Receptor, Platelet-Derived Growth Factor beta

受体,血小板源性生长因子 β
  • 文章类型: Journal Article
    平滑肌细胞(SMC)表型调制,主要由PDGFRβ信号驱动,与闭塞性心血管疾病有关。然而,PDGFRβ的促进和限制性调节机制以及蛋白酪氨酸磷酸酶非受体14(PTPN14)在新生内膜增生中的作用尚不清楚。我们的研究观察到新内膜增生期间SMC中PTPN14的显着上调。PTPN14过表达以磷酸酶活性依赖性方式加剧新生内膜增生,而SMC特异性PTPN14缺乏减轻小鼠的这一过程。RNA-seq表明PTPN14缺陷抑制PDGFRβ信号传导诱导的SMC表型调节。此外,PTPN14与PDGFRβ的胞内区域相互作用并介导其在Y692位点上的去磷酸化。PDGFRβY692的磷酸化负调控PDGFRβ信号传导激活。PTPN14和磷酸-PDGFRβY692的水平与人冠状动脉的狭窄程度相关。我们的发现表明PTPN14是SMC的关键调节剂,促进新生内膜增生。由PTPN14去磷酸化的PDGFRβY692作为控制PDGFRβ活化的自我抑制位点。
    Smooth muscle cell (SMC) phenotypic modulation, primarily driven by PDGFRβ signaling, is implicated in occlusive cardiovascular diseases. However, the promotive and restrictive regulation mechanism of PDGFRβ and the role of protein tyrosine phosphatase non-receptor type 14 (PTPN14) in neointimal hyperplasia remain unclear. Our study observes a marked upregulation of PTPN14 in SMCs during neointimal hyperplasia. PTPN14 overexpression exacerbates neointimal hyperplasia in a phosphatase activity-dependent manner, while SMC-specific deficiency of PTPN14 mitigates this process in mice. RNA-seq indicates that PTPN14 deficiency inhibits PDGFRβ signaling-induced SMC phenotypic modulation. Moreover, PTPN14 interacts with intracellular region of PDGFRβ and mediates its dephosphorylation on Y692 site. Phosphorylation of PDGFRβY692 negatively regulates PDGFRβ signaling activation. The levels of both PTPN14 and phospho-PDGFRβY692 are correlated with the degree of stenosis in human coronary arteries. Our findings suggest that PTPN14 serves as a critical modulator of SMCs, promoting neointimal hyperplasia. PDGFRβY692, dephosphorylated by PTPN14, acts as a self-inhibitory site for controlling PDGFRβ activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    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
    血小板衍生生长因子(PDGF)是肺动脉高压(PAH)中与肺血管重构相关的重要细胞因子之一。PDGF受体(PDGFR)抑制在临床试验中对PAH产生治疗作用,但是严重的副作用需要停用现有药物。在这项研究中,开发了一种新型的高选择性PDGFR抑制剂WQ-C-401,研究其对PAH中PDGFR信号通路和肺血管重构的影响。细胞增殖测定和PDGFRα/β磷酸化的蛋白质印迹分析显示WQ-C-401以浓度依赖性方式抑制PDGFR介导的细胞增殖测定并抑制PDGFR磷酸化。DiscoverX的KinomeScanTM技术证实了WQ-C-401良好的运动学选择性(PDGFR的S评分(1)=(0.01))。在野百合碱(MCT)诱导的PAH大鼠中,WQ-C-401(25、50、100mg/kg/d)或伊马替尼(50mg/kg/d,阳性对照)显著降低右心室收缩压(RVSP)。组织学分析表明,WQ-C-401通过减少肌肉化和纤维化抑制肺血管重塑,以及减轻MCT治疗大鼠的右心室肥厚。此外,WQ-C-401抑制MCT诱导的细胞过度增殖和肺动脉周围CD68+巨噬细胞浸润。体外,WQ-C-401抑制PDGF-BB诱导的人肺动脉平滑肌细胞(PASMC)增殖和迁移。此外,westernblot分析表明,WQ-C-401协同依赖性地抑制PDGF-BB诱导的ERK1/2和PDGFRβY751磷酸化,减少胶原蛋白Ⅰ的合成,增加PASMC中α-平滑肌肌动蛋白(α-SMA)的表达。总的来说,我们的结果表明,WQ-C-401是一种选择性和有效的PDGFR抑制剂,它可能是通过预防肺血管重塑治疗PAH的有前景的药物.
