TGF‐β

TGF - β
  • 文章类型: Journal Article
    在临床环境中,解决大骨缺损仍然是骨科医生面临的重大挑战。使用遗传修饰的骨髓间充质干细胞(BMSCs)已成为这些治疗的非常有前途的方法。信号肽-CUB-EGF域含蛋白3(SCUBE3)是一种多功能分泌型糖蛋白,其在人hBMSCs中的作用尚不清楚。本研究采用多种实验方法阐明了SCUBE3在体外影响hBMSCs成骨分化的潜在机制。此外,SCUBE3与多孔GeLMA微球的治疗效果,使用小鼠骨缺损模型进行体内评估。我们的发现表明,在hBMSCs成骨分化的早期,SCUBE3水平显着增加。降低SCUBE3水平会阻碍这种分化。过表达SCUBE3可升高成骨基因和蛋白质水平并增强钙沉积。此外,用重组人SCUBE3(rhSCUBE3)蛋白治疗可增强BMP2和TGF-β的表达,hBMSCs中激活的线粒体自噬,改善氧化应激,并通过SMAD磷酸化恢复成骨功能。在体内,GELMA/OE处理有效地加速了小鼠的骨愈合。总之,SCUBE3通过激活BMP2/TGF-β信号通路促进hBMSCs成骨分化和线粒体自噬。当与工程水凝胶细胞疗法结合时,它可以为广泛骨缺损的临床治疗提供有价值的指导。
    In clinical settings, addressing large bone defects remains a significant challenge for orthopedic surgeons. The use of genetically modified bone marrow mesenchymal stem cells (BMSCs) has emerged as a highly promising approach for these treatments. Signal peptide-CUB-EGF domain-containing protein 3 (SCUBE3) is a multifunctional secreted glycoprotein, the role of which remains unclear in human hBMSCs. This study used various experimental methods to elucidate the potential mechanism by which SCUBE3 influences osteogenic differentiation of hBMSCs in vitro. Additionally, the therapeutic efficacy of SCUBE3, in conjunction with porous GeLMA microspheres, was evaluated in vivo using a mouse bone defect model. Our findings indicate that SCUBE3 levels increase significantly during early osteogenic differentiation of hBMSCs, and that reducing SCUBE3 levels can hinder this differentiation. Overexpressing SCUBE3 elevated osteogenesis gene and protein levels and enhanced calcium deposition. Furthermore, treatment with recombinant human SCUBE3 (rhSCUBE3) protein boosted BMP2 and TGF-β expression, activated mitophagy in hBMSCs, ameliorated oxidative stress, and restored osteogenic function through SMAD phosphorylation. In vivo, GELMA/OE treatment effectively accelerated bone healing in mice. In conclusion, SCUBE3 fosters osteogenic differentiation and mitophagy in hBMSCs by activating the BMP2/TGF-β signaling pathway. When combined with engineered hydrogel cell therapy, it could offer valuable guidance for the clinical management of extensive bone defects.
    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
    转化生长因子β(TGF-β)信号通路和N6-甲基腺苷(m6A)mRNA修饰在肝细胞癌(HCC)的进展中起重要作用。然而,TGF-β和m6A在肝细胞癌(HCC)中的关系尚不清楚。这里,发现TGF-β可以促进METTL3的液相分离,进一步导致ITIH1mRNA稳定性降低。作为一种分泌蛋白,ITIH1可以作为整合素α5β1的配体拮抗纤连蛋白,诱导粘着斑激酶信号通路的抑制,并抑制HCC的进展。在临床前模型(小鼠模型,患者来源的类器官,患者来源的异种移植物),纯化的重组ITIH1(r-ITIH1)蛋白可靶向治疗肝癌。更重要的是,r-ITIH1可以在TGF-β抑制剂靶向HCC中发挥协同作用。揭示了TGF-β和m6A修饰的下游ITIH1调控机制,ITIH1可以作为肝癌的潜在目标。
    Both the transforming growth factor beta (TGF-β) signaling pathway and N6-methyladenosine (m6A) modification for mRNA play an important role in hepatocellular carcinoma (HCC) progression. However, the relationship between TGF-β and m6A in hepatocellular carcinoma (HCC) remains unclear. Here, it is found that TGF-β can promote the liquid phase separation of METTL3, which further leads to the reduction of mRNA stability of ITIH1. As a secreted protein, ITIH1 can act as a ligand of integrin α5β1 to antagonize fibronectin, induce the inhibition of focal adhesion kinase signaling pathway, and inhibit the progression of HCC. In the preclinical model (mouse model, patient-derived organoid, patient-derived xenografts), purified recombinant ITIH1 (r-ITIH1) protein can be targeted for HCC. More importantly, r-ITIH1 can play a synergistic role in targeting HCC with TGF-β inhibitor. The downstream ITIH1 regulatory mechanism of TGF-β and m6A modification is revealed, and ITIH1 can be translational as a potential target for HCC.
