Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Tumour Biol ; 39(7): 1010428317712592, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28718374

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/genética , Metaloproteinase 3 da Matriz/genética , Neoplasias Ovarianas/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Fatores de Transcrição da Família Snail/genética , Adulto , Idoso , Animais , Caderinas/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Fator 1 de Crescimento de Fibroblastos/biossíntese , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Metaloproteinase 3 da Matriz/biossíntese , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Neoplasias Ovarianas/patologia , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/biossíntese , Transdução de Sinais , Fatores de Transcrição da Família Snail/biossíntese
2.
Tumour Biol ; 39(5): 1010428317705512, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28475003

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/genética , Quimiocina CCL7/genética , Quimiocina CXCL12/genética , Fator 1 de Crescimento de Fibroblastos/genética , Fator de Crescimento de Hepatócito/genética , Neoplasias Laríngeas/genética , Apoptose/genética , Benzilaminas , Fibroblastos Associados a Câncer/metabolismo , Carcinoma de Células Escamosas/patologia , Proliferação de Células/genética , Quimiocina CCL7/biossíntese , Quimiocina CXCL12/biossíntese , Ciclamos , Fator 1 de Crescimento de Fibroblastos/biossíntese , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/biossíntese , Compostos Heterocíclicos/administração & dosagem , Humanos , Neoplasias Laríngeas/patologia , Masculino , Estadiamento de Neoplasias , Receptores CXCR4/antagonistas & inibidores
3.
Zhonghua Jie He He Hu Xi Za Zhi ; 39(9): 714-8, 2016 Sep.
Artigo em Zh | MEDLINE | ID: mdl-27600422

RESUMO

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.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibrose Pulmonar Idiopática/metabolismo , MicroRNAs/genética , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Actinas/genética , Actinas/metabolismo , Animais , Benzamidas , Movimento Celular/fisiologia , Células Cultivadas/efeitos dos fármacos , Dioxóis , Fator 1 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fibroblastos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Proteínas Serina-Treonina Quinases , Fibrose Pulmonar , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Receptores de Fatores de Crescimento Transformadores beta , Fator de Crescimento Transformador beta
4.
Respir Res ; 16: 83, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-26138239

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/biossíntese , Fibrose Pulmonar Idiopática/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Movimento Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia
5.
J Cell Physiol ; 229(12): 2038-48, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24777817

RESUMO

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.


Assuntos
Matriz Extracelular/genética , Fator 1 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/biossíntese , Nicotina/toxicidade , Osteoblastos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/biossíntese , Matriz Extracelular/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Osteopontina/biossíntese , Transdução de Sinais
6.
Protein Expr Purif ; 81(1): 119-125, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21963769

RESUMO

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.


Assuntos
Catelicidinas/biossíntese , Escherichia coli/genética , Fator 1 de Crescimento de Fibroblastos/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos , Bactérias/efeitos dos fármacos , Catelicidinas/química , Catelicidinas/genética , Catelicidinas/farmacologia , Proliferação de Células/efeitos dos fármacos , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Fator 1 de Crescimento de Fibroblastos/química , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Células NIH 3T3 , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia
7.
J Biol Chem ; 285(18): 13885-95, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20189986

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/fisiologia , Fator 1 de Crescimento de Fibroblastos/biossíntese , Elementos de Resposta/fisiologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Fator 1 de Crescimento de Fibroblastos/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Células-Tronco Neoplásicas/metabolismo , Interferência de RNA , Fatores de Transcrição de Fator Regulador X , Fator Regulador X1 , Fatores de Transcrição/genética
8.
Nucleic Acids Res ; 37(16): 5267-78, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19561198

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/genética , Desenvolvimento Muscular/genética , Biossíntese de Proteínas , Ativação Transcricional , Animais , Diferenciação Celular , Linhagem Celular , Fator 1 de Crescimento de Fibroblastos/biossíntese , Camundongos , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Regiões Promotoras Genéticas , Regeneração
9.
Int J Biol Sci ; 17(7): 1795-1807, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33994863

RESUMO

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.


