Fibroblast growth factor (FGF)

  • 文章类型: Journal Article
    成纤维细胞生长因子(FGF)信号包括多种功能,包括调节细胞增殖,分化,形态发生,和图案。FGFs及其受体(FGFR)对于成人组织修复过程至关重要。FGF信号转导异常与软骨损伤等各种病理状况有关,骨丢失,肌肉减少,以及在骨科退行性疾病如骨关节炎(OA)中观察到的其他核心病理变化,椎间盘退变(IVDD),骨质疏松症(OP),和肌少症.特别是在OA和IVDD病理学中,FGF1,FGF2,FGF8,FGF9,FGF18,FGF21和FGF23调节合成,分解代谢,软骨组织骨化。此外,FGFR表达失调(FGFR1和FGFR3)促进软骨降解的病理过程。在OP和肌少症中,内分泌衍生的FGFs(FGF19,FGF21和FGF23)调节骨矿物质的合成和分解以及肌肉组织。FGF2和其他FGF也发挥调节作用。越来越多的研究集中在理解FGF信号在骨科变性中的意义。此外,已经确定了FGF信号中越来越多的潜在靶标,例如FGF9、FGF18和FGF23。然而,应该指出的是,这些发现中的大多数仍处于实验阶段,在考虑临床应用之前,还需要进一步的研究。目前,本综述旨在记录FGF信号通路与骨科疾病发生发展的关系。此外,将评估目前针对FGF信号通路预防和治疗骨科变性的治疗策略。
    Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    已在头颈部鳞状细胞癌(HNSCC)中观察到成纤维细胞生长因子受体(FGFR)信号通路的失调,并且是选择性酪氨酸激酶抑制剂(TKIs)的有希望的治疗靶标。迫切需要对FGFR靶向疗法的反应的潜在预测性生物标志物。了解FGF通路相关基因的表观遗传调控,即FGFRs,FGFs,和CCND1,可以启发生物标志物选择FGFR靶向治疗的方式。
    我们在单个CpG位点分辨率(840CpG位点)下对编码基因FGFR1,FGFR2,FGFR3,FGFR4,FGF1-14,FGF16-23和CCND1进行了DNA甲基化分析使用癌症基因组研究图谱(TCGA)HNSCC队列,包括N=530个肿瘤组织和N=50个正常相邻组织样本。我们将DNA甲基化与人乳头瘤病毒(HPV)和基因扩增状态的mRNA表达相关联。此外,我们研究了N=40个HPV(-)HNSCC细胞系中甲基化与选择性FGFR抑制剂PD173074和AZD4547敏感性的相关性.
    我们发现了与表观遗传调节基因一致的序列-上下文细微的CpG甲基化模式。高甲基化水平主要存在于启动子侧翼和基因体区域,而大多数分析的CpG位点的中央启动子区域存在低甲基化水平。FGFRs,FGFs,CCND1甲基化在肿瘤和正常癌旁组织之间存在显著差异,并与HPV和基因扩增状态相关。CCND1启动子甲基化与CCND1扩增相关。对于大多数分析的CpG位点,甲基化水平与肿瘤组织中mRNA表达相关。此外,我们发现特定CpG位点的DNA甲基化与FGFR1/3选择性抑制剂PD173074和AZD4547的应答显著相关,主要在CCND1的转录起始位点内.
    我们的结果表明CCND1,FGFRs,和FGFs通过HNSCC中的DNA甲基化,并保证进一步研究DNA甲基化作为临床试验中对选择性FGFR抑制剂反应的潜在预测生物标志物。
    Dysregulation of fibroblast growth factor receptor (FGFR) signaling pathway has been observed in head and neck squamous cell carcinoma (HNSCC) and is a promising therapeutic target for selective tyrosine kinase inhibitors (TKIs). Potential predictive biomarkers for response to FGFR-targeted therapies are urgently needed. Understanding the epigenetic regulation of FGF pathway related genes, i.e. FGFRs, FGFs, and CCND1, could enlighten the way towards biomarker-selected FGFR-targeted therapies.