    Platelet-derived growth factor (PDGF) is one of the most important cytokines associated with pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). PDGF receptor (PDGFR) inhibition exerted therapeutic effects on PAH in clinical trials, but serious side effects warrant the withdrawal of existing drugs. In this study, a novel highly selective PDGFR inhibitor WQ-C-401 was developed, and its effects on PDGFR signaling pathway and pulmonary vascular remodeling in PAH were investigated. Cell proliferation assays and Western blot analysis of PDGFRα/β phosphorylation showed that WQ-C-401 inhibited PDGFR-mediated cell proliferation assay and suppressed PDGFR phosphorylation in a concentration-dependent manner. DiscoverX\'s KinomeScanTM technology confirmed the good kinome selectivity of WQ-C-401 (S score (1) of PDGFR = (0.01)). In monocrotaline (MCT)-induced PAH rats, intragastric administration of WQ-C-401 (25, 50, 100 mg/kg/d) or imatinib (50 mg/kg/d, positive control) significantly decreased right ventricular systolic pressure (RVSP). Histological analysis demonstrated that WQ-C-401 inhibited pulmonary vascular remodeling by reducing muscularization and fibrosis, as well as alleviated right ventricular hypertrophy in MCT-treated rats. In addition, WQ-C-401 suppressed MCT-induced cell hyperproliferation and CD68+ macrophage infiltration around the pulmonary artery. In vitro, WQ-C-401 inhibited PDGF-BB-induced proliferation and migration of human pulmonary arterial smooth muscle cells (PASMCs). Moreover, Western blot analysis showed that WQ-C-401 concertration-dependently inhibited PDGF-BB-induced phosphorylation of ERK1/2 and PDGFRβ Y751, decreased collagen Ⅰ synthesis and increased alpha smooth muscle actin (α-SMA) expression in PASMCs. Collectively, our results suggest that WQ-C-401 is a selective and potent PDGFR inhibitor which could be a promising drug for the therapeutics of PAH by preventing pulmonary vascular remodeling.
    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
    背景:肝纤维化是慢性肝病中普遍存在的病理过程,其特征是细胞外基质(ECM)沉积过多和血管生成异常。值得注意的是,肝星状细胞(HSC)是ECM的主要来源。活化的HSC不仅分泌大量促纤维化细胞因子,而且被赋予促血管生成表型以促进病理性血管生成。因此,HSC的靶向调节已成为解决肝纤维化的关键策略。羟基红花黄色素A(HSYA)是一种药食同源色素,具有良好的药理活性。然而,HSYA抗肝纤维化的确切机制尚不清楚.