Assuntos
Asma/genética , Fator 1 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Macrófagos Alveolares/metabolismo , MicroRNAs/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Fator de Transcrição STAT1/genética , Animais , Asma/metabolismo , Asma/patologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Exossomos/genética , Exossomos/metabolismo , Fator 1 de Crescimento de Fibroblastos/biossíntese , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , Quinases de Proteína Quinase Ativadas por Mitógeno/biossíntese , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fator de Transcrição STAT1/biossíntese , Transdução de Sinais
10.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 27(1): 126-31, 2010 Feb.
Artigo em Zh | MEDLINE | ID: mdl-20337039

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/biossíntese , Vetores Genéticos/genética , Nicotiana/metabolismo , Plantas Geneticamente Modificadas/metabolismo , Agrobacterium/genética , Fator 1 de Crescimento de Fibroblastos/genética , Plantas Geneticamente Modificadas/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Nicotiana/genética
11.
Appl Biochem Biotechnol ; 191(4): 1562-1579, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32166590

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno/metabolismo , Escherichia coli/metabolismo , Fermentação , Fator 1 de Crescimento de Fibroblastos/biossíntese , Células 3T3 , Animais , Cromatografia por Troca Iônica , Códon , Enteropeptidase/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Microbiologia Industrial , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/biossíntese , Reprodutibilidade dos Testes , Temperatura
12.
JCI Insight ; 5(11)2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32493839

RESUMO

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.


Assuntos
MicroRNAs/genética , Músculo Esquelético , Distrofia Muscular de Duchenne , Células Satélites de Músculo Esquelético , Animais , Regulação para Baixo , Fator 1 de Crescimento de Fibroblastos/biossíntese , Fator 1 de Crescimento de Fibroblastos/genética , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia
13.
Breast Cancer Res Treat ; 117(1): 183-91, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18563556

RESUMO

BACKGROUND: BRCA1- and BRCA2-associated tumors appear to have distinct molecular signatures. BRCA1-associated tumors are predominantly basal-like cancers, whereas BRCA2-associated tumors have a predominant luminal-like phenotype. These two molecular signatures reflect in part the two cell types found in the terminal duct lobular unit of the breast. To elucidate novel genes involved in these two spectra of breast tumorigenesis we performed global gene expression analysis on breast tumors from germline BRCA1 and BRCA2 mutation carriers. METHODOLOGY: Breast tumor RNAs from 7 BRCA1 and 6 BRCA2 mutation carriers were profiled using UHN human 19K cDNA microarrays. Supervised univariate analyses were conducted to identify genes differentially expressed between BRCA1 and BRCA2-associated tumors. Selected discriminatory genes were validated using real time reverse transcription polymerase chain reaction in the tumor RNAs, and/or by immunohistochemistry (IHC) or by in situ hybridization (ISH) on tissue microarrays (TMAs) containing an independent set of 58 BRCA1 and 64 BRCA2-associated tumors. RESULTS: Genes more highly expressed in BRCA1-associated tumors included stathmin, osteopontin, TGFbeta2 and Jagged 1 in addition to genes previously identified as characteristic of basal-like breast cancers. BRCA2-associated cancers were characterized by the higher relative expression of FGF1 and FGFR2. FGFR2 protein was also more highly expressed in BRCA2-associated cancers (P = 0.004). SIGNIFICANCE: BRCA1-associated tumours demonstrated increased expression of component genes of the Notch and TGFbeta pathways whereas the higher expression of FGFR2 and FGF1 in BRCA2-associated cancers suggests the existence of an autocrine stimulatory loop.


Assuntos
Neoplasias da Mama/genética , Perfilação da Expressão Gênica , Genes BRCA2 , Predisposição Genética para Doença , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Biomarcadores Tumorais/genética , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Feminino , Fator 1 de Crescimento de Fibroblastos/biossíntese , Fator 1 de Crescimento de Fibroblastos/genética , Expressão Gênica , Genes BRCA1 , Humanos , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Osteopontina/biossíntese , Osteopontina/genética , RNA Mensageiro/análise , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptores Notch/biossíntese , Receptores Notch/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Serrate-Jagged , Transdução de Sinais/fisiologia , Estatmina/biossíntese , Estatmina/genética , Análise Serial de Tecidos , Fator de Crescimento Transformador beta2/biossíntese , Fator de Crescimento Transformador beta2/genética
14.
Oncol Rep ; 21(1): 211-6, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19082464

