carcinoma cell line, we found that laryngeal cancer-associated fibroblasts appeared to strongly inhibit the apoptosis of primarily cultured laryngeal squamous cell carcinoma cells in vitro. In this study, we investigated whether paired laryngeal cancer-associated fibroblasts alone can effectively support the growth of primarily cultured laryngeal squamous cell carcinoma cells in vitro. In all, 29 laryngeal squamous cell carcinoma specimens were collected and primarily cultured. The laryngeal squamous cell carcinoma cells were separated from cancer-associated fibroblasts by differential trypsinization and continuously subcultured. Morphological changes of the cultured laryngeal squamous cell carcinoma cells were observed. Immunocytofluorescence was used to authenticate the identity of the cancer-associated fibroblasts and laryngeal squamous cell carcinoma cells. Flow cytometry was used to quantify the proportion of apoptotic cells. Western blot was used to detect the protein levels of caspase-3. Enzyme-linked immunosorbent assay was used to detect the levels of chemokine (C-X-C motif) ligand 12, chemokine (C-X-C motif) ligand 7, hepatocyte growth factor, and fibroblast growth factor 1 in the supernatants of the laryngeal squamous cell carcinoma and control cells. AMD3100 (a chemokine (C-X-C motif) receptor 4 antagonist) and an anti-chemokine (C-X-C motif) ligand 7 antibody were used to block the tumor-supporting capacity of cancer-associated fibroblasts. Significant apoptotic changes were detected in the morphology of laryngeal squamous cell carcinoma cells detached from cancer-associated fibroblasts. The percentage of apoptotic laryngeal squamous cell carcinoma cells and the protein levels of caspase-3 increased gradually in subsequent subcultures. In contrast, no significant differences in the proliferation capacity of laryngeal squamous cell carcinoma cells cocultured with cancer-associated fibroblasts were detected during subculturing. High level of chemokine (C-X-C motif) ligand 12 was detected in the culture supernatant of cancer-associated fibroblasts. The tumor-supporting effect of cancer-associated fibroblasts was significantly inhibited by AMD3100. Our findings demonstrate that the paired laryngeal cancer-associated fibroblasts alone are sufficient to support the primary growth of laryngeal squamous cell carcinoma cells in vitro and that the chemokine (C-X-C motif) ligand 12/chemokine (C-X-C motif) receptor 4 axis is one of the major contributors.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chemokine CCL7/genetics , Chemokine CXCL12/genetics , Fibroblast Growth Factor 1/genetics , Hepatocyte Growth Factor/genetics , Laryngeal Neoplasms/genetics , Apoptosis/genetics , Benzylamines , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Chemokine CCL7/biosynthesis , Chemokine CXCL12/biosynthesis , Cyclams , Fibroblast Growth Factor 1/biosynthesis , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/biosynthesis , Heterocyclic Compounds/administration & dosage , Humans , Laryngeal Neoplasms/pathology , Male , Neoplasm Staging , Receptors, CXCR4/antagonists & inhibitors
8.
Zhonghua Jie He He Hu Xi Za Zhi ; 39(9): 714-8, 2016 Sep.
Article in Chinese | MEDLINE | ID: mdl-27600422

