Large Neutral Amino Acid-Transporter 1

大型中性氨基酸转运蛋白 1
  • 文章类型: Journal Article
    大多数头颈部鳞状细胞癌(HNSCC)患者被诊断为晚期局部阶段。虽然免疫疗法提高了生存率,只有少数患者对靶向免疫疗法有持久的反应,带来了巨大的临床挑战。我们通过分析从公共数据库检索的单细胞RNA测序(scRNA-seq)数据和批量RNA测序数据集,调查了免疫治疗前后HNSCC队列中肿瘤微环境的异质性。
    我们构建了免疫治疗前后HNSCC患者的单细胞转录组景观,并分析了细胞组成,发展轨迹,基因调控网络,和不同细胞类型亚群的通讯模式。此外,我们通过蛋白质印迹评估了HNSCC细胞中相关指标的表达水平,ELISA,和荧光探针技术。
    在单细胞级别,我们确定了TP63+SLC7A5+HNSCC的亚群表现出铁凋亡抗性表型。该亚群通过高TP63表达介导的SLC7A5的转录上调抑制恶性细胞中的铁凋亡,从而促进肿瘤生长和对免疫疗法的抵抗。实验结果表明,TP63的过表达上调了SLC7A5的表达,并抑制了HNSCC细胞中Fe2和ROS的浓度。通过整合大量转录组数据,我们开发了一个基于TP63和SLC7A5的临床评分模型,这两个模型与肿瘤分期密切相关,揭示TP63+SLC7A5+HNSCC介导的铁凋亡机制在HNSCC患者中的显着预后功效。
    我们的研究阐明了免疫疗法前后HNSCC中的TME,揭示了TP63+SLC7A5+HNSCC抑制铁凋亡并通过TP63诱导的SLC7A5上调增强肿瘤抗性的新机制。这些见解为开发更有效的HNSCC治疗奠定了基础。
    UNASSIGNED: Most head and neck squamous cell carcinoma (HNSCC) patients are diagnosed at an advanced local stage. While immunotherapy has improved survival rates, only a minority of patients respond durably to targeted immunotherapies, posing substantial clinical challenges. We investigated the heterogeneity of the tumor microenvironment in HNSCC cohorts before and after immunotherapy by analyzing single-cell RNA sequencing (scRNA-seq) data and bulk RNA sequencing datasets retrieved from public databases.
    UNASSIGNED: We constructed a single-cell transcriptome landscape of HNSCC patients before and after immunotherapy and analyzed the cellular composition, developmental trajectories, gene regulatory networks, and communication patterns of different cell type subpopulations. Additionally, we assessed the expression levels of relevant indicators in HNSCC cells via western blot, ELISA, and fluorescent probe techniques.
    UNASSIGNED: At the single-cell level, we identified a subpopulation of TP63+ SLC7A5+ HNSCC that exhibited a ferroptosis-resistant phenotype. This subpopulation suppresses ferroptosis in malignant cells through the transcriptional upregulation of SLC7A5 mediated by high TP63 expression, thereby promoting tumor growth and resistance to immunotherapy. The experimental results demonstrated that the overexpression of TP63 upregulated the expression of SLC7A5 and suppressed the concentrations of Fe2+ and ROS in HNSCC cells. By integrating bulk transcriptome data, we developed a clinical scoring model based on TP63 and SLC7A5, which are closely associated with tumor stage, revealing the significant prognostic efficacy of the TP63+ SLC7A5+ HNSCC-mediated ferroptosis mechanism in HNSCC patients.
    UNASSIGNED: Our research elucidates the TME in HNSCC before and after immunotherapy, revealing a novel mechanism by which TP63+ SLC7A5+ HNSCC inhibits ferroptosis and enhances tumor resistance via TP63-induced SLC7A5 upregulation. These insights lay the foundation for the development of more effective treatments for HNSCC.