    We performed DNA methylation analysis of the encoding genes FGFR1, FGFR2, FGFR3, FGFR4, FGF1-14, FGF16-23, and CCND1 at single CpG site resolution (840 CpG sites) employing The Cancer Genome Research Atlas (TCGA) HNSCC cohort comprising N = 530 tumor tissue and N = 50 normal adjacent tissue samples. We correlated DNA methylation to mRNA expression with regard to human papilloma virus (HPV) and gene amplification status. Moreover, we investigated the correlation of methylation with sensitivity to the selective FGFR inhibitors PD 173074 and AZD4547 in N = 40 HPV(-) HNSCC cell lines.
    We found sequence-contextually nuanced CpG methylation patterns in concordance with epigenetically regulated genes. High methylation levels were predominantly found in the promoter flank and gene body region, while low methylation levels were present in the central promoter region for most of the analyzed CpG sites. FGFRs, FGFs, and CCND1 methylation differed significantly between tumor and normal adjacent tissue and was associated with HPV and gene amplification status. CCND1 promoter methylation correlated with CCND1 amplification. For most of the analyzed CpG sites, methylation levels correlated to mRNA expression in tumor tissue. Furthermore, we found significant correlations of DNA methylation of specific CpG sites with response to the FGFR1/3-selective inhibitors PD 173074 and AZD4547, predominantly within the transcription start site of CCND1.
    Our results suggest an epigenetic regulation of CCND1, FGFRs, and FGFs via DNA methylation in HNSCC and warrants further investigation of DNA methylation as a potential predictive biomarker for response to selective FGFR inhibitors in clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    In mice, male sex determination depends on FGF9 signalling via FGFR2c in the bipotential gonads to maintain the expression of the key testis gene SOX9. In humans, however, while FGFR2 mutations have been linked to 46,XY disorders of sex development (DSD), the role of FGF9 is unresolved. The only reported pathogenic mutations in human FGF9, FGF9S99N and FGF9R62G, are dominant and result in craniosynostosis (fusion of cranial sutures) or multiple synostoses (fusion of limb joints). Whether these synostosis-causing FGF9 mutations impact upon gonadal development and DSD etiology has not been explored. We therefore examined embryonic gonads in the well-characterized Fgf9 missense mouse mutants, Fgf9S99N and Fgf9N143T, which phenocopy the skeletal defects of FGF9S99N and FGF9R62G variants, respectively. XY Fgf9S99N/S99N and XY Fgf9N143T/N143T fetal mouse gonads showed severely disorganized testis cords and partial XY sex reversal at 12.5 days post coitum (dpc), suggesting loss of FGF9 function. By 15.5 dpc, testis development in both mutants had partly recovered. Mitotic analysis in vivo and in vitro suggested that the testicular phenotypes in these mutants arise in part through reduced proliferation of the gonadal supporting cells. These data raise the possibility that human FGF9 mutations causative for dominant skeletal conditions can also lead to loss of FGF9 function in the developing testis, at least in mice. Our data suggest that, in humans, testis development is largely tolerant of deleterious FGF9 mutations which lead to skeletal defects, thus offering an explanation as to why XY DSDs are rare in patients with pathogenic FGF9 variants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Initiation of expression of fibroblast growth factor receptor 1 (FGFR1) concurrent with loss of FGFR2 expression is a well-documented event in the progression of prostate cancer (PCa). Although it is known that some FGFR isoforms confer advantages in cell proliferation and survival, the mechanism by which the subversion of different FGFR isoforms contributes to PCa progression is incompletely understood. Here, we report that fibroblast growth factor (FGF) promotes NF-κB signaling in PCa cells and that this increase is associated with FGFR1 expression. Disruption of FGFR1 kinase activity abrogated both FGF activity and NF-κB signaling in PCa cells. Of note, the three common signaling pathways downstream of FGFR1 kinase, extracellular signal-regulated kinase 1/2 (ERK1/2), phosphoinositide 3-kinase (PI3K/AKT), and phosphoinositide phospholipase Cγ (PLCγ), were not required for FGF-mediated NF-κB signaling. Instead, transforming growth factor β-activating kinase 1 (TAK1), a central regulator of the NF-κB pathway, was required for FGFR1 to stimulate NF-κB signaling. Moreover, we found that FGFR1 promotes NF-κB signaling in PCa cells by reducing TAK1 degradation and thereby supporting sustained NF-κB activation. Consistently, Fgfr1 ablation in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model reduced inflammation in the tumor microenvironment. In contrast, activation of the FGFR1 kinase in the juxtaposition of chemical-induced dimerization (CID) and kinase 1 (JOCK1) mouse model increased inflammation. As inflammation plays an important role in PCa initiation and progression, these findings suggest that ectopically expressed FGFR1 promotes PCa progression, at least in part, by increasing inflammation in the tumor microenvironment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    骨髓间充质干细胞(BMSCs)易于收集和培养,并确定它具有多方向的分化潜力,此外,它具有低免疫原性,因此,它可以用作皮肤伤口愈合的同种异体细胞来源。水凝胶已被广泛用于皮肤伤口愈合本身,它能够模拟细胞的3D微环境,支持细胞增殖,迁移和分泌。在这项研究中,我们创造了一种新型的生物相容性热敏水凝胶携带BMSCs用于全层皮肤伤口愈合。负载有BMSCs的热敏水凝胶在植入伤口后可以快速实现溶胶-凝胶转变。组织学结果证实,水凝胶-BMSCs组合组对伤口闭合有显著的促进作用,上皮细胞增殖和再上皮化,减少伤口和伤口周围组织的炎症反应。联合治疗还可以促进胶原蛋白沉积,TGF-β1和bFGF分泌与组织重塑。本研究为皮肤创伤的临床治疗提供了一种有希望的策略。
    Bone marrow-derived mesenchymal stem cells (BMSCs) are easy to collect and culture, and it is identified that it has multi-directional differentiation potential, moreover it has low immunogenicity, hence it can be used as an allogeneic cell source for skin wound healing. Hydrogel has been widely used in skin wound healing own to it is able to mimic the 3D microenvironment of cells, which supports cell proliferation, migration and secretion. In this study, we created a novel biocompatible thermo-sensitive hydrogel to carry BMSCs for full-thickness skin wound healing. The thermo-sensitive hydrogel loaded with BMSCs can fast achieve sol-gel transition after implanting to the wound. Histological results confirmed that hydrogel-BMSCs combination group showed significant promotion of wound closure, epithelial cells\' proliferation and re-epithelialization, and reduced inflammatory responses in the wounds and in the tissues surrounding the wounds. The combination therapy also can promote collagen deposition, TGF-β1 and bFGF secretion and tissue remodeling. The present study provides a promising strategy for the clinical treatment of skin wounds.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在表达FGFR1c的BaF3细胞中,不同的硫酸多糖具有不同的激活特定成纤维细胞生长因子(FGF)信号通路的能力。在目前的研究中,我们首先从马尾藻和日本糖精中分离并表征了不同的岩藻依聚糖级分。在FGF-1、-2、-7、-8、-9和-10的存在下,将所有岩藻依聚糖级分与BaF3细胞一起孵育。评估其FGFs/FGFR1c信号激活能力。我们的数据表明,在所有六个测试的FGFs中,来自具有最高硫酸盐含量的粳稻的低分子量岩藻依聚糖级分(LMWF-2M)具有最有效的活性。低硫酸化杂多糖组分LMWF-0.5M,与SMP-1,SMP-D-1和SMP-A-1一起,仅激活FGF-2/FGFR1c对(P<0.05)。相比之下,SMP,SMP-A,和SMP-D刺激除FGF-8外的BaF3细胞增殖。LMWF-1M和LMWF-2M促进FGF-1-,-2-,-8-,和基于-9的信令。硫酸盐含量是观察到的活性的主要因素,其次是分子量。单糖组成也影响了活性,其中SMP及其衍生物在FGF-8存在下不能诱导BaF3细胞增殖。当前研究中揭示的结构-活性关系为岩藻依聚糖在FGF/FGFR信号调节中的潜在应用提供了有用的信息。
    Different sulfated polysaccharides have distinct abilities to activate specific fibroblast growth factor (FGF) signaling pathways in FGFR1c-expressing BaF3 cells. In the current study, we first isolated and characterized different fucoidan fractions from Sargassum maclurei and Saccharina japonica. All of the fucoidan fractions were incubated with BaF3 cells in the presence of FGF-1, -2, -7, -8, -9, and -10, respectively, to evaluate their FGFs/FGFR1c signal-activating ability. Our data showed that low molecular weight fucoidan fraction from S. japonica with highest sulfate content (LMWF-2M) had the most potent activity among all of the six tested FGFs. Low sulfated heteropolysacchairde fraction LMWF-0.5M, along with SMP-1, SMP-D-1, and SMP-A-1, only activated the FGF-2/FGFR1c pair (P<0.05). In contrast, SMP, SMP-A, and SMP-D stimulated BaF3 cell proliferation except for FGF-8. Both LMWF-1M and LMWF-2M facilitated FGF-1-, -2-, -8-, and -9-based signaling. The sulfate content was the major contributing factor to the observed activity followed by the molecular weight. The monosaccharide composition also affected the activity, in that SMP and its derivatives with varied monosaccharide composition could not induce BaF3 cell proliferation in the presence of FGF-8. The structure-activity relationships revealed in current study provided useful information for the potential application of fucoidans in FGF/FGFR signaling regulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    用热水提取Costariacostata的粗多糖,并通过阴离子交换色谱进一步分离为三个多糖级分。然后通过用过氧化氢和抗坏血酸降解多糖来制备三个低分子量片段。基于HGPC首次阐明了多糖及其低分子量片段的结构特征,FT-IR,NMR,MS,单糖组成,和其他化学分析。还检查了它们在BaF3细胞中的抗凝剂和FGF-1、-2、-7、-8、-9、-10/FGFR1c信号传导激活活性。我们的研究表明,多糖在半乳糖和岩藻糖残基的不同位置被硫酸化。APTT-,基于PT和TT的抗凝剂测定结果表明,高分子量和较高的硫酸化程度对于其抗凝活性至关重要。相比之下,不仅多糖而且解聚的片段在FGF-,分子量-,和硫酸化依赖的方式。当前研究中呈现的结果证明了多糖及其片段作为抗凝剂和FGF信号调节剂的潜在用途。
    Crude polysaccharides from Costaria costata were extracted by hot water and further fractionated by anion exchange chromatography into three polysaccharide fractions. Three low molecular weight fragments were then prepared by degradation of the polysaccharides with hydrogen peroxide and ascorbic acid. The structural features of the polysaccharides and their low molecular weight fragments were elucidated for the first time based on the HGPC, FT-IR, NMR, MS, monosaccharide composition, and other chemical analyses. Their anticoagulant and FGF-1, -2, -7, -8, -9, -10/FGFR1c signaling activation activities in BaF3 cells were also examined. Our studies showed that the polysaccharides were sulfated at different positions of galactose and fucose residues. The APTT-, PT- and TT-based anticoagulant assay results indicated that a high molecular weight and a higher degree of sulfation were essential for their anticoagulant activities. In contrast, not only the polysaccharides but also the depolymerized fragments showed significant FGF/FGFR signal activating activities in a FGF-, molecular weight-, and sulfation-dependent manner. The results presented in current study demonstrated the potential use of the polysaccharides and their fragments as anticoagulants and FGF signal regulators.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Idiopathic pulmonary fibrosis (IPF) is characterized by progressive pulmonary scarring, decline in lung function, and often results in death within 3-5 five years after diagnosis. Fibroblast growth factor (FGF) signaling has been implicated in the pathogenesis of IPF; however, the mechanism through which FGF signaling contributes to pulmonary fibrosis remains unclear. We hypothesized that FGF receptor (FGFR) signaling in fibroblasts is required for the fibrotic response to bleomycin. To test this, mice with mesenchyme-specific tamoxifen-inducible inactivation of FGF receptors 1, 2, and 3 (Col1α2-CreER; TCKO mice) were lineage labeled and administered intratracheal bleomycin. Lungs were collected for histologic analysis, whole lung RNA and protein, and dissociated for flow cytometry and FACS. Bleomycin-treated Col1α2-CreER; TCKO