ABSTRACT

OBJECTIVE: To investigate the effect of fibroblasts on regulating airway stem cell proliferation in idiopathic pulmonary fibrosis. METHODS: Lung cell suspension was prepared from ß-actin-GFP mice. Airway stem cells were obtained by fluorescence activated cell sorting and co-cultured with lung fibroblasts. The fibroblasts were treated with TGF-ß inhibitor SB43142. The expression of growth factors FGF1/2 and the effect of FGF1/2 on stem cell proliferation were observed. RESULTS: The cloning efficiency of airway stem cells, when co-cultured with normal lung fibroblast cells for 8 days, was (3.5±1.1)%, while the cloning efficiency was reduced to (0.04±0.04)% when co-cultured with lung fibroblasts from idiopathic pulmonary fibrosis patients. The difference between the 2 groups was statistically significant(P=0.002 5). TGF-ß receptor inhibitor SB431542 increased lung fibroblast growth factors FGF1/2 expression.FGF1 mRNA expression was increased to the experimental group 0.005 5 from 0.000 2 in the control group.FGF2 mRNA expression of the amount raised to the experimental group 0.000 15 from 0.000 8 in the control group.FGF1/2 promoted the growth of airway stem cells. After FGF1/2 was co-cultured with normal lung fibroblast cells for 8 days, the cloning efficiency of airway stem cells was (0.3±0.1)%. CONCLUSION: During the development of idiopathic pulmonary fibrosis, fibroblast secreted FGF1/2 regulate airway stem cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/pharmacology , Idiopathic Pulmonary Fibrosis/metabolism , MicroRNAs/genetics , Receptors, Fibroblast Growth Factor/biosynthesis , Actins/genetics , Actins/metabolism , Animals , Benzamides , Cell Movement/physiology , Cells, Cultured/drug effects , Dioxoles , Fibroblast Growth Factor 1/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Fibroblasts , Gene Expression Regulation/drug effects , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Mice , Protein Serine-Threonine Kinases , Pulmonary Fibrosis , Receptor, Transforming Growth Factor-beta Type II , Receptors, Fibroblast Growth Factor/drug effects , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , Receptors, Transforming Growth Factor beta , Transforming Growth Factor beta
9.
Sci Rep ; 5: 16323, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26567460

ABSTRACT

With an increasing clinical demand for functional therapeutic proteins every year, there is an increasing requirement for the massive production of bioactive recombinant human acidic fibroblast growth factor (r-haFGF). In this present study, we delicately explore a strategy for the mass production of r-haFGF protein with biological activity in the transgenic silkworm cocoons. The sequence-optimized haFGF was inserted into an enhanced sericin-1 expression system to generate the original transgenic silkworm strain, which was then further crossed with a PIG jumpstarter strain to achieve the remobilization of the expression cassette to a "safe harbor" locus in the genome for the efficient expression of r-haFGF. In consequence, the expression of r-haFGF protein in the mutant line achieved a 5.6-fold increase compared to the original strain. The high content of r-haFGF facilitated its purification and large-scald yields. Furthermore, the r-haFGF protein bioactively promoted the growth, proliferation and migration of NIH/3T3 cells, suggesting the r-haFGF protein possessed native mitogenic activity and the potential for wound healing. These results show that the silk gland of silkworm could be an efficient bioreactor strategy for recombinant production of bioactive haFGF in silkworm cocoons.


Subject(s)
Bombyx/genetics , Fibroblast Growth Factor 1/biosynthesis , Animals , Animals, Genetically Modified , Bioreactors , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA Transposable Elements , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/isolation & purification , Fibroblast Growth Factor 1/pharmacology , Gene Expression , Humans , Mice , NIH 3T3 Cells , Protein Refolding , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
10.
Hum Pathol ; 46(12): 1821-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391572