    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
    高分子量聚乙烯亚胺(HMWPEI;支链25kDaPEI)由于其高转染效率而被广泛研究用于基因递送。然而,毒性和缺乏对特定细胞的靶向限制了其临床应用。在目前的调查中,将L-3,4-二羟基苯丙氨酸(L-DOPA)缀合在HMWPEI上,以靶向L型氨基酸转运蛋白1(LAT-1)并调节聚合物/质粒复合物(聚合复合物)表面上的正电荷密度。生物物理表征的结果表明,PEI缀合物能够形成Zeta电位在+9.5-12.4mV范围内的≤180nm的纳米颗粒。这些复合物可以浓缩质粒DNA,并在载体与质粒的比率高于4时保护其免受核酸酶消化。将L-DOPA缀合的PEI衍生物与编码人白细胞介素-12(hIL-12)的质粒复合。靶向复合物在4T1鼠乳腺癌细胞系中显示出高达2.5倍的转染效率,其表达LAT-1,而不是以相同方式制备的25kDaPEI聚合复合物。这些聚合复合物的细胞毒性也显著低于未修饰的母体HMWPEI。这些结果支持PEI的L-3,4-二羟基苯丙氨酸衍生物在开发LAT-1靶向基因载体的任何尝试中的用途。
    High molecular weight polyethylenimine (HMW PEI; branched 25 kDa PEI) has been widely investigated for gene delivery due to its high transfection efficiency. However, the toxicity and lack of targeting to specific cells have limited its clinical application. In the present investigation, L-3, 4-dihydroxyphenylalanine (L-DOPA) was conjugated on HMW PEI in order to target L-type amino acid transporter 1 (LAT-1) and modulate positive charge density on the surface of polymer/plasmid complexes (polyplexes). The results of biophysical characterization revealed that the PEI conjugates are able to form nanoparticles ≤ 180 nm with the zeta potential ranging from + 9.5-12.4 mV. These polyplexes could condense plasmid DNA and protect it against nuclease digestion at the carrier to plasmid ratios higher than 4. L-DOPA conjugated PEI derivatives were complexed with a plasmid encoding human interleukin-12 (hIL-12). Targeted polyplexes showed up to 2.5 fold higher transfection efficiency in 4T1 murine mammary cancer cell line, which expresses LAT-1, than 25 kDa PEI polyplexes prepared in the same manner. The cytotoxicity of these polyplexes was also substantially lower than the unmodified parent HMW PEI. These results support the use of L-3, 4-dihydroxyphenylalanine derivatives of PEI in any attempt to develop a LAT-1 targeted gene carrier.
    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
    激活的Wnt/β-catenin通路是肝癌发生的关键遗传事件。溶质载体(SLC)转运体是有前景的药物靶标。这里,我们确定SLC13A3是肝癌β-catenin下游的药物靶向效应物.SLC13A3表达在具有功能获得(GOF)突变体CTNNB1(编码β-连环蛋白的基因)的人肝癌样品中升高。β-连环蛋白的激活上调SLC13A3,导致内源性SLC13A3底物的细胞内积累。SLC13A3被鉴定为谷胱甘肽(GSH)的低亲和力转运蛋白。SLC13A3的沉默通过c-MYC信号传导下调亮氨酸转运蛋白SLC7A5,导致亮氨酸耗竭和mTOR失活。此外,SLC13A3的沉默会消耗GSH并诱导β-catenin激活的肝癌细胞中的自噬铁凋亡。重要的是,SLC13A3的基因抑制和小分子SLC13A3抑制剂均抑制小鼠β-catenin驱动的肝癌发生。总之,我们的研究表明,SLC13A3可能是治疗具有GOFCTNNB1突变的人类肝癌的有前景的治疗靶点.
    Activated Wnt/β-catenin pathway is a key genetic event in liver cancer development. Solute carrier (SLC) transporters are promising drug targets. Here, we identify SLC13A3 as a drug-targetable effector downstream of β-catenin in liver cancer. SLC13A3 expression is elevated in human liver cancer samples with gain of function (GOF) mutant CTNNB1, the gene encoding β-catenin. Activation of β-catenin up-regulates SLC13A3, leading to intracellular accumulation of endogenous SLC13A3 substrates. SLC13A3 is identified as a low-affinity transporter for glutathione (GSH). Silencing of SLC13A3 downregulates the leucine transporter SLC7A5 via c-MYC signaling, leading to leucine depletion and mTOR inactivation. Furthermore, silencing of SLC13A3 depletes GSH and induces autophagic ferroptosis in β-catenin-activated liver cancer cells. Importantly, both genetic inhibition of SLC13A3 and a small molecule SLC13A3 inhibitor suppress β-catenin-driven hepatocarcinogenesis in mice. Altogether, our study suggests that SLC13A3 could be a promising therapeutic target for treating human liver cancers with GOF CTNNB1 mutations.