RESUMO

Expression of the fibroblast growth factor (FGF)-1, FGF-2, fibroblast growth factor receptor (FGFR)-1, and FGFR-2 genes has been reported in various cancers and is associated with poor outcomes in patients with solid tumors. This study examined the relations between the relative expression of the FGF genes and clinicopathological factors, especially invasion and metastasis, in patients with colorectal cancer. We studied surgical specimens of cancer tissue and adjacent normal mucosa obtained from 202 patients with untreated colorectal carcinoma. The relative expression levels of FGF-1, FGF-2, FGFR-1, and FGFR-2 mRNA in cancer and in normal adjacent mucosa were measured by quantitative real-time, reverse-transcription polymerase chain reaction. The relative expression level of the FGFR-2 gene was higher in normal adjacent mucosa than in cancer, whereas the relative expression levels of the FGF-1, FGF-2, and FGFR-1 genes were similar. FGFR-1 gene expression levels were higher in the presence than in the absence of liver metastasis. An analysis of the relation between clinicopathological features and gene expression showed that overexpression of FGFR-1 correlated with liver metastasis. Our results suggested that overexpression of the FGFR-1 gene might lead to liver metastasis in colorectal cancer. Overexpression of the FGFR-1 gene may thus be a useful predictor of liver metastasis in patients with colorectal cancer.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Idoso , Biomarcadores Tumorais/genética , Neoplasias Colorretais/metabolismo , Feminino , Fator 1 de Crescimento de Fibroblastos/biossíntese , Fator 1 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Circulation ; 116(17): 1915-24, 2007 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-17909102

RESUMO

BACKGROUND: The distal portion of the long arm of chromosome 5 is linked to hypertension and contains functional candidate blood pressure-regulating genes. METHODS AND RESULTS: Tightening the grid of microsatellite markers under this quantitative trait locus in the Silesian Hypertension Study (629 individuals from 207 Polish hypertensive families) provided enhanced support for linkage of this region to blood pressure (maximal Z=3.51, P=0.0002). The fine mapping, comparative genomics, and functional prioritization identified fibroblast growth factor 1 gene (FGF1) as the positional candidate. Linkage disequilibrium mapping based on 51 single nucleotide polymorphisms spanning the locus showed no overlap between 3 independent haploblocks of FGF1 and the adjacent extragenic chromosomal regions. Single and multilocus family-based analysis revealed that genetic variation within FGF1 haploblock 1 was associated with hypertension and identified a common intronic single nucleotide polymorphism, rs152524, as the major driver of this association (P=0.0026). Real-time quantitative polymerase chain reaction and Western blotting analysis of renal tissue obtained from subjects undergoing unilateral nephrectomy showed an increase in both mRNA and protein FGF1 expression in hypertensive patients compared with normotensive controls. Renal immunohistochemistry revealed that FGF1 was expressed exclusively within the glomerular endothelial and mesangial cells. CONCLUSIONS: Our data demonstrate that genetic variation within FGF1 cosegregates with elevated blood pressure in hypertensive families and that this association is likely to be mediated by upregulation of renal FGF1 expression. The results of our study will need to be replicated in other cohorts.


Assuntos
Cromossomos Humanos Par 5/genética , Fator 1 de Crescimento de Fibroblastos/genética , Hipertensão/genética , Desequilíbrio de Ligação , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas/genética , Pressão Sanguínea/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 5/metabolismo , Família , Feminino , Fator 1 de Crescimento de Fibroblastos/biossíntese , Humanos , Hipertensão/metabolismo , Masculino , Células Mesangiais/metabolismo , Pessoa de Meia-Idade , Polônia , Regulação para Cima/genética
16.
Mol Cell Biol ; 14(6): 4244-50, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7515150

RESUMO

Acidic fibroblast growth factor (FGF) and related family members regulate differentiation in organisms as diverse as Xenopus laevis and mammals. We utilized a well-characterized model of myogenic development to directly assess the importance of endogenously produced FGF in controlling differentiation. A role for endogenous FGF is suggested by the previous finding that acidic and basic FGF abundance in cultured myocytes decreases during differentiation. In this study we inhibited the endogenous production of FGF in murine Sol 8 myoblasts by using antisense RNA and observed precocious myogenic differentiation. Exogenously supplied acidic FGF rescues this phenotype. Further results suggest that the effect of FGF on myogenic differentiation is mediated in part through inhibition of myogenin expression. These results demonstrate a direct role for endogenously synthesized growth factors in regulating myogenesis and provide support for a general role for related proteins in mammalian development.