ABSTRACT

Fibroblast growth factor (FGF) 1 is identified as a candidate cancer biomarker in several kinds of cancer. However, little is known about its expression and function in non-small cell lung cancer (NSCLC). The aim of this study is to identify the expression of FGF1 in primary human NSCLC tissues and evaluate its clinical significance for NSCLC patients. Archived tissues from NSCLC (n = 113) and adjacent normal lung tissues (n = 71) were examined for the immunohistochemical expression of FGF1; then we analyzed the correlations of FGF1 expression with clinicopathological factors and overall survival of the patients. FGF1 expression was identified in the cytoplasm or both cytoplasm and nucleus of NSCLC cells. Immunoreactive scores of FGF1 were significantly higher in NSCLC specimens than in peritumoral normal tissues. High expression of FGF1 (immunoreactive score >3) was detected in 61.9% (70/113) of NSCLC specimens, and high FGF1 expression in cancer cells was significantly correlated with larger primary tumor size, squamous cell carcinoma (SQCC), and vascular invasion. In addition, FGF1 expression was correlated with intratumoral microvessel density in both SQCC and adenocarcinoma subgroups. Moreover, NSCLC patients with high FGF1 expression had a significantly lower overall survival rate, compared with those with low FGF1 expression. Furthermore, subgroup analyses showed that FGF1 expression was associated with poor prognosis in lung SQCC, but not in adenocarcinoma. These findings suggest that the presence of FGF1 in NSCLC cells may serve as a prognostic indicator and a potential therapeutic target for NSCLC patients, especially for lung SQCC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Fibroblast Growth Factor 1/biosynthesis , Lung Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/pathology , Female , Fibroblast Growth Factor 1/analysis , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis
11.
Respir Res ; 16: 83, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26138239

ABSTRACT

BACKGROUND: Recent clinical studies show that tyrosine kinase inhibitors slow the rate of lung function decline and decrease the number of acute exacerbations in patients with Idiopathic Pulmonary Fibrosis (IPF). However, in the murine bleomycin model of fibrosis, not all tyrosine kinase signaling is detrimental. Exogenous ligands Fibroblast Growth Factor (FGF) 7 and 10 improve murine lung repair and increase survival after injury via tyrosine kinase FGF receptor 2b-signaling. Therefore, the level and location of FGF/FGFR expression as well as the exogenous effect of the most highly expressed FGFR2b ligand, FGF1, was analyzed on human lung fibroblasts. METHODS: FGF ligand and receptor expression was evaluated in donor and IPF whole lung homogenates using western blotting and qPCR. Immunohistochemistry for FGF1 and FGFR1/2/3/4 were performed on human lung tissue. Lastly, the effects of FGF1, a potent, multi-FGFR ligand, were studied on primary cultures of IPF and non-IPF donor fibroblasts. Western blots for pro-fibrotic markers, proliferation, FACS for apoptosis, transwell assays and MetaMorph analyses on cell cultures were performed. RESULTS: Whole lung homogenate analyses revealed decreased FGFR b-isoform expression, and an increase in FGFR c-isoform expression. Of the FGFR2b-ligands, FGF1 was the most significantly increased in IPF patients; downstream targets of FGF-signaling, p-ERK1/2 and p-AKT were also increased. Immunohistochemistry revealed FGF1 co-localization within basal cell sheets, myofibroblast foci, and Surfactant protein-C positive alveolar epithelial type-II cells as well as co-localization with FGFR1, FGFR2, FGFR3, FGFR4 and myofibroblasts expressing the migratory marker Fascin. Both alone and in the presence of heparin, FGF1 led to increased MAPK-signaling in primary lung fibroblasts. While smooth muscle actin was unchanged, heparin + FGF1 decreased collagen production in IPF fibroblasts. In addition, FGF1 + heparin increased apoptosis and cell migration. The FGFR inhibitor (PD173074) attenuated these effects. CONCLUSIONS: Strong expression of FGF1/FGFRs in pathogenic regions of IPF suggest that aberrant FGF1-FGFR signaling is increased in IPF patients and may contribute to the pathogenesis of lung fibrosis by supporting fibroblast migration and increased MAPK-signaling.


Subject(s)
Fibroblast Growth Factor 1/biosynthesis , Idiopathic Pulmonary Fibrosis/metabolism , Receptors, Fibroblast Growth Factor/biosynthesis , Cell Movement/physiology , Cells, Cultured , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology
12.
Vasc Endovascular Surg ; 48(5-6): 372-7, 2014.
Article in English | MEDLINE | ID: mdl-24951292

ABSTRACT

Therapeutic angiogenesis using gene therapy is a novel strategy for the treatment of critical limb ischemia (CLI). We conducted a meta-analysis to evaluate the efficacy and safety of gene therapy for the treatment of CLI with no option of revascularization. Randomized placebo controlled trials of gene therapy on CLI were identified by searching PubMed (from 1990 to October 2013) and EMBASE (from 1990 to October 2013). Five eligible studies were selected for the meta-analysis. Among these studies, a total of 425 patients received gene therapy of either fibroblast growth factor 1 or hepatocyte growth factor, and 365 patients were given placebo. No statistical differences were observed between the 2 groups in major amputation or death at 1 year (risk ratio [RR], 0.83; 95% confidence interval [CI], 0.51-1.39; P = .48) and wound healing at 6 months (RR, 1.55; 95% CI, 0.73-3.28; P = .25). Gene therapy had similar occurrence of serious adverse events as control (RR, 1.05; 95% CI, 0.97-1.14; P = .23).


Subject(s)
Angiogenic Proteins/biosynthesis , Extremities/blood supply , Genetic Therapy , Ischemia/therapy , Neovascularization, Physiologic/genetics , Amputation, Surgical , Angiogenic Proteins/genetics , Chi-Square Distribution , Critical Illness , Evidence-Based Medicine , Fibroblast Growth Factor 1/biosynthesis , Fibroblast Growth Factor 1/genetics , Genetic Therapy/adverse effects , Genetic Therapy/mortality , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/genetics , Humans , Ischemia/diagnosis , Ischemia/genetics , Ischemia/metabolism , Ischemia/mortality , Ischemia/physiopathology , Limb Salvage , Odds Ratio , Randomized Controlled Trials as Topic , Risk Factors , Time Factors , Treatment Outcome , Wound Healing
13.
J Cell Physiol ; 229(12): 2038-48, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24777817

ABSTRACT

Exposure to nicotine and other compounds contained in cigarette smoking affects human health. This study examined the effects of exposure to a single or multiple sub-toxic nicotine concentrations on human osteoblasts. Cell growth and expression of genes involved in bone differentiation, extracellular matrix (ECM) metabolism, and growth factor signaling pathways were investigated in nicotine-treated cells compared to untreated cells. Depending on osteoblast concentration and maturation stages, nicotine differently regulated cell growth. Real-time PCR showed regulated expressions of genes expressed by nicotine-treated osteoblasts compared to untreated cells. Among ECM genes, type I collagen was down-regulated and osteonectin was up-regulated in nicotine-treated osteoblasts; similarly, fibroblast growth factor-1 (FGF1) and fibroblast growth factor-2 (FGF2), two members of FGF signaling system, were discordantly modulated; genes involved in osteoblast maturation and differentiation such as alkaline phosphatase (ALP), runt-related transcription factor-2 (RUNX2), and bone sialoprotein (BSP) were over-expressed after drug treatment. Our results show a positive association between nicotine exposure and osteoblast phenotype and illustrate for the first time a mechanism whereby acute or chronic exposure to sub-toxic nicotine concentrations may affect bone formation through the impairment of growth factor signaling system and ECM metabolism.


Subject(s)
Extracellular Matrix/genetics , Fibroblast Growth Factor 1/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Nicotine/toxicity , Osteoblasts/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Collagen Type I/biosynthesis , Extracellular Matrix/drug effects , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Osteopontin/biosynthesis , Signal Transduction
14.
Neurochem Int ; 62(1): 43-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23147682