    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
    L型氨基酸转运蛋白1(LAT1)在各种癌症类型中上调,并有助于疾病进展。以前的研究已经证明或暗示缺氧诱导因子(HIF),缺氧反应中的关键转录因子,控制LAT1基因在几种类型的癌细胞中的表达。然而,这种调节关系尚未在结直肠癌(CRC)中进行研究,LAT1表达增加的癌症类型之一具有预后意义。在这项研究中,我们发现,在缺氧条件下(1%O2)在体外对CRCHT-29细胞中LAT1mRNA和蛋白均无诱导作用,无论显著的HIF-1/2α积累和HIF依赖性的葡萄糖转运蛋白1上调。在8个测试的CRC细胞系中,低氧处理通常不会增加LAT1的mRNA或蛋白表达。与氨基酸限制引起的明显上调相反。有趣的是,敲低vonHippel-Lindau泛素连接酶以抑制HIF的蛋白酶体降解,导致HIF-2α的积累,并增加某些CRC细胞系中的LAT1表达。本研究有助于描述LAT1在CRC细胞中病理表达的分子机制。强调HIFs依赖性LAT1在癌细胞中转录上调的模糊性。
    L-type amino acid transporter 1 (LAT1) is upregulated in various cancer types and contributes to disease progression. Previous studies have demonstrated or suggested that hypoxia-inducible factors (HIFs), the key transcription factors in hypoxic responses, control the expression of LAT1 gene in several types of cancer cells. However, this regulatory relationship has not been investigated yet in colorectal cancer (CRC), one of the cancer types in which the increased LAT1 expression holds prognostic significance. In this study, we found that neither LAT1 mRNA nor protein is induced under hypoxic condition (1% O2) in CRC HT-29 cells in vitro, regardless of the prominent HIF-1/2α accumulation and HIFs-dependent upregulation of glucose transporter 1. The hypoxic treatment generally did not increase either the mRNA or protein expression of LAT1 in eight CRC cell lines tested, in contrast to the pronounced upregulation by amino acid restriction. Interestingly, knockdown of von Hippel-Lindau ubiquitin ligase to inhibit the proteasomal degradation of HIFs caused an accumulation of HIF-2α and increased the LAT1 expression in certain CRC cell lines. This study contributes to delineating the molecular mechanisms responsible for the pathological expression of LAT1 in CRC cells, emphasizing the ambiguity of HIFs-dependent transcriptional upregulation of LAT1 across cancer cells.
    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
    Claudin-1(CLDN1)在人肺腺癌衍生的A549细胞中高表达,并参与化学耐药性的增强。然而,化学抗性的机制尚未完全了解。在肿瘤微环境中,癌细胞暴露于压力条件,如缺氧和营养不良。这里,我们使用A549细胞研究了CLDN1表达对氨基酸(AA)通量和化学抗性的影响。L型AA转运蛋白的表达,LAT1和LAT3在A549球状体中通过CLDN1沉默而降低。细胞外AA浓度的降低增加了这些AA转运蛋白在二维(2D)培养细胞中的表达。除Ser外的细胞旁AA通量,Thr,Tyr,阿拉,通过CLDN1沉默增强了Gly。这些结果表明,CLDN1形成了一些AAs的细胞旁屏障,导致球状体中LAT1/3表达升高。球状体中的CLDN1沉默减少了线粒体和细胞质中活性氧的产生,导致核因子红系2相关因子2(Nrf2)及其靶抗氧化基因的表达下调。CLDN1沉默增强了抗癌药物包括阿霉素和顺铂的细胞毒性,被萝卜硫素堵塞了,Nrf2信号的诱导剂。同样,Nrf2沉默增强了抗癌诱导的毒性。在2D培养的细胞中,AA缺乏和萝卜硫烷可减弱抗癌诱导的毒性。我们建议CLDN1在球体中形成AA屏障,导致A549细胞中Nrf2依赖性化学抗性的增强。
    Claudin-1 (CLDN1) is highly expressed in human lung adenocarcinoma-derived A549 cells and is involved in the augmentation of chemoresistance. However, the mechanism of chemoresistance is not fully understood. In the tumor microenvironment, cancer cells are exposed to stress conditions such as hypoxia and malnutrition. Here, we investigated the effect of CLDN1 expression on amino acid (AA) flux and chemoresistance using A549 cells. The expression of L-type AA transporters, LAT1 and LAT3, was decreased by CLDN1 silencing in A549 spheroids. A reduction in extracellular AA concentration increased the expression of these AA transporters in two-dimensional (2D) cultured cells. The paracellular AA flux except for Ser, Thr, Tyr, Ala, and Gly was enhanced by CLDN1 silencing. These results suggest that CLDN1 forms a paracellular barrier to some AAs, leading to the elevation of LAT1/3 expression in spheroids. The production of reactive oxygen species in the mitochondria and cytosol was decreased by CLDN1 silencing in spheroids, resulting in downregulation of the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its target antioxidant genes. CLDN1 silencing enhanced the cytotoxicity of anticancer drugs including doxorubicin and cisplatin, which was blocked by sulforaphane, an inducer of Nrf2 signaling. Similarly, the anticancer-induced toxicity was enhanced by Nrf2 silencing. In 2D cultured cells, the anticancer-induced toxicity was attenuated by AA deficiency and sulforaphane. We suggest that CLDN1 forms an AA barrier in spheroids, leading to the augmentation of Nrf2-dependent chemoresistance in A549 cells.