    目的:本研究的目的是阐明HSYA对肝纤维化和病理性血管生成的影响,以及体外和体内研究的潜在机制。
    方法:采用MTT法研究HSYA对TGF-β1诱导的HSCs和VEGFA诱导的内皮细胞的作用及机制。EdU细胞增殖试验,细胞划痕试验,Elisa分析,免疫荧光测定,分子对接,细胞转染试验,蛋白质印迹分析和RT-qPCR分析。在CCl4诱导的肝纤维化小鼠模型中,H&E,Masson,采用天狼星红染色进行组织病理学观察。采用生化试剂盒检测血清转氨酶活性和肝脏生化指标。免疫组织化学,荧光原位杂交(FISH),采用westernblot分析和RT-qPCR分析来确定HSYA在体内的作用机制。
    结果:这里,我们的发现证实了HSYA抑制增殖,HSC的迁移和激活,细胞活力的降低证明了这一点,相对迁移率,EdU染色强度,以及体外促纤维化mRNA和蛋白质表达。机械上,HSYA通过部分沉默活化的HSC中的PDGFRB发挥抗纤维化和抗血管生成作用,从而破坏PDGFRB/MEK/ERK信号转导,抑制HIF-1α的表达,VEGFA和VEGFR2蛋白。重要的是,PDGFRB是miR-29a-3p的靶基因。在用miR-29a-3p抑制剂转染的TGF-β1诱导的HSC中,用HSYA处理逆转miR-29a-3p的下调并拮抗PDGFRB信号通路。与我们的体外研究一致,HSYA通过降低血清ALT和AST水平在CCl4诱导的肝纤维化小鼠中表现出良好的肝保护作用,降低四个纤维化指标的含量(HA,PIIIP,ColIV和LN)和羟脯氨酸,并抑制TGF-β1/TGFBR信号通路。在机制方面,HSYA通过使PDGFRB信号通路失活并损害CD31的阳性表达,减轻纤维化肝病理性血管生成。随后,FISH结果进一步证实HSYA影响HSC的活化和通过miR-29a-3p的同时上调和α-SMA和VEGFA的下调而实现的血管生成。此外,用HSYA治疗也在HSC和内皮细胞之间建立了联系,通过抑制内皮细胞的异常增殖得到支持。
    结论:从根本上说,目前的研究表明,HSYA通过抑制HSC介导的促纤维化和促血管生成过程来改善肝纤维化,这取决于HSYA对miR-29a-3p/PDGFRB轴的调节作用。这些发现提供了令人信服的证据支持HSYA作为肝纤维化治疗剂的潜力。
    BACKGROUND: Liver fibrosis is a prevalent pathological process in chronic liver diseases characterized by excessive extracellular matrix (ECM) deposition and abnormal angiogenesis. Notably, hepatic stellate cells (HSCs) are the primary source of ECM. Activated HSCs not only secrete numerous pro-fibrotic cytokines but also are endowed with a pro-angiogenic phenotype to promote pathological angiogenesis. Therefore, targeted modulation of HSCs has emerged as a pivotal strategy for addressing liver fibrosis. Hydroxysafflor yellow A (HSYA) is a homology of medicine and food colourant with good pharmacological activity. However, the precise mechanisms of HSYA against liver fibrosis remain unclear.
    OBJECTIVE: The objective of this study was to elucidate the impact of HSYA on liver fibrosis and pathological angiogenesis, as well as the underlying mechanisms in vitro and in vivo studies.
    METHODS: The efficacy and mechanisms of HSYA on TGF-β1-induced HSCs and VEGFA-induced endothelial cells were investigated by MTT assay, EdU cell proliferation assay, cell scratch assay, Elisa assay, immunofluorescence assay, molecular docking, cell transfection assay, western blot analysis and RT-qPCR analysis. In CCl4-induced liver fibrosis mice model, H&E, Masson, and Sirius red staining were used to observe histopathology. Serum transaminase activity and liver biochemical indexes were tested by biochemical kit. Immunohistochemical, fluorescence in situ hybridization (FISH), western blot analysis and RT-qPCR analysis were implemented to determine the mechanism of HSYA in vivo.
    RESULTS: Herein, our findings confirmed that HSYA inhibited the proliferation, migration and activation of HSCs, as evidenced by a reduction in cell viability, relative migration rate, EdU staining intensity, and pro-fibrotic mRNAs and proteins expression in vitro. Mechanistically, HSYA played an anti-fibrotic and anti-angiogenic role by partially silencing PDGFRB in activated HSCs, thereby disrupting PDGFRB/MEK/ERK signal transduction and inhibiting the expression of HIF-1α, VEGFA and VEGFR2 proteins. Importantly, PDGFRB was a target gene of miR-29a-3p. Treatment with HSYA reversed the down-regulation of miR-29a-3p and antagonized PDGFRB signaling pathway in TGF-β1-induced HSCs transfected with miR-29a-3p inhibitor. Consistent with our in vitro study, HSYA exhibited a good hepatoprotective effect in CCl4-induced liver fibrosis mice by reducing serum ALT and AST levels, decreasing the contents of four fibrosis indicators (HA, PIIIP, ColIV and LN) and hydroxyproline, and inhibiting the TGF-β1/TGFBR signaling pathway. In terms of mechanisms, HSYA alleviated pathological angiogenesis in fibrotic liver by deactivating PDGFRB signaling pathway and impairing the positive expression of CD31. Subsequently, FISH results further corroborated HSYA affected the activation of HSCs and angiogenesis achieved by the concurrent upregulation of miR-29a-3p and downregulation of α-SMA and VEGFA. Additionally, treatment with HSYA also forged a link between HSCs and endothelial cells, as supported by inhibiting the aberrant proliferation of endothelial cells.
    CONCLUSIONS: Fundamentally, the current study has illustrated that HSYA ameliorates liver fibrosis by repressing HSCs-mediated pro-fibrotic and pro-angiogenic processes, which is contingent upon the regulatory effect of HSYA on the miR-29a-3p/PDGFRB axis. These findings provide compelling evidence bolstering the potential of HSYA as a therapeutic agent in liver fibrosis.
    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
    恶性黑色素瘤由于其侵袭性转移行为和对当前治疗的有限反应而提出了巨大的挑战。为了解决这个问题,我们的研究使用恶性黑色素瘤细胞和人脐静脉内皮细胞探讨了安洛替尼对血管生成和血管生成拟态的影响.评估管状结构的形成,细胞增殖,迁移,入侵,和血管生成中的关键信号分子,我们证明了安洛替尼对肾小管结构具有剂量依赖性抑制作用,并有效抑制两种细胞类型的细胞生长和侵袭.此外,在小鼠异种移植模型中,安洛替尼治疗导致肿瘤生长和血管密度降低.值得注意的是,VEGFR-2、FGFR-1、PDGFR-β的下调,PI3K强调了安洛替尼的多靶向抗肿瘤活性。我们的发现强调了安洛替尼在靶向血管生成和血管生成拟态方面的治疗潜力,有助于开发对抗恶性黑色素瘤的新策略。
    Malignant melanoma presents a formidable challenge due to its aggressive metastatic behavior and limited response to current treatments. To address this, our study delves into the impact of anlotinib on angiogenesis and vasculogenic mimicry using malignant melanoma cells and human umbilical vein endothelial cells. Evaluating tubular structure formation, cell proliferation, migration, invasion, and key signaling molecules in angiogenesis, we demonstrated that anlotinib exerts a dose-dependent inhibition on tubular structures and effectively suppresses cell growth and invasion in both cell types. Furthermore, in a mouse xenograft model, anlotinib treatment resulted in reduced tumor growth and vascular density. Notably, the downregulation of VEGFR-2, FGFR-1, PDGFR-β, and PI3K underscored the multitargeted antitumor activity of anlotinib. Our findings emphasize the therapeutic potential of anlotinib in targeting angiogenesis and vasculogenic mimicry, contributing to the development of novel strategies for combating malignant melanoma.
    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
    对于准确的药物筛选和配体-蛋白质亲和力分析,膜受体固定的准确方向和稳定构象尤为重要。然而,仍然存在与(1)传统重组相关的挑战,净化,和膜受体的固定,这是耗时和劳动密集型的;(2)固定相的方向不容易控制。在这里,开发了一种新颖的一步合成和定向固定化膜受体亲和层析(oSOMAC)方法,以实现针对膜受体特定域的高通量和准确的药物筛选。我们使用Strep-tagII作为通过CFPS融合到血小板衍生生长因子受体β(PDGFRβ)中的非共价固定标签,同时,分批制备Strep-Tactin修饰的整体柱。oSOMAC的优点如下:(1)靶向膜受体在1-2h内可以独立于活细胞表达;(2)膜受体的取向可以灵活控制,活性部位可以准确暴露;(3)靶向膜受体可以合成,纯化,一步定向固定在整体柱上。因此,三种潜在的PDGFRβ胞内结构域靶向配体:丹参酮IIA(TanIIA),羟基丹参酮IIA,通过oSOMAC从丹参提取物中成功筛选出脱氢丹参酮IIA。药理学实验和分子对接进一步证明,TanIIA可以通过靶向PDGFRβ的蛋白激酶结构域来减弱肝星状细胞的活化,KD值为9.7μM。最终,新的oSOMAC方法为准确的药物筛选和相互作用分析提供了原始见解,可应用于其他膜受体。
    Accurate orientations and stable conformations of membrane receptor immobilization are particularly imperative for accurate drug screening and ligand-protein affinity analysis. However, there remain challenges associated with (1) traditional recombination, purification, and immobilization of membrane receptors, which are time-consuming and labor-intensive; (2) the orientations on the stationary phase are not easily controlled. Herein, a novel one-step synthesis and oriented-immobilization membrane-receptor affinity chromatography (oSOMAC) method was developed to realize high-throughput and accurate drug screening targeting specific domains of membrane receptors. We employed Strep-tag II as a noncovalent immobilization tag fused into platelet-derived growth factor receptor β (PDGFRβ) through CFPS, and meanwhile, the Strep-Tactin-modified monolithic columns are prepared in batches. The advantages of oSOMAC are as follows: (1) targeted membrane receptors can be expressed independent of living cell within 1-2 h; (2) orientation of membrane receptors can be flexibly controlled and active sites can expose accurately; and (3) targeted membrane receptors can be synthesized, purified, and orientation-immobilized on monolithic columns in one step. Accordingly, three potential PDGFRβ intracellular domain targeted ligands: tanshinone IIA (Tan IIA), hydroxytanshinone IIA, and dehydrotanshinone IIA were successfully screened out from Salvia miltiorrhiza extract through oSOMAC. Pharmacological experiments and molecular docking further demonstrated that Tan IIA could attenuate hepatic stellate cells activation by targeting the protein kinase domain of PDGFRβ with a KD value of 9.7 μM. Ultimately, the novel oSOMAC method provides an original insight for accurate drug screening and interaction analysis which can be applied in other membrane receptors.
    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: Case Reports
    这里,我们介绍了一例罕见的骨髓增殖性肿瘤(MPN)伴嗜酸性粒细胞增多,同时伴有BCR::ABL1和PDGFRB重排,构成分类困境。患者的临床和实验室特征提示慢性粒细胞白血病(CML)和伴有嗜酸性粒细胞增多和酪氨酸激酶基因融合(MLN-TK)的骨髓/淋巴肿瘤,强调与重叠表型相关的诊断挑战。尽管复杂,伊马替尼治疗迅速实现深度分子缓解,强调酪氨酸激酶抑制剂在这种情况下的治疗效果。此外,该患者对伊马替尼的反应迅速达到深度缓解与MLN-TK伴PDGFRB重排患者中观察到的非常相似.需要进一步的研究来阐明驱动MPN中多种致癌重排共存的潜在机制,并优化这些复杂病例的治疗策略。
    Here, we present a rare case of myeloproliferative neoplasms (MPN) with eosinophilia harboring both BCR::ABL1 and PDGFRB rearrangements, posing a classification dilemma. The patient exhibited clinical and laboratory features suggestive of chronic myeloid leukemia (CML) and myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions (MLN-TK), highlighting the diagnostic challenges associated with overlapping phenotypes. Despite the complexity, imatinib treatment swiftly achieved deep molecular remission, underscoring the therapeutic efficacy of tyrosine kinase inhibitors in such scenarios. Furthermore, the rapid attainment of deep remission by this patient in response to imatinib closely resembles that observed in MLN-TK patients with PDGFRB rearrangements. Further research is warranted to elucidate the underlying mechanisms driving the coexistence of multiple oncogenic rearrangements in MPNs and to optimize therapeutic strategies for these complex cases.
    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
    血小板衍生生长因子受体β(PDGFRβ)在小鼠造血中起着至关重要的作用。黄芩素(BAI),一种天然存在的类黄酮,可以通过抗氧化来减轻疾病损伤,抗凋亡,和抗炎机制。然而,BAI是否通过PDGFRβ减弱小鼠造血细胞的氧化损伤仍未研究。在这项研究中,我们利用叔丁基过氧化氢(TBHP)诱导的BaF3细胞损伤模型和电离辐射(IR)诱导的小鼠损伤模型来研究PDGFRβ的存在或不存在对BAI药理作用的影响。此外,通过分子对接和动力学模拟表征了BAI-PDGFRβ相互作用。结果表明,特定浓度的BAI导致细胞活力增加,降低活性氧(ROS)含量,核因子红系2相关因子2(NRF2)表达上调,及其下游靶基因血红素加氧酶1(HO-1)和NAD(P)H醌脱氢酶1(NQO1),在表达PDGFRβ质粒并经历损伤的细胞中激活蛋白激酶B(AKT)通路。同样,BAI升高谱系-Sca1+cKIT+(LSK)细胞比例,促进造血恢复,PDGFRβ+/+小鼠NRF2介导的抗氧化反应增强。然而,尽管使用了BAI,PDGFRβ敲除小鼠(PDGFRβ-/-)表现出比全身照射(TBI)组低的LSK比例和低的抗氧化能力。此外,我们在分子水平上证明了BAI和PDGFRβ之间的相互作用。总的来说,我们的结果表明,BAI通过调节PDGFRβ减轻氧化应激损伤,并有助于促进造血细胞恢复.
    Platelet-derived growth factor receptor β (PDGFRβ) plays a crucial role in murine haematopoiesis. Baicalein (BAI), a naturally occurring flavonoid, can alleviate disease damage through anti-oxidative, anti-apoptotic, and anti-inflammatory mechanisms. However, whether BAI attenuates oxidative damage in murine haematopoietic cells by PDGFRβ remains unexplored. In this study, we utilized a tert-butyl hydroperoxide (TBHP)-induced BaF3 cell injury model and an ionising radiation (IR)-induced mice injury model to investigate the impact of the presence or absence of PDGFRβ on the pharmacological effects of BAI. In addition, the BAI-PDGFRβ interaction was characterized by molecular docking and dynamics simulations. The results show that a specific concentration of BAI led to increased cell viability, reduced reactive oxygen species (ROS) content, upregulated nuclear factor erythroid 2-related factor 2 (NRF2) expression, and its downstream target genes heme oxygenase 1 (HO-1) and NAD(P)H Quinone Dehydrogenase 1 (NQO1), and activated protein kinase B (AKT) pathway in cells expressing PDGFRβ plasmid and experiencing damage. Similarly, BAI elevated lineage-Sca1+cKIT+ (LSK) cell proportion, promoted haematopoietic restoration, enhanced NRF2-mediated antioxidant response in PDGFRβ+/+ mice. However, despite BAI usage, PDGFRβ knockout mice (PDGFRβ-/-) showed lower LSK proportion and less antioxidant capacity than the total body irradiation (TBI) group. Furthermore, we demonstrated an interaction between BAI and PDGFRβ at the molecular level. Collectively, our results indicate that BAI attenuates oxidative stress injury and helps promote haematopoietic cell recovery through regulation of PDGFRβ.
    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
    背景:舒尼替尼已成为晚期或转移性透明细胞肾细胞癌(ccRCC)的主要治疗方法,因为它显著改善了患者的平均生存时间。然而,舒尼替尼的耐药性和不良反应对其临床获益构成挑战.
    方法:研究ccRCC中与舒尼替尼敏感性和耐药性相关的差异表达基因(DEGs)。细胞计数试剂盒-8,平板集落形成,采用流式细胞术和皮下移植瘤模型检测来探讨PDZK1对ccRCC的影响。通过蛋白质印迹对其分子机制进行了进一步的研究,免疫共沉淀,免疫荧光共定位和免疫组织化学染色。
    结果:我们阐明了PDZK1在舒尼替尼耐药的ccRCC标本中显著下调,和PDZK1通过与PDGFR-β的相互作用负调节PDGFR-β的磷酸化及其下游途径的激活。PDZK1的低水平失调导致细胞增殖的抑制不足,肿瘤生长,对舒尼替尼治疗不敏感。值得注意的是,我们的临床前研究表明,miR-15bantagomirs通过上调PDZK1水平增强舒尼替尼对ccRCC细胞的细胞毒性作用,表明它们在克服舒尼替尼耐药性方面的潜力。
    结论:我们的发现确立了miR-15b/PDZK1/PDGFR-β轴作为ccRCC患者对舒尼替尼治疗反应的一个有前景的治疗靶点和新的预测因子。
    BACKGROUND: Sunitinib has emerged as the primary treatment for advanced or metastatic clear cell renal cell carcinoma (ccRCC) due to its significant improvement in patients\' average survival time. However, drug resistance and adverse effects of sunitinib pose challenges to its clinical benefits.