ABSTRACT

Stroke-prone spontaneously hypertensive rats (SHRSP) have an abnormality in cholesterol synthesis, but the pathological relevance of this to stroke and related neuronal disorders is not yet clear. The induction of astrocyte-derived cholesterol transportation to neurons by apolipoprotein E (apoE) promotes neuronal repair after brain injuries such as stroke. Such repair is reduced by interleukin-1 beta (IL-1ß) and stroke conditions. Furthermore, fibroblast growth factor 1 (FGF1) regulates the production of apoE-cholesterol-rich high density lipoproteins (HDL) and induces gliosis of astrocytes. On the other hand, high levels of plasma carotenoids reduce the risk of ischemic stroke. Thus, we investigated the expression of apoE in primary astrocytes that had been treated with IL-1ß or ß-carotene. In addition, we compared the expression levels of Apoe genes in astrocytes from SHRSP/Izm and normal control rats, Wistar-Kyoto rats (WKY/Izm) following hypoxia/reoxygenation (H/R). The expression levels of genes and proteins were investigated by RT-PCR, Western blotting (WB), and immunofluorescence analysis. IL-1ß decreased the expression levels of the Apoe gene. Conversely, ß-carotene significantly enhanced the expression levels of genes related to cholesterol regulation, including Abca1, Abcg1, Hmgcr as well as Apoe. During H/R, the gene expression levels of Apoe were decreased in the SHRSP/Izm rats in comparison with the WKY/Izm rats. These results suggest that IL-1ß decreases Apoe expression levels, whereas ß-carotene strongly elevates Apoe levels and inhibits FGF1-mediated gliosis of astrocytes. Furthermore, under hypoxic stress, astrocytes isolated from SHRSP/Izm rats displayed altered regulation of Apoe compared with those from WKY/Izm rats.


Subject(s)
Apolipoproteins E/biosynthesis , Astrocytes/metabolism , Diet , Interleukin-1beta/biosynthesis , beta Carotene/pharmacology , Animals , Astrocytes/drug effects , Blotting, Western , Cell Hypoxia/physiology , Cell Survival , Cholesterol/metabolism , Coloring Agents , Fibroblast Growth Factor 1/biosynthesis , Fibroblast Growth Factor 1/genetics , Fluorescent Antibody Technique , Gene Expression/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Species Specificity , Stroke/genetics , Stroke/metabolism , Tetrazolium Salts , Thiazoles
15.
Protein Expr Purif ; 81(1): 119-125, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21963769

ABSTRACT

Human acidic fibroblast growth factor (haFGF) stimulates repair of delayed healing which still remains a tremendously world-wide issue. However, most of the patients with delayed healings have to face another creeping problem - microbial infection, which is one of the most frequent complications that still lead to wound healing failure. LL-37/hCAP-18 is the only cathelicidin-derived antimicrobial peptide found in human with a wide range of antimicrobial activities. In the present study, a novel hybrid protein combining LL-37 with haFGF was designed. The DNA sequence encoding recombination fusion protein LL-37-haFGF was subcloned into the pET-21b vector for protein expression in Escherichia coli strain BL21 (DE3). The recombinant protein was expressed as a His-tagged protein and purified using a combination of Ni affinity and CM-Sepharose chromatography at a purity of 95.43% as detected by RP-HPLC and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Antimicrobial activity assays showed that the purified LL-37-haFGF had improved antimicrobial activities in vitro compared with LL-37. Methylthiazoletetrazolium (MTT) assay showed that the purified LL-37-haFGF also had a distinct mitogenic activity in NIH 3T3 cells. These data suggests the recombinant protein LL-37-haFGF has pharmaceutical potential for applications in wound healing.


Subject(s)
Cathelicidins/biosynthesis , Escherichia coli/genetics , Fibroblast Growth Factor 1/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides , Bacteria/drug effects , Cathelicidins/chemistry , Cathelicidins/genetics , Cathelicidins/pharmacology , Cell Proliferation/drug effects , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Fibroblast Growth Factor 1/chemistry , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/pharmacology , Humans , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , NIH 3T3 Cells , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
16.
Int J Cardiol ; 152(3): 307-13, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-20674996