Assuntos
Diferenciação Celular , Fator 1 de Crescimento de Fibroblastos/genética , Músculos/citologia , Miogenina/biossíntese , RNA Antissenso/metabolismo , Actinas/biossíntese , Animais , Linhagem Celular , Fator 1 de Crescimento de Fibroblastos/biossíntese , Expressão Gênica , Cinética , Mamíferos , Camundongos , RNA Mensageiro/isolamento & purificação , RNA Mensageiro/metabolismo , Transfecção , Xenopus laevis
17.
J Mol Med (Berl) ; 84(6): 491-502, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16389546

RESUMO

Fibroblast growth factor 1 (FGF1) is an angiogenic factor known to play a role in the growth of arteries. The purpose of this study was to evaluate the usefulness of direct intramuscular injection of an optimized expression plasmid encoding FGF1 to augment collateral formation and tissue perfusion in a rabbit ischemic hindlimb model. Truncated FGF1 fused to the human fibroblast interferon (FIN) signal peptide was expressed from a newly designed plasmid backbone with an improved safety profile for gene therapy applications. In vitro, optimization of plasmid design yielded in a dramatic increase in expression efficiency for FGF1, independent of the presence of a signal peptide, as analyzed by Western Blotting. In vivo, successful transgene expression could be demonstrated by FGF1 immunostaining after gene application. FGF1 plasmid containing FIN signal peptide (100, 500, and 1,000 mug), when injected into ischemic muscle areas of rabbits 10 days after ligation of the external iliac artery, exhibited a pronounced therapeutic effect on collateral formation to the ischemic hindlimb in a dose-depending manner, as assessed by physiological (blood pressure ratio, maximal intra-arterial Doppler flow) and anatomical (angiographic score, histologic evaluation of capillary density) measurements 30 days after therapy, compared to saline or lacZ control plasmid. FGF1 plasmid without a signal peptide sequence resulted in a comparable therapeutic effect on collateral formation at comparable doses (500 and 1,000 mug). Our results indicate that intramuscular FGF1 gene application could be useful to stimulate collateral formation in a situation of chronic peripheral ischemia. The presence of a signal peptide does not seem to be obligatory to achieve bioactivity of intramuscular transfected FGF1. An optimized vector design improved both biosafety of gene transfer and expression efficiency of the transgene, rendering this vector highly suitable for human gene therapy. Therefore, this new generation vector encoding FGF1 might be useful as an alternative treatment for patients with chronic ischemic disorders not amenable to conventional therapy.


Assuntos
Fator 1 de Crescimento de Fibroblastos/genética , Técnicas de Transferência de Genes , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Sequência de Aminoácidos , Animais , Pressão Sanguínea , Células Cultivadas , Fator 1 de Crescimento de Fibroblastos/biossíntese , Vetores Genéticos , Humanos , Injeções Intramusculares , Interferon beta/genética , Interferon beta/fisiologia , Masculino , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Plasmídeos , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Coelhos
18.
Biotechnol Appl Biochem ; 48(Pt 3): 143-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17484724

RESUMO

The aFGF (acidic fibroblast growth factor) plays an important role in morphogenesis, angiogenesis and wound healing and is therefore of potential medical interest. A DNA fragment encoding haFGF (human aFGF) has been cloned into the PVX (potato virus X)-based binary vector (pgR107) and transiently expressed in Nicotiana benthamiana (a wild Australian tobacco) by agroinfection. Approx. 1 week after agroinfection, the recombinant haFGF accumulated in the agroinfected plants reached up to 1% of the total soluble protein. haFGF was then purified on heparin-Sepharose CL-6B. The purified haFGF could stimulate the growth of NIH 3T3 cells, suggesting that the recombinant haFGF expressed via PVX viral vector in N. benthamiana was active biologically.


Assuntos
Fator 1 de Crescimento de Fibroblastos/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Nicotiana/genética , Potexvirus , Animais , Fator 1 de Crescimento de Fibroblastos/biossíntese , Humanos , Camundongos , Células NIH 3T3 , Rhizobium
19.
Mol Cell Biol ; 37(18)2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28652266

RESUMO

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.


Assuntos
Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias/patologia , Neovascularização Patológica/genética , Inanição/genética , Animais , Calgranulina A/biossíntese , Calgranulina B/biossíntese , Linhagem Celular Tumoral , Proliferação de Células , Fator 1 de Crescimento de Fibroblastos/biossíntese , Perfilação da Expressão Gênica , Fator de Crescimento de Hepatócito/biossíntese , Humanos , Metaloproteinase 3 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias/genética , Interferência de RNA , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , RNA Interferente Pequeno/genética , Transplante Heterólogo , Microambiente Tumoral/fisiologia
20.
Mol Med Rep ; 16(2): 1643-1652, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28656229

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma de Células Escamosas/metabolismo , Fator 1 de Crescimento de Fibroblastos/biossíntese , Neoplasias Pulmonares/metabolismo , Glicoproteínas de Membrana/biossíntese , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Células Endoteliais/patologia , Feminino , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Inativação Gênica , Humanos , Neoplasias Pulmonares/patologia , Linfangiogênese , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA