首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 156 毫秒
1.
成纤维细胞生长因子(FGF)有许多重要的生理功能,并与肿瘤的形成有关.为了弄清FGF与成纤维细胞生长因子受体(FGFR)相互作用的机制,人们对FGF和RGFR的各个结合结构域进行了深入、细致的研究,定位了aFGF、bFGF的肝素结合区、bFGF的受体结合区、FGF受体的肝素结合区、配体结合区和FGF受体相互结合区,提出了两个FGF与FGFR相互作用的模型,在此基础上设计了FGF的核酸类、糖类和多肽类抑制剂,为寻找新一代抗癌药物打下了理论基础.  相似文献   

2.
成纤维细胞生长因子9(fibroblast growth factor,FGF9)最初发现于人类神经胶质瘤细胞,是成纤维细胞生长因子家族的成员之一.研究发现FGF9在多种组织的发育及疾病的发生中起重要作用.FGF9与肝素结合活化FGFR3受体,可作用于软骨细胞,在骨骼发育及损伤过程中抑制软骨细胞增生和软骨内骨化.FGF9基因缺失或突变可分别导致骨骼发育不良或肿瘤.本文简要综述FGF9与FGFR3受体在骨发育中的作用及其致病机制的研究进展.  相似文献   

3.
成纤维细胞生长因子在骨修复中的作用和应用   总被引:4,自引:0,他引:4  
成纤维细胞生长因子 ( FGF)是一类与肝素有高亲和性的多聚肽 ,最初根据它能刺激成纤维细胞再生而命名。现已发现有 1 8个成员 ,即 FGF1- FGF18。目前研究得最多的是 FGF1和 FGF2 。根据它们等电点 ( PI)的不同 ,FGF1又称为酸性成纤维细胞生长因子 ( a FGF,PI为 5 .6) ,FGF2 又称为碱性成纤维细胞生长因子 ( b FGF,PI为 9.6) ,它们均通过自分泌和 /或旁分泌途径在组织修复中发挥重要作用。成纤维细胞生长因子受体 ( FGFR)属于免疫球蛋白超家族成员 ,目前已确定了 4种由独立基因编码的人 FGFR。它们是一种跨膜蛋白质 ,分为细…  相似文献   

4.
成纤维细胞生长因子的结构与功能研究进展   总被引:4,自引:0,他引:4  
成纤维细胞生长因子(FGF)是一种重要的多肽生长因子,它可分为酸性(aFGF)和碱性(bFGF)两类.这两类 FGF 在结构与功能的许多方面相似而又有所不同,特别是在肝素结合力以及肝素依赖性上的差异,引起人们的研究兴趣.文中从 FGF 功能区研究,肝素作用机理和结构特征等方面概述了 FGF 结构与功能关系的研究进展.FGF 结构与功能关系的阐明,不仅有助于揭示生物大分子的活性调节机理,也有助于推动 FGF 临床应用研究的开展.  相似文献   

5.
角质细胞生长因子 2 (KGF 2 )也叫成纤维细胞生长因子 10 (FGF 10 ) ,是成纤维细胞生长因子家族的一员 .能特异性促进上皮细胞的增殖、分化和迁移 ,对脊椎动物多种组织和器官的发育起重要调控作用 ,对临床上多种疾病的治疗也有很好的应用前景 .1 KGF 2与受体KGF 2有两种细胞膜表面受体 :FGFR1Ⅲb和FGFR2Ⅲb .KGF 2与FGFR2Ⅲb的亲和力很高 ,而与FGFR1Ⅲb的亲和力很低 ,只有在高浓度KGF 2存在时才与FGFR1Ⅲb结合 .KGF 2与受体结合后 ,促使受体胞内的C端酪氨酸残基磷酸化 ,磷酸化的受体具有了酪氨酸蛋白激酶活性 ,并与一系…  相似文献   

6.
碱性成纤维细胞生长因子(bFGF)相关结合蛋白   总被引:1,自引:0,他引:1  
有四种不同类型的细胞表面或细胞外基质中的蛋白质分子在结合碱性成纤维细胞生长因子 (bFGF)、辅助其发挥生物功能活性方面起着重要的作用。它们是 :(1)细胞膜上的具有酪氨酸激酶活性的FGF受体家族 (FGFRs) ;(2 )细胞外基质中的硫酸乙酰肝素蛋白多糖家族 (HSPGs) ;(3)细胞内富含半胱氨酸的FGF受体 (CFR) ;(4)分泌型的FGF结合蛋白 (FGF BP)。本文试图从它们在bFGF生物功能发挥中可能起到的作用对它们进行简单综述。  相似文献   

7.
碱性成纤维细胞生长因子(bFGF)相关结合蛋白   总被引:1,自引:0,他引:1  
有四种不同类型的细胞表面或细胞外基质中的蛋白质分子在结合碱性成纤维细胞生长因子(bFGF)、辅助其发挥生物功能活性方面起着重要的作用。它们是:(1)细胞膜上的具有酪氨酸激酶活性的FGF受体家族(FGFRs);(2)细胞外基质中的硫酸乙酰肝素蛋白多糖家族(HSPGs);(3)细胞内富含半胱氨酸的FGF受体(CFR);(4)分泌型的FGF结合蛋白(FGF-BP)。本文试图从它们在bFGF生物功能发挥中可能起到的作用对它们进行简单综述。  相似文献   

8.
角质细胞生长因子(KGF)属于成纤维细胞生长因子(FGFs)家族的成员之一,又称FGF一7,最早由Rubin等(1980)从人类胎肺成纤维细胞培养液中分离提纯获得的。人KGF的cDNA编码一含194个氨基酸的单链多肽,其分子量为26-28KD。KGF由各种来源的间质细胞分泌,与肝素有较强的亲和力,其受体KGFR属于蛋白酪氨酸激酶受体家族,目前已知该家族主要包括FGFR1、FGFR2、FGFR3和FGFR4四位成员,KGFR由FGFR2基因编码,为FGFR-2的剪接形式,即FGFR2Ⅲb,其主要分布于上皮细胞,KGF与靶细胞膜上的受体KGFR特异性结合后,促使受体自身磷酸化,从而启动细胞内信号级联反应,进而发挥多种生物学功能:参与组织器官发育、促进细胞增殖及组织损伤修复、减少放化疗引起的副反应,尤其与癌症的发生发展有着密切的联系。该文就KGF的研究进展进行综述。  相似文献   

9.
成纤维细胞生长因子9(fibroblast growth factor 9, FGF9)是成纤维细胞生长因子(fibroblast growth factor,FGF)家族成员之一,属于一种自分泌或旁分泌生长因子。在脑组织中,FGF9主要表达于海马和皮质区,具有促进细胞增殖和维持细胞存活的功能。研究发现,FGF家族在抑郁症患者的多个脑区出现表达紊乱,FGF9在抑郁行为中扮演着负调控角色,但其介导抑郁行为的分子机制尚不清楚。本文综述了FGF9及其家族成员在抑郁中的作用; 围绕其受体(FGFR)信号在中枢神经系统中的功能特点,深入分析FGF9调节抑郁行为中的作用机制;结合运动抗抑郁的神经营养假说,提出经由FGFR/GSK3β/β-catenin通路的FGF信号,可能介导抑郁症的运动干预机制的假设。这些将为FGF9介导抑郁行为和运动抗抑郁的有关研究提供理论的基础和探索的思路。  相似文献   

10.
成纤维细胞生长因子(fibroblast growth factors,FGFs)以旁分泌或内分泌的方式参与多种生理活动的调节,维护成体组织的正常结构、功能并参与代谢调控。FGFs通过结合配体硫酸乙酰肝素或klotho使成纤维细胞生长因子受体二聚化而发挥生物学作用。过去的十年中,FGFs结构和分子机制的研究成果改变了人们对FGF信号在人类健康和疾病发生、发展中的认识,为以FGF及其受体为靶点的药物开发带来新的突破。文章综述了FGF的生理、病理作用及最新应用研究进展。  相似文献   

11.
Heparan sulfate proteoglycans on the cell surface act as low affinity binding sites for acidic and basic fibroblast growth factor (FGF) [Moscatelli (1887): J Cell Physiol 131:123–130] and play an important role in the interaction of FGF with the FGF receptor (FGFR). In this study, several aspects of the interaction of FGFs with cell surface heparan sulfate proteoglycans were examined. Reciprocal cross blocking studies demonstrated that acidic FGF (aFGF) and basic FGF (bFGF) bind to identical or closely associated heparan sulfate motifs on BALB/c 3T3 cell surface heparan sulfate proteoglycans. However, the binding affinity of the two growth factros for these heparan sulfate proteoglycans differs considerably, competition binding data indicating that aFGF has a 4.7-fold lower affinity than bFGF for 3T3 heparan sulfate proteoglycan. Subsequent studies of dissociation kinetics demonstrated that bFGF dissociates form the FGFR at least 10-fold slower than aFGF, whereas, following removal of cell surface heparan sulfate proteoplycan. Subsequent studies of dissociation kinetic demonstrated that bFGF dissociates from the FGFR at least 10-fold slwer than aFGF, whereas, following removal of cell surface heparan sulfate proteoglycans by heparinase treatment, the dissociation rate of both FGFs is similar and rapid. These results support the concept that cell surface heparan sulfate proteoglycans stabilize the interactio fo FGF with FGFR, possibly by the formatin of a ternary complex. © Wiley-Liss, Inc.  相似文献   

12.
The family of fibroblast growth factor receptors (FGFRs) plays an important and well-characterized role in a variety of pathological disorders. FGFR4 is involved in myogenesis and muscle regeneration. Mutations affecting the kinase domain of FGFR4 may cause cancer, for example, breast cancer or rhabdomyosarcoma. Whereas FGFR1–FGFR3 have been structurally characterized, the structure of the FGFR4 kinase domain has not yet been reported. In this study, we present four structures of the kinase domain of FGFR4, in its apo-form and in complex with different types of small-molecule inhibitors. The two apo-FGFR4 kinase domain structures show an activation segment similar in conformation to an autoinhibitory segment observed in the hepatocyte growth factor receptor kinase but different from the known structures of other FGFR kinases. The structures of FGFR4 in complex with the type I inhibitor Dovitinib and the type II inhibitor Ponatinib reveal the molecular interactions with different types of kinase inhibitors and may assist in the design and development of FGFR4 inhibitors.  相似文献   

13.
Fibroblast growth factor (FGF) plays an important role in human embryogenesis, angiogenesis, cell proliferation, and differentiation. Carcinogenesis is accompanied by aberrant constitutive activation of FGF receptors (FGFRs) resulting from missense mutation in the FGFR1-4 genes, generation of chimeric oncogenes, FGFR1-4 gene amplification, alternative splicing shift toward formation of mesenchymal FGFR isoforms, and FGFR overexpression. Altogether, these alterations contribute to auto-and paracrine stimulation of cancer cells and neoangiogenesis. Certain missense mutations are found at a high rate in urinary bladder cancer and can be used for non-invasive cancer recurrence diagnostics by analyzing urine cell pellet DNA. Chimeric FGFR1/3 and amplified FGFR1/2 genes can predict cell response to the targeted therapy in various oncological diseases. In recent years, high-throughput sequencing has been used to analyze exomes of virtually all human tumors, which allowed to construct phylogenetic trees of clonal cancer evolution with special emphasis on driver mutations in FGFR1-4 genes. At present, FGFR blockers, such as multi-kinase inhibitors, specific FGFR inhibitors, and FGF ligand traps are being tested in clinical trials. In this review, we discuss current data on the functioning of the FGFR family proteins in both normal and cancer cells, mutations in the FGFR1-4 genes, and mechanisms underlying their oncogenic potential, which might be interesting to a broad range of scientists searching for specific tumor markers and targeted anti-cancer drugs.  相似文献   

14.
Abstract

Sugar induced protein-protein interactions play an important role in several biological processes. The carbohydrate moieties of proteoglycans, the glycosaminoglycans, bind to growth factors with a high degree of specificity and induce interactions with growth factor receptors, thereby regulate the growth factor activity. We have used molecular modeling method to study the modes of binding of heparin or heparan sulfate proteoglycans (HSPGs) to bFGF that leads to the dimerization of FGF receptor 1 (FGFR1) and activation of receptor tyrosine kinase. Homology model of FGFR1 Ig D(II)-D(III) domains was built to investigate the interactions between heparin, bFGF and FGFR1. The structural requirements to bridge the two monomeric bFGF molecules by heparin or HSPGs and to simulate the dimerization and activation of FGFR1 have been examined. A structural model of the biologically functional dimeric bFGF-heparin complex is proposed based on: (a) the stability of dimeric complex, (b) the favorable binding energies between heparin and bFGF molecules, and (c) its accessibility to FGFR1. The modeled complex between heparin, bFGF and FGFR1 has a stoichiometry of 1 heparin: 2 bFGF: 2 FGFR1. The structural properties of the proposed model of bFGF/heparin/FGFR1 complex are consistent with the binding mechanism of FGF to its receptor, the receptor dimerization, and the reported site-specific mutagenesis and biochemical cross-linking data. In the proposed model heparin bridges the two bFGF monomers in a specific orientation and the resulting complex induces FGF receptor dimerization, suggesting that in the oligosaccharide induced recognition process sugars orient the molecules in a way that brings about specific protein-protein or protein-carbohydrate interactions.  相似文献   

15.
Activation of fibroblast growth factor (FGF) signaling is initiated by a multiprotein complex formation between FGF, FGF receptor (FGFR), and heparan sulfate proteoglycan on the cell membrane. Cross-talk with other factors could affect this complex assembly and modulate the biological response of cells to FGF. We have previously demonstrated that anosmin-1, a glycosylated extracellular matrix protein, interacts with the FGFR1 signaling complex and enhances its activity in an IIIc isoform-specific and HS-dependent manner. The molecular mechanism of anosmin-1 action on FGFR1 signaling, however, remains unknown. Here, we show that anosmin-1 directly binds to FGFR1 with high affinity. This interaction involves domains in the N terminus of anosmin-1 (cysteine-rich region, whey acidic protein-like domain and the first fibronectin type III domain) and the D2–D3 extracellular domains of FGFR1. In contrast, anosmin-1 binds to FGFR2IIIc with much lower affinity and displays negligible binding to FGFR3IIIc. We also show that FGFR1-bound anosmin-1, although capable of binding to FGF2 alone, cannot bind to a FGF2·heparin complex, thus preventing FGFR1·FGF2·heparin complex formation. By contrast, heparin-bound anosmin-1 binds to pre-formed FGF2·FGFR1 complex, generating an anosmin-1·FGFR1·FGF2·heparin complex. Furthermore, a functional interaction between anosmin-1 and the FGFR1 signaling complex is demonstrated by immunofluorescence co-localization and Transwell migration assays where anosmin-1 was shown to induce opposing effects during chemotaxis of human neuronal cells. Our study provides molecular and cellular evidence for a modulatory action of anosmin-1 on FGFR1 signaling, whereby binding of anosmin-1 to FGFR1 and heparin can play a dual role in assembly and activity of the ternary FGFR1·FGF2·heparin complex.FGF5 signaling plays an important role in a wide range of fundamental biological responses (13). Both FGF and FGFR bind to heparan sulfate (HS) and heparin, a highly sulfated type of HS produced in connective tissue mast cells. Heparan sulfate proteoglycans (HSPG) are the cell surface co-receptors essential for the formation of functional FGF·FGFR signaling complex (4, 5). There are four structurally related FGFRs (FGFR1–4), which consist of an extracellular ligand-binding region containing three immunoglobulin (Ig)-like domains (D1–D3), a single transmembrane domain, and a cytoplasmic domain with protein-tyrosine kinase catalytic activity. The 22 members of the FGF family bind to the interface formed by the D2/D3 domains and the linker between these domains (6, 7), whereas a conserved positively charged region in D2 serves as the HS binding site (8). An unusual stretch of seven to eight acidic residues designated as the “acid box” is present in the linker connecting D1 and D2. Alternative splicing events occur to generate various isoforms, including a truncated receptor lacking D1 and the D1–D2 linker or a full-length receptor that differs in the second half of D3, designated as IIIb and IIIc isoforms (5). Two crystal structures have been proposed to demonstrate how the FGF·FGFR·heparin complex is assembled (9, 10). Recent evidence suggests that both may be biologically relevant (11, 12).The diversity of FGF signaling pathways and consequent biological functions require that activation of FGFR should be tightly regulated. Such regulation can occur either at the level of the extracellular receptor-ligand complex assembly or via intracellular modulation of downstream effectors (13). Extracellular regulation mainly involves the interaction between each component of the FGF·FGFR·HS signaling complex. For example, FGF8 is shown to bind mostly to the FGFR IIIc isoforms, whereas FGF7 acts as the preferential ligand for the FGFR2 IIIb isoform (13, 14). Sequence specificity, length, and sulfation patterns of HS are also important regulators of the FGF·FGFR interaction (15, 16).Cell surface proteins other than FGFs and HSPGs participate in FGFR signaling regulation. FLRT3 (a member of the fibronectin-leucine-rich transmembrane protein family) promotes FGF signaling and interacts with FGFR1 and FGFR4 via its extracellular fibronectin type III (FnIII) domain (17). Sef (similar expression to fgf genes) functions as an antagonist of FGF signaling in zebrafish. The two FnIII regions of Sef are essential for its function and interaction with FGFR1 and FGFR2 (18). Neuronal cell adhesion molecule (NCAM), N-cadherin, and L1 have also been identified as functionally relevant in FGFR-mediated neurite outgrowth (1922). The FnIII domains of NCAM bind to the D2 and D3 domains of FGFR1 (19) and FGFR2 (23) to induce ligand-independent receptor phosphorylation.Anosmin-1, an extracellular matrix-associated glycosylated protein, appears to be a novel member of the extracellular FGFR signaling modulators (24, 25). Loss-of-function mutations of anosmin-1 and FGFR1 are associated with Kallmann syndrome (KS), underlying X-linked, and autosomal dominant/recessive inheritance mode, respectively (2628). KS is a human developmental genetic disorder characterized by loss of sense of smell (anosmia) caused by abnormal olfactory bulb development and delayed, even arrested puberty caused by disrupted migration of the gonadotropin-releasing hormone (GnRH)-secreting neuron. We previously reported that anosmin-1 acts as an FGFR1IIIc isoform-specific co-ligand, which enhances signaling activity. In human embryonic GnRH olfactory neuroblast FNC-B4 cells, anosmin-1 induced neurite outgrowth and cytoskeletal rearrangements through FGFR1-dependent mechanisms involving p42/44 and p38 mitogen-activated protein kinases and Cdc42/Rac1 activation (25). A functional interaction is also demonstrable between anosmin-1 and FGFR1 in optic nerve oligodendrocyte precursor development (24). Structurally, anosmin-1 comprises an N-terminal cysteine-rich domain (CR) and a whey acidic protein-like (WAP) domain, followed by four tandem FnIII repeats and a C-terminal histidine rich region (Fig. 1a). Current evidence suggests that anosmin-1 functions by affecting FGF2-induced activation of FGFR1 signaling rather than by directly stimulating the receptor. However, the precise molecular mechanism of this interaction remains unclear.Open in a separate windowFIGURE 1.Generation of recombinant anosmin-1, anosmin-1 mutants, FGFR1D1D3, and FGFR1D2D3 proteins. a, the schematic structures of recombinant proteins of anosmin-1 and FGFR1. Each domain in the wild type (PIWF4), point mutants (mPIWF4N267K, mPIWF4E514K, and mPIWF4F517L), and truncated (PIWF1, PIWF2, and PIF4) anosmin-1 protein analogues are represented by a shaded rectangle. V5 and 6His epitopes at the C terminus are represented by a clear rectangle. Each immunoglobulin-like domain in the full ectodomain (FGFR1D1D3) and truncated form (FGFR1D2D3) of FGFR1 is represented by a half circle. The acid box (AB) is represented by a filled rectangle. H, histidine-rich region. b, 0.5–1 μg of purified recombinant proteins are loaded in each lane and visualized by colloidal blue staining. Molecular mass markers in kilodaltons are shown on the left.We now report for the first time that anosmin-1 directly binds to FGFR1 using surface plasmon resonance (SPR), chemical cross-linking, and immunofluorescence co-localization studies in living cells. This interaction occurs between the N-terminal CR, WAP, and the first FnIII domain of anosmin-1 and D2 and D3 ectodomains of FGFR1. Moreover, SPR studies using sequential injections and Transwell migration assays in immortalized FNC-B4-hTERT cells suggest that anosmin-1 can have opposing effects in the formation and activation of the FGF2·FGFR1·heparin complex depending on the order of their binding interactions with anosmin-1.  相似文献   

16.
Among the seven tyrosine autophosphorylation sites identified in the intracellular domain of tyrosine kinase fibroblast growth factor receptor-1 (FGFR1), five of them are dispensable for FGFR1-mediated mitogenic signaling. The possibility of dissociating the mitogenic activity of basic FGF (FGF2) from its urokinase-type plasminogen activator (uPA)-inducing capacity both at pharmacological and structural levels prompted us to evaluate the role of these autophosphorylation sites in transducing FGF2-mediated uPA upregulation. To this purpose, L6 myoblasts transfected with either wild-type (wt) or various FGFR1 mutants were evaluated for the capacity to upregulate uPA production by FGF2. uPA was induced in cells transfected with wt-FGFR1, FGFR1-Y463F, -Y585F, -Y730F, -Y766F, or -Y583/585F mutants. In contrast, uPA upregulation was prevented in L6 cells transfected with FGFR1-Y463/583/585/730F mutant (FGFR1–4F) or with FGFR1-Y463/583/585/730/766F mutant (FGFR1–5F) that retained instead a full mitogenic response to FGF2; however, preservation of residue Y730 in FGFR1-Y463/583/585F mutant (FGFR1–3F) and FGFR1-Y463/583/585/766F mutant (FGFR1–4Fbis) allows the receptor to transduce uPA upregulation. Wild-type FGFR1, FGFR1–3F, and FGFR1–4F similarly bind to a 90-kDa tyrosine-phosphorylated protein and activate Shc, extracellular signal-regulated kinase (ERK)2, and JunD after stimulation with FGF2. These data, together with the capacity of the ERK kinase inhibitor PD 098059 to prevent ERK2 activation and uPA upregulation in wt-FGFR1 cells, suggest that signaling through the Ras/Raf-1/ERK kinase/ERK/JunD pathway is necessary but not sufficient for uPA induction in L6 transfectants. Accordingly, FGF2 was able to stimulate ERK1/2 phosphorylation and cell proliferation, but not uPA upregulation, in L6 cells transfected with the FGFR1-Y463/730F mutant, whereas the FGFR1-Y583/585/730F mutant was fully active. We conclude that different tyrosine autophosphorylation requirements in FGFR1 mediate cell proliferation and uPA upregulation induced by FGF2 in L6 cells. In particular, phosphorylation of either Y463 or Y730, dispensable for mitogenic signaling, represents an absolute requirement for FGF2-mediated uPA induction.  相似文献   

17.
18.
Expression of both basic fibroblast growth factor (bFGF) and FGF receptors (FGFR) by vascular smooth muscle cells suggests that autocrine FGF signaling mechanisms may have important functions. Inhibition of smooth muscle cell bFGF expression provokes apoptosis, suggesting that endogenous bFGF generates an anti-apoptotic signal. The purpose of this study was to determine whether the survival function of endogenous bFGF requires signaling through FGFR. A recombinant adenovirus encoding a truncated murine FGFR-1 lacking the kinase domain (DN-FGFR) efficiently expressed the transgene in cultured rat aortic smooth muscle cells. The truncated receptor acted in a dominant negative fashion to effectively prevent receptor-mediated signaling, assessed by phosphorylation of p42/p44 MAP kinase. Expression of DN-FGFR provoked apoptosis of SMC in a dose-dependent fashion that was insensitive to recombinant bFGF but could be rescued by platelet derived growth factor or epidermal growth factor. Heterologous growth factor rescue was inhibited by PD98059, an inhibitor of MEK (MAP kinase-kinase). These data demonstrate that inhibition of FGF receptor activation results in apoptosis and suggest that an intact autocrine FGF signaling loop is required for vascular smooth muscle cell survival in vitro. These findings also implicate the Ras/Raf/MEK /MAP kinase cascade in generating or sustaining the survival signal. The functional significance of an autocrine FGF signaling loop in non-transformed cells has important implications for cardiovascular development, remodeling and disease. J. Cell. Physiol. 177:58–67, 1998. © 1998 Wiley-Liss, Inc.  相似文献   

19.
It has been widely known that the giant panda (Ailuropoda melanoleuca) is one of the most endangered species in the world. An optimized platform for maintaining the proliferation of giant panda mesenchymal stem cells (MSCs) is very necessary for current giant panda protection strategies. Basic fibroblast growth factor (bFGF), a member of the FGF family, is widely considered as a growth factor and differentiation inducer within the stem cell research field. However, the role of bFGF on promoting the proliferation of MSCs derived from giant panda bone marrow (BM) has not been reported. In this study, we aimed to investigate the role of bFGF on the proliferation of BM-MSCs derived from giant panda. MSCs were cultured for cell proliferation analysis at 24, 48 and 72 hrs following the addition of bFGF. With increasing concentrations of bFGF, cell numbers gradually increased. This was further demonstrated by performing 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) cell proliferation assay, 5-Bromo-2-deoxyUridine (BrdU) labeling and cell cycle testing. Furthermore, the percentage of MSCs that were OCT4 positive increased slightly following treatment with 5 ng/ml bFGF. Moreover, we demonstrated that the extracellular signal-regulated kinase (ERK) signaling pathway may play an important role in the proliferation of panda MSCs stimulated by bFGF. In conclusion, this study suggests that giant panda BM-MSCs have a high proliferative capacity with the addition of 5 ng/ml bFGF in vitro.  相似文献   

20.
Abstract

In order to map in detail the ligand binding sites of fibroblast growth factor receptor 2 (FGFR2) and keratinocyte growth factor receptor (KGFR), we have generated receptor molecules that are chimeric within the membrane proximal sequence that varies between them. The chimeric molecules are found to bind aFGF with a greater than 5-fold difference in affinity, indicating that there is coupling between the chimeric regions with respect to aFGF binding. Further, binding of bFGF and KGF is abolished in the chimeras, showing that the binding site for these ligands requires the whole of the 48- or 50- amino acid variable sequence to be intact. Direct interactions between the different regions exchanged in the chimeras are most probably involved in forming KGF or bFGF binding sites.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号