    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
    TGF-β信号通路异常可导致结直肠癌(CRC)的侵袭表型,导致预后不良。在TGF-β相关基因的基础上建立有效的预后因子对准确识别CRC患者的风险至关重要。
    我们从数据库和以前的文献中对CRC患者的TGF-β相关基因进行了差异分析,以获得TGF-β相关的差异表达基因(TRDEGs)。利用LASSO-Cox回归建立基于TRDEGs的CRC预后特征模型。使用两个GEO验证集对模型进行了验证。采用Wilcoxon秩和检验模型与临床因素的相关性。使用ESTIMATE算法和ssGSEA和肿瘤突变负荷(TMB)分析来分析高风险(HR)和低风险(LR)组的免疫状况和突变负荷。利用CellMiner数据库来鉴定对特征基因具有高敏感性的治疗药物。
    我们建立了具有良好预测准确性的六基因风险预后模型,独立预测CRC患者的预后。由于较高的免疫浸润和TMB,HR组更有可能经历免疫疗法益处。特征基因TGFB2能够抑制XAV-939、星孢菌素、和达沙替尼,但促进药物如CUDC-305和CUDC-305的副产品的疗效。同样,RBL1可以抑制氟奋乃静和咪喹莫特的药物作用,但可以促进伊罗芬的药物作用。
    根据TGF-β相关基因开发了CRC风险预后特征,为CRC患者的风险和进一步的治疗选择提供参考。
    UNASSIGNED: Aberrant TGF-β signaling pathway can lead to invasive phenotype of colorectal cancer (CRC), resulting in poor prognosis. It is pivotal to develop an effective prognostic factor on the basis of TGF-β-related genes to accurately identify risk of CRC patients.
    UNASSIGNED: We performed differential analysis of TGF-β-related genes in CRC patients from databases and previous literature to obtain TGF-β-related differentially expressed genes (TRDEGs). LASSO-Cox regression was utilized to build a CRC prognostic feature model based on TRDEGs. The model was validated using two GEO validation sets. Wilcoxon rank-sum test was utilized to test correlation of model with clinical factors. ESTIMATE algorithm and ssGSEA and tumor mutation burden (TMB) analysis were used to analyze immune landscape and mutation burden of high-risk (HR) and low-risk (LR) groups. CellMiner database was utilized to identify therapeutic drugs with high sensitivity to the feature genes.
    UNASSIGNED: We established a six-gene risk prognostic model with good predictive accuracy, which independently predicted CRC patients\' prognoses. The HR group was more likely to experience immunotherapy benefits due to higher immune infiltration and TMB. The feature gene TGFB2 could inhibit the efficacy of drugs such as XAV-939, Staurosporine, and Dasatinib, but promote the efficacy of drugs such as CUDC-305 and by-product of CUDC-305. Similarly, RBL1 could inhibit the drug action of Fluphenazine and Imiquimod but promote that of Irofulven.
    UNASSIGNED: A CRC risk prognostic signature was developed on basis of TGF-β-related genes, which provides a reference for risk and further therapeutic selection of CRC patients.
    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
    透明细胞肾细胞癌(ccRCC)代表与预后最差相关的高度异质性肾脏恶性肿瘤。晚期ccRCC肿瘤的转移潜力非常高,带来了重大的临床挑战。迫切需要开发新的治疗方法来解决ccRCC转移。最近的研究表明GBP2与肿瘤免疫之间存在潜在的关联。然而,GBP2在ccRCC进展中的确切功能作用尚不清楚.本研究揭示了GBP2和M2巨噬细胞之间的强相关性。具体来说,我们的研究结果表明,GBP2的抑制显著阻碍了ccRCC细胞的迁移和侵袭能力.我们观察到M2巨噬细胞的存在可以逆转GBP2敲低对肿瘤细胞迁移和侵袭的影响。机械上,我们证明,M2巨噬细胞通过分泌白细胞介素-10(IL-10)和转化生长因子-β(TGF-β)促进肿瘤细胞GBP2/p-STAT3和p-ERK轴的表达,从而显著增强肿瘤细胞的迁移和侵袭能力。同时,我们已经确定GBP2通过刺激白细胞介素-18(IL-18)的分泌促进巨噬细胞向M2表型的极化.总之,我们的研究预计GBP2/IL-18/M2巨噬细胞/IL-10和TGF-β/GBP2,p-STAT3,p-ERK环在ccRCC转移中起着至关重要的作用。我们研究的集体发现强调了GBP2在肿瘤免疫中的重要作用,并强调了调节GBP2作为靶向ccRCC转移的有希望的治疗策略的潜力。
    Clear cell renal cell carcinoma (ccRCC) represents a highly heterogeneous kidney malignancy associated with the poorest prognosis. The metastatic potential of advanced ccRCC tumors is notably high, posing significant clinical challenges. There is an urgent imperative to develop novel therapeutic approaches to address ccRCC metastasis. Recent investigations indicated a potential association between GBP2 and tumor immunity. However, the precise functional role of GBP2 in the progression of ccRCC remains poorly understood. The present study revealed a strong correlation between GBP2 and M2 macrophages. Specifically, our findings demonstrated that the inhibition of GBP2 significantly impedes the migratory and invasive capabilities of ccRCC cells. We observed that the presence of M2 macrophages can reverse the effects of GBP2 knockdown on tumor cell migration and invasion. Mechanistically, we demonstrated that M2 macrophages promote the expression of the GBP2/p-STAT3 and p-ERK axis in tumor cells through the secretion of interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), thereby substantially enhancing the migratory and invasive capacities of the tumor cells. Simultaneously, we have identified that GBP2 promotes the polarization of macrophages to the M2 phenotype by stimulating the secretion of interleukin-18 (IL-18). In summary, our investigation anticipates that the GBP2/IL-18/M2 macrophages/IL-10 and the TGF-β/GBP2, p-STAT3, p-ERK loop plays a crucial role in ccRCC metastasis. The collective findings from our research underscore the significant role of GBP2 in tumor immunity and emphasize the potential for modulating GBP2 as a promising therapeutic strategy for targeting ccRCC metastasis.
    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
    来自五个生命王国的生物使用矿物质来硬化组织并制造牙齿,贝壳和骨骼,在生物矿化过程中。海胆幼虫骨骼是研究生物矿化及其进化的生物学调控的优秀系统。控制海胆骨骼生成的基因调节网络(GRN)非常详细,与控制脊椎动物血管化的GRN相似,而与驱动脊椎动物骨形成的GRN截然不同。然而,转化生长因子β(TGF-β)信号调节海胆和脊椎动物骨骼形成。这里,我们研究了地中海海胆物种中TGF-β的上游调控和转录目标,Paracentrotuslividus.TGF-βRII在血管内皮生长因子(VEGF)信号下游的骨架细胞中短暂活跃,在P.lividus.TGF-βRII活性的连续扰动显著损害骨骼伸长和关键成骨基因的表达。骨骼启动后TGF-βRII的扰动导致骨骼伸长的延迟和基因表达的微小变化。TGF-β靶标与其在脊椎动物骨形成过程中的转录靶标不同,这表明TGF-β在这两个门的生物矿化中的作用是趋同进化的结果。
    Organisms from the five kingdoms of life use minerals to harden their tissues and make teeth, shells and skeletons, in the process of biomineralization. The sea urchin larval skeleton is an excellent system to study the biological regulation of biomineralization and its evolution. The gene regulatory network (GRN) that controls sea urchin skeletogenesis is known in great details and shows similarity to the GRN that controls vertebrates\' vascularization while it is quite distinct from the GRN that drives vertebrates\' bone formation. Yet, transforming growth factor beta (TGF-β) signaling regulates both sea urchin and vertebrates\' skeletogenesis. Here, we study the upstream regulation and identify transcriptional targets of TGF-β in the Mediterranean Sea urchin species, Paracentrotus lividus. TGF-βRII is transiently active in the skeletogenic cells downstream of vascular endothelial growth factor (VEGF) signaling, in P. lividus. Continuous perturbation of TGF-βRII activity significantly impairs skeletal elongation and the expression of key skeletogenic genes. Perturbation of TGF-βRII after skeletal initiation leads to a delay in skeletal elongation and minor changes in gene expression. TGF-β targets are distinct from its transcriptional targets during vertebrates\' bone formation, suggesting that the role of TGF-β in biomineralization in these two phyla results from convergent evolution.