    METHODS: The differentially expressed genes (DEGs) associated with sunitinib sensitivity and resistance in ccRCC were investigated. Cell counting kit-8, plate colony formation, flow cytometry and subcutaneous xenograft tumor model assays were employed to explore the effects of PDZK1 on ccRCC. Further research on the molecular mechanism was conducted through western blot, co-immunoprecipitation, immunofluorescence co-localization and immunohistochemical staining.
    RESULTS: We elucidated that PDZK1 is significantly downregulated in sunitinib-resistant ccRCC specimens, and PDZK1 negatively regulates the phosphorylation of PDGFR-β and the activation of its downstream pathways through interaction with PDGFR-β. The dysregulated low levels of PDZK1 contribute to inadequate inhibition of cell proliferation, tumor growth, and insensitivity to sunitinib treatment. Notably, our preclinical investigations showed that miR-15b antagomirs enhance sunitinib cytotoxic effects against ccRCC cells by upregulating PDZK1 levels, suggesting their potential in overcoming sunitinib resistance.
    CONCLUSIONS: Our findings establish the miR-15b/PDZK1/PDGFR-β axis as a promising therapeutic target and a novel predictor for ccRCC patients\' response to sunitinib treatment.
    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
    环状RNA(circularRNAs)代表一类共价闭合的,单链RNA,并与癌症进展有关。N6-甲基腺苷(m6A)甲基化是癌细胞中普遍存在的RNA修饰。越来越多的证据表明,m6A可以介导circRNAs在癌症生物学中的作用。相比之下,子宫内膜癌(EC)进展过程中m6A和circRNA的转录后系统仍不清楚.当前的研究鉴定了一种具有m6A修饰的新型circRNA,hsa_circ_0084582(circhd7),在EC组织中上调。功能上,发现circCHD7促进EC细胞的增殖。机械上,circCHD7与胰岛素样生长因子2mRNA结合蛋白(IGF2BP2)相互作用以增强其富集。此外,circCHD7以m6A依赖性方式增加血小板衍生生长因子受体β(PDGFRB)的mRNA稳定性,从而增强其表达。此外,circCHD7/IGF2BP2/PDGFRB轴激活JAK/STAT信号通路,促进EC细胞增殖。总之,这些发现为circRNA介导的m6A修饰的调控提供了新的见解,新的“circCHD7-PDGFRB”调控模型为circCHD7作为EC治疗的潜在靶点提供了新的视角。
    Circular RNAs (circRNAs) represent a class of covalently closed, single-stranded RNAs and have been linked to cancer progression. N6-methyladenosine (m6A) methylation is a ubiquitous RNA modification in cancer cells. Increasing evidence suggests that m6A can mediate the effects of circRNAs in cancer biology. In contrast, the post-transcriptional systems of m6A and circRNA in the progression of endometrial cancer (EC) remain obscure. The current study identified a novel circRNA with m6A modification, hsa_circ_0084582 (circCHD7), which was upregulated in EC tissues. Functionally, circCHD7 was found to promote the proliferation of EC cells. Mechanistically, circCHD7 interacted with insulin-like growth factor 2 mRNA-binding protein (IGF2BP2) to amplify its enrichment. Moreover, circCHD7 increased the mRNA stability of platelet-derived growth factor receptor beta (PDGFRB) in an m6A-dependent manner, thereby enhancing its expression. In addition, the circCHD7/IGF2BP2/PDGFRB axis activated the JAK/STAT signaling pathway and promoted EC cell proliferation. In conclusion, these findings provide new insights into the regulation of circRNA-mediated m6A modification, and the new \"circCHD7-PDGFRB\" model of regulation offers new perspectives on circCHD7 as a potential target for EC therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号