mice have decreased pulmonary fibrosis, collagen production, and fewer α-smooth muscle actin-positive (αSMA+) myofibroblasts compared with controls. Freshly isolated Col1α2-CreER; TCKO mesenchymal cells from bleomycin-treated mice have decreased collagen expression compared with wild type mesenchymal cells. Furthermore, lineage labeled FGFR-deficient fibroblasts have decreased enrichment in fibrotic areas and decreased proliferation. These data identify a cell autonomous requirement for mesenchymal FGFR signaling in the development of pulmonary fibrosis, and for the enrichment of the Col1α2-CreER-positive (Col1α2+) mesenchymal lineage in fibrotic tissue following bleomycin exposure. We conclude that mesenchymal FGF signaling is required for the development of pulmonary fibrosis, and that therapeutic strategies aimed directly at mesenchymal FGF signaling could be beneficial in the treatment of IPF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    异位骨化(HO),严重的骨骼外骨形成障碍,作为创伤的常见并发症或罕见的遗传疾病发生。许多保守的信号通路与HO有关;然而,许多形式的HO的确切潜在分子机制仍不清楚.新出现的情况是,骨形态发生蛋白(BMP)信号的失调在该过程中起着核心作用,但是其他保守的信号通路,例如刺猬(HH),Wnt/β-catenin和成纤维细胞生长因子(FGF),也参与其中,通过与BMP信号的串扰或通过其他独立机制。深入了解保守的信号通路是有效预防和治疗HO的必要条件。在这次审查中,我们更新和整合这方面的最新进展。希望,我们的讨论将指向有希望的新颖,用于进一步转化研究和成功临床应用的药物位点。
    Heterotopic ossification (HO), a serious disorder of extra-skeletal bone formation, occurs as a common complication of trauma or in rare genetic disorders. Many conserved signaling pathways have been implicated in HO; however, the exact underlying molecular mechanisms for many forms of HO are still unclear. The emerging picture is that dysregulation of bone morphogenetic protein (BMP) signaling plays a central role in the process, but that other conserved signaling pathways, such as Hedgehog (HH), Wnt/β-catenin and Fibroblast growth factors (FGF), are also involved, either through cross-talk with BMP signaling or through other independent mechanisms. Deep understanding of the conserved signaling pathways is necessary for the effective prevention and treatment of HO. In this review, we update and integrate recent progress in this area. Hopefully, our discussion will point to novel promising, druggable loci for further translational research and successful clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    星形胶质细胞通过反应性星形胶质细胞对中枢神经系统损伤作出反应,但潜在的机制尚不清楚。在这项研究中,我们表明,雷帕霉素复合物(mTORC1)信号的机制靶标在出生后神经元中的失活诱导小鼠反应性星形胶质细胞增生。Raptor(一种mTORC1特异性成分)在有丝分裂后神经元中的消融消除了mTORC1的活性,并产生了具有较小体细胞和较少树突的神经元,导致成年小鼠的小头畸形和异常行为。有趣的是,在这些小鼠中观察到广泛的星形胶质细胞增生而没有明显的星形胶质细胞增生和胶质瘢痕形成。神经元mTORC1的抑制可能通过减少神经元来源的成纤维细胞生长因子2(FGF-2)激活星形胶质细胞增生,这可能会触发星形胶质细胞中的FGF受体信号以维持其非反应状态,和FGF-2注射成功地预防了Raptor敲除小鼠的星形胶质增生。这项研究表明,神经元mTORC1抑制反应性星形胶质细胞增生,并在CNS病理中起重要作用。
    Astrocytes respond to CNS insults through reactive astrogliosis, but the underlying mechanisms are unclear. In this study, we show that inactivation of mechanistic target of rapamycin complex (mTORC1) signaling in postnatal neurons induces reactive astrogliosis in mice. Ablation of Raptor (an mTORC1-specific component) in postmitotic neurons abolished mTORC1 activity and produced neurons with smaller soma and fewer dendrites, resulting in microcephaly and aberrant behavior in adult mice. Interestingly, extensive astrogliosis without significant astrocyte proliferation and glial scar formation was observed in these mice. The inhibition of neuronal mTORC1 may activate astrogliosis by reducing neuron-derived fibroblast growth factor 2 (FGF-2), which might trigger FGF receptor signaling in astrocytes to maintain their nonreactive state, and FGF-2 injection successfully prevented astrogliosis in Raptor knock-out mice. This study demonstrates that neuronal mTORC1 inhibits reactive astrogliosis and plays an important role in CNS pathologies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号