    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
    肝星状细胞(HSC)活化是肝纤维化(LF)的重要病理进程。调节HSC活化和LF的分子机制尚不完全清楚。这里,我们探讨了转录因子SRY相关的高迁移率族蛋白7(SOX7)对HSC激活和LF的影响,以及潜在的分子机制。我们发现SOX7在人和小鼠纤维化肝脏中的表达水平降低,特别是在纤维化病灶。SOX7在原代活化的HSC和TGF-β1刺激的LX-2细胞中也下调。SOX7敲低可促进LX-2细胞的活化和增殖,同时抑制其凋亡。另一方面,SOX7的过表达抑制了HSC的活化和增殖。机械上,SOX7通过降低TGF-β1诱导的β-连环蛋白的表达和Smad2和Smad3的磷酸化来减弱HSC活化和LF。此外,使用AAV8-SOX7小鼠模型的SOX7的过表达改善了在体内响应于CCl4处理的LF的程度。总的来说,SOX7抑制HSC激活和LF。因此,以SOX7为目标,可能是一种潜在的新策略,以防止LF。
    Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-β1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of β-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-β1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.
    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
    转化生长因子-β(TGF-β)是一种多效性细胞因子,通过调节靶基因表达来调节多种细胞反应。它主要通过受体激活的转录因子Smad2和Smad3传递信号,它们在激活时与Smad4形成三聚体复合物,并通过与基因组DNA结合来调节基因表达。这里,我们通过筛选与内源性激活的Smad复合物结合的DNA序列的双链DNA寡核苷酸文库,研究了TGF-β以细胞环境依赖性方式调节靶基因转录的机制.使用抗Smad2/3抗体和分离自用TGF-β刺激的三种细胞系(A549、HepG2和HaCaT)的核提取物,通过循环扩增选定的靶标(CASTing)进行筛选。活化的Smad复合物对常规Smad结合基序如Smad结合元件(SBE)和CAGA基序的偏好在HepG2中与其他两种细胞系中不同,这可能表明活化的Smad复合物的不同组成。SBE或CAGA以外的几个转录因子结合基序,包括Fos/Jun绑定基序,在富集的序列中检测到。使用包含这些转录因子结合基序和Smad结合基序的序列进行的报告测定表明,某些基序可能参与TGF-β的细胞类型依赖性转录激活。结果表明,CASTing方法可用于阐明上下文依赖性Smad信号传导的分子基础。
    Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that modulates a wide variety of cellular responses by regulating target gene expression. It principally transmits signals via receptor-activated transcription factors Smad2 and Smad3, which form trimeric complexes with Smad4 upon activation and regulate gene expression by binding to genomic DNA. Here, we examined the mechanisms by which TGF-β regulates the transcription of target genes in a cell context-dependent manner by screening a double-stranded DNA oligonucleotide library for DNA sequences bound to endogenous activated Smad complexes. Screening was performed by cyclic amplification of selected targets (CASTing) using an anti-Smad2/3 antibody and nuclear extracts isolated from three cell lines (A549, HepG2, and HaCaT) stimulated with TGF-β. The preference of the activated Smad complexes for conventional Smad-binding motifs such as Smad-binding element (SBE) and CAGA motifs was different in HepG2 than in the other two cell lines, which may indicate the distinct composition of the activated Smad complexes. Several transcription factor-binding motifs other than SBE or CAGA, including the Fos/Jun-binding motifs, were detected in the enriched sequences. Reporter assays using sequences containing these transcription factor-binding motifs together with Smad-binding motifs indicated that some of the motifs may be involved in cell type-dependent transcriptional activation by TGF-β. The results suggest that the CASTing method is useful for elucidating the molecular basis of context-dependent Smad signaling.
    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
    上皮-间质转化(EMT)通过促进远处转移和抗癌药物耐药而导致癌症患者的不良预后。几种不同的代谢改变已被鉴定为关键的EMT表型。在本研究中,我们进一步研究了转化生长因子-β(TGF-β)诱导的EMT在非小细胞肺癌中的作用.我们的研究表明,TGF-β通过上调胞苷5三磷酸合成酶1(CTPS)在EMT功能中起作用,嘧啶代谢途径中CTP生物合成的重要酶。CTPS的敲除和酶抑制均可减少TGF-β诱导的EMT标记表达变化,体外化学抗性和迁移。此外,CTPS敲低抵消了TGF-β介导的UDP-葡糖醛酸的下调,戊二酸,肌酸,牛磺酸和烟酰胺.这些发现表明,CTPS在EMT代谢中起着多方面的作用,通过EMT对癌症的恶性转化至关重要,并强调其作为预防非小细胞肺癌耐药和转移的有希望的治疗靶点的潜力。
    Epithelial-to-mesenchymal transition (EMT) contributes to the poor prognosis of patients with cancer by promoting distant metastasis and anti-cancer drug resistance. Several distinct metabolic alterations have been identified as key EMT phenotypes. In the present study, we further characterize the role of transforming growth factor-β (TGF-β)-induced EMT in non-small-cell lung cancer. Our study revealed that TGF-β plays a role in EMT functions by upregulation of cytidine 5\'-triphosphate synthetase 1 (CTPS), a vital enzyme for CTP biosynthesis in the pyrimidine metabolic pathway. Both knockdown and enzymatic inhibition of CTPS reduced TGF-β-induced changes in EMT marker expression, chemoresistance and migration in vitro. Moreover, CTPS knockdown counteracted the TGF-β-mediated downregulation of UDP-glucuronate, glutarate, creatine, taurine and nicotinamide. These findings indicate that CTPS plays a multifaceted role in EMT metabolism, which is crucial for the malignant transformation of cancer through EMT, and underline its potential as a promising therapeutic target for preventing drug resistance and metastasis in non-small-cell lung cancer.
    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
    尽管有各种临床选择,人类前交叉韧带(ACL)损伤不能完全愈合。使用重组腺相关病毒(rAAV)载体的生物材料指导的基因治疗可能会改善ACL修复的内在机制。这里,我们研究了聚(苯乙烯磺酸钠)接枝的聚(ε-己内酯)(pNaSS接枝的PCL)膜是否可以在人骨髓间充质基质细胞(hMSCs)中递送编码修复性碱性成纤维细胞生长因子(FGF-2)和转化生长因子β(TGF-β)的rAAV载体,作为ACL病变中可植入细胞的来源.相对于所有其他条件,在细胞中实现有效和持续的rAAV介导的报道分子(红色荧光蛋白)和治疗性(FGF-2和TGF-β)基因过表达至少21天,特别是用pNaSS移植的PCL膜(差异高达5.2倍)。应用PCL膜对rAAV介导的FGF-2和TGF-β的表达增加了细胞增殖水平,DNA含量,随着时间的推移,在转录因子表达水平较高的细胞中,蛋白聚糖和I型和III型胶原蛋白的沉积(差异高达2.9倍)(Mohawk,巩膜轴)(差异高达1.9倍),与对照组相比,尤其是使用pNaSS移植的PCL膜时,不会激活炎性肿瘤坏死α。总的来说,TGF-β介导的作用高于FGF-2促进的作用,这可能是由于rAAV基因转移后基因表达水平较高。该研究显示了使用官能化PCL膜应用rAAV载体进行ACL修复的潜力。
    Despite various clinical options, human anterior cruciate ligament (ACL) lesions do not fully heal. Biomaterial-guided gene therapy using recombinant adeno-associated virus (rAAV) vectors may improve the intrinsic mechanisms of ACL repair. Here, we examined whether poly(sodium styrene sulfonate)-grafted poly(ε-caprolactone) (pNaSS-grafted PCL) films can deliver rAAV vectors coding for the reparative basic fibroblast growth factor (FGF-2) and transforming growth factor beta (TGF-β) in human mesenchymal stromal cells (hMSCs) as a source of implantable cells in ACL lesions. Efficient and sustained rAAV-mediated reporter (red fluorescent protein) and therapeutic (FGF-2 and TGF-β) gene overexpression was achieved in the cells for at least 21 days in particular with pNaSS-grafted PCL films relative to all other conditions (up to 5.2-fold difference). Expression of FGF-2 and TGF-β mediated by rAAV using PCL films increased the levels of cell proliferation, the DNA contents, and the deposition of proteoglycans and of type-I and -III collagen (up to 2.9-fold difference) over time in the cells with higher levels of transcription factor expression (Mohawk, Scleraxis) (up to 1.9-fold difference), without activation of inflammatory tumor necrosis alpha especially when using pNaSS-grafted PCL films compared with the controls. Overall, the effects mediated by TGF-β were higher than those promoted by FGF-2, possibly due to higher levels of gene expression achieved upon rAAV gene transfer. This study shows the potential of using functionalized PCL films to apply rAAV vectors for ACL repair.
    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
    我们以前的研究表明,SLC7A1在上皮性卵巢癌(EOC)肿瘤细胞中的上调显着增加癌细胞的增殖,迁移,和顺铂耐药;然而,SLC7A1在EOC中起作用的分子机制尚不清楚。在后来的研究中,我们发现SLC7A1在高级别浆液性卵巢癌(HGSOC)的间质部分也高表达,但是在间质中这种高表达的意义尚不清楚。这里,我们通过免疫组织化学显示SLC7A1在HGSOC中的间质高表达。富含癌相关成纤维细胞(CAF)的SLC7A1被TGF-β1上调。Transwell分析,划痕试验,cck8实验和细胞粘附实验显示SLC7A1在CAFs中高表达促进肿瘤细胞侵袭,体外迁移和转移。通过RNA测序和蛋白质印迹法验证了SLC7A1对卵巢癌(OC)中MAPK和EMT通路蛋白的影响。SLC7A1在OC中的过表达与MAPK/ERK通路和EMT有关。总的来说,在HGSOC,过表达SLC7A1的CAFs支持肿瘤细胞的迁移和侵袭;SLC7A1在卵巢癌中高表达,并参与MAPK信号通路中的ERK磷酸化和EMT信号传导。这表明SLC7A1可能是OC转移的潜在治疗靶标。
    Our previous studies have shown that upregulation of SLC7A1 in epithelial ovarian cancer (EOC) tumor cells significantly increases cancer cell proliferation, migration, and cisplatin resistance; however, the molecular mechanism by which SLC7A1 functions in EOC remains unknown. In later studies, we found that SLC7A1 is also highly expressed in the interstitial portion of high-grade serous ovarian cancer (HGSOC), but the significance of this high expression in the interstitial remains unclear. Here, we showed the Interstitial high expression of SLC7A1 in HGSOC by immunohistochemistry. SLC7A1 enriched in cancer-associated fibroblasts (CAFs) was upregulated by TGF-β1. Transwell assay, scratch assay, cck8 assay and cell adhesion assay showed that SLC7A1 highly expressed in CAFs promoted tumor cells invasion, migration and metastasis in vitro. The effect of SLC7A1 on MAPK and EMT pathway proteins in ovarian cancer (OC) was verified by RNA sequencing and western blotting. Overexpression of SLC7A1 in OC is involved in MAPK/ ERK pathway and EMT. In general, in HGSOC, CAFs overexpressing SLC7A1 supported the migration and invasion of tumor cells; SLC7A1 is highly expressed in ovarian cancer and is involved in ERK phosphorylation and EMT signaling in MAPK signaling pathway. This suggests that SLC7A1 may be a potential therapeutic target for OC metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号