ABSTRACT

Acidic and basic fibroblast growth factors (FGF-1/FGF-2) promote angiogenesis in cancer. Angiogenesis is integral to cardiac repair following myocardial infarction (MI). The potential regulation of FGF-1/FGF-2 in cardiac angiogenesis postMI remains unexplored. Herein, we examined the temporal and spatial expression of FGF-1/FGF-2 and FGF receptors (FGFR) in the infarcted rat heart at days 1, 3, 7, and 14 postMI. FGF-1/-2 gene and protein expression, cells expressing FGF-1/-2 and FGFR expression were examined by quantitative in situ hybridization, RT-PCR; western blot, immunohistochemistry and quantitative in vitro autoradiography. Compared to the normal heart, we found that in the border zone and infarcted myocardium 1) FGF-1 gene expression was increased in the first week postMI and returned to control levels at week 2; FGF-1 protein levels were, however, largely reduced at day 1, then elevated at day 3 peaked at day 7 and declined at day 14; and cells expressing FGF-1 were primarily inflammatory cells; 2) FGF-2 gene expression was significantly elevated from day 1 to day 14; the increase in FGF-2 protein level was most evident at day 7 and cells expressing FGF-2 were primarily endothelial cells; 3) FGFR expression started to increase at day 3 and remained elevated thereafter; and 4) FGF-1/FGF-2 and FGFR expression remained unchanged in the noninfarcted myocardium. Thus, FGF-1/FGF-2 and FGFR expression are enhanced in the infarcted myocardium in the early stage after MI, which is spatially and temporally coincident with angiogenesis, suggesting that FGF-1/FGF-2 are involved in regulating cardiac angiogenesis and repair.


Subject(s)
Fibroblast Growth Factor 1/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Myocardial Infarction/metabolism , Neovascularization, Pathologic/metabolism , Animals , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/physiology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/physiology , Male , Myocardial Infarction/pathology , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley
17.
J Biol Chem ; 285(18): 13885-95, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20189986

ABSTRACT

Fibroblast growth factor 1 (FGF1) has been suggested to have an important role in cell growth, proliferation, and neurogenesis. Human FGF1 gene 1B promoter (-540 to +31)-driven green fluorescence (F1BGFP) has been shown to monitor endogenous FGF1 expression. F1BGFP could also be used to isolate neural stem/progenitor cells from embryonic, neonatal, and adult mouse brains or to isolate glioblastoma stem cells (GBM-SCs) from human glioblastoma tissues. Here, we present evidence that transcription factor RFX1 could bind the 18-bp cis-elements (-484 to -467) of the F1B promoter, modulate F1BGFP expression and endogenous FGF1 expression, and further regulate the maintenance of GBM-SCs. These observations were substantiated by using yeast one-hybrid assay, electrophoretic mobility shift assay, chromatin immunoprecipitation assay, gain- and loss-of-function assays, and neurosphere assays. Overexpression of RFX1 was shown to down-regulate FGF-1B mRNA expression and neurosphere formation in human glioblastoma cells, whereas RNA interference knockdown of RFX1 demonstrated the opposite effects. Our findings provide insight into FGF1 gene regulation and suggest that the roles of FGF1 and RFX1 in the maintenance of GBM-SCs. RFX1 may negatively regulate the self-renewal of GBM-SCs through modulating FGF-1B and FGF1 expression levels by binding the 18-bp cis-elements of the F1B promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation/physiology , Fibroblast Growth Factor 1/biosynthesis , Response Elements/physiology , Transcription Factors/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 1/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Mice , Neoplastic Stem Cells/metabolism , RNA Interference , Regulatory Factor X Transcription Factors , Regulatory Factor X1 , Transcription Factors/genetics
18.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 27(1): 126-31, 2010 Feb.
Article in Chinese | MEDLINE | ID: mdl-20337039

ABSTRACT

Acid fibroblast growth factor (aFGF) has great potential in clinical application, but it is very expensive. In order to reduce the cost of production and to make full use of the merits integrated with plant bioreator, we have explored the aFGF in transgenic Tobacco expression. AFGF gene was inserted into plant expression vector pBI121; the acquired plants contained aFGF gene expression vector pBI121-TOAB-aF. Using Agrobacterium-mediated gene transformation of Tobacco and using transgenic Tobacco containing kanamycin and cephalosporin culture medium, we obtained kanamycin resistant transgenic Tobacco plants. PCR detection, RT-PCR detection and Western blot detection confirmed that foreign genes were successfully expressed in Tobacco. These data could serve as a theoretical foundation on which to use the plant bioreactor for production of aFGF.


Subject(s)
Fibroblast Growth Factor 1/biosynthesis , Genetic Vectors/genetics , Nicotiana/metabolism , Plants, Genetically Modified/metabolism , Agrobacterium/genetics , Fibroblast Growth Factor 1/genetics , Plants, Genetically Modified/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Nicotiana/genetics
19.
Nucleic Acids Res ; 37(16): 5267-78, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19561198

ABSTRACT

Fibroblast growth factor 1 (FGF1) is involved in muscle development and regeneration. The FGF1 gene contains four tissue-specific promoters allowing synthesis of four transcripts with distinct leader regions. Two of these transcripts contain internal ribosome entry sites (IRESs), which are RNA elements allowing mRNA translation to occur in conditions of blockade of the classical cap-dependent mechanism. Here, we investigated the function and the regulation of FGF1 during muscle differentiation and regeneration. Our data show that FGF1 protein expression is induced in differentiating myoblasts and regenerating mouse muscle, whereas siRNA knock-down demonstrated FGF1 requirement for myoblast differentiation. FGF1 induction occurred at both transcriptional and translational levels, involving specific activation of both promoter A and IRES A, whereas global cap-dependent translation was inhibited. Furthermore, we identified, in the FGF1 promoter A distal region, a cis-acting element able to activate the IRES A-driven translation. These data revealed a mechanism of molecular coupling of mRNA transcription and translation, involving a unique process of IRES activation by a promoter element. The crucial role of FGF1 in myoblast differentiation provides physiological relevance to this novel mechanism. This finding also provides a new insight into the molecular mechanisms linking different levels of gene expression regulation.


Subject(s)
Fibroblast Growth Factor 1/genetics , Muscle Development/genetics , Protein Biosynthesis , Transcriptional Activation , Animals , Cell Differentiation , Cell Line , Fibroblast Growth Factor 1/biosynthesis , Mice , Muscle, Skeletal/physiology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Promoter Regions, Genetic , Regeneration
20.
Curr Opin Investig Drugs ; 10(3): 259-68, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19333884

ABSTRACT

Gene transfer of FGF1 has been demonstrated to successfully promote angiogenesis. NV1FGF, a novel pCOR (conditional origin of replication) DNA plasmid-based gene delivery system, is in development by Sanofi-Aventis for the local expression of FGF1 in the treatment of peripheral vascular disease, and has demonstrated potential to induce therapeutic angiogenesis. Preclinical studies using NV1FGF demonstrated restoration in capillary and arteriolar density in rabbit and hamster models of hind limb ischemia. In phase I and phase II clinical trials, NV1FGF effectively reduced the number of amputations and deaths in the trials, with minimal toxicity, and in conjunction with a sustained increase in mRNA and protein levels of FGF1 and its receptors in patients with critical limb ischemia (CLI). These results prompted a phase III trial of NV1FGF in patients with CLI, with the aim of improving quality of life. Thus, the results of the phase III clinical trial may be a significant advancement in the field of medical science, widening the reach of this therapeutic approach to effectively cure intermittent claudication and CLI.


Subject(s)
Extremities/blood supply , Fibroblast Growth Factor 1/genetics , Genetic Therapy/methods , Intermittent Claudication/therapy , Ischemia/therapy , Neovascularization, Physiologic/genetics , Animals , Critical Illness , Disease Models, Animal , Fibroblast Growth Factor 1/biosynthesis , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Vectors , Humans , Intermittent Claudication/etiology , Intermittent Claudication/genetics , Intermittent Claudication/metabolism , Intermittent Claudication/physiopathology , Ischemia/complications , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Patents as Topic , Plasmids , Quality of Life , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...