    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
    系统性硬化症(SSc)是一种慢性自身免疫性结缔组织疾病。血管损害是SSc的重要特点之一,影响疾病的进展和预后。MiR-126-3p是调节血管结构和功能的重要microRNA(miRNA),可以通过外泌体运输。然而,miR-126-3p在SSc血管损伤中的作用尚不清楚.因此,我们专注于miR-126-3p与SSc血管损伤之间的联系,以及研究miR-126-3p在SSc血管损伤中的潜在作用。首先,这项研究成功地从临床血浆样本中提取了细胞外囊泡,并对其中的外泌体进行了表征。然后,我们通过在线数据库预测并筛选了哺乳动物/机制雷帕霉素靶蛋白(mTOR)的靶途径和miR-126-3p的靶基因SLC7A5。接下来,我们构建了SSc小鼠用于体内研究。结果显示血浆外泌体中miR-126-3p的表达降低,而SLC7A5表达式,自噬,和脂质过氧化在主动脉中增加。荧光素酶报告基因测定表明miR-126-3p可以与SLC7A5结合,导致其表达降低。体外实验表明,外泌体miR-126-3p可以被人脐静脉内皮细胞(HUVEC)内化。miR-126-3p组表现出增强的细胞活力和试管形成能力,随着血管形成标志物CD31的表达增加。此外,miR-126-3p下调HUVECs中SLC7A5和LC3的蛋白表达,在上调mTOR蛋白表达的同时,P62,PPARγ,和CPT-1。然而,miR-126-3p对HUVECs的作用被mTOR抑制剂抵消,被mTOR激活剂增强.结果表明,外泌体miR-126-3p有可能通过调节HUVECs中的SLC7A5/mTOR信号通路来保护SSc免受血管损伤。
    Systemic sclerosis (SSc) is a chronic autoimmune connective tissue disease. Vascular damage is one of the important features of SSc, which affects the progression and prognosis of the disease. MiR-126-3p is an important microRNA (miRNA) that regulates vascular structure and function, which can be transported through exosomes. However, the role of miR-126-3p in vascular damage in SSc is still unclear. Therefore, we focused on the connection between miR-126-3p and vascular damage in SSc, as well as investigated the potential role of miR-126-3p in vascular damage in SSc. First, this study successfully extracted extracellular vesicles from clinical plasma samples and characterized the exosomes within them. Then, we predicted and screened the target pathway mammalian/mechanistic target of rapamycin (mTOR) and the target gene SLC7A5 of miR-126-3p through online databases. Next, we constructed SSc mice for in vivo studies. The results showed that the expression of miR-126-3p was decreased in the plasma exosomes, while the SLC7A5 expression, autophagy, and lipid peroxidation were increased in the aorta. Luciferase reporter gene assays demonstrated that miR-126-3p can bind to SLC7A5, resulting in a decrease in its expression. In vitro experiments have shown that exosomal miR-126-3p can be internalized by human umbilical vein endothelial cells (HUVECs). The miR-126-3p group exhibited enhanced cell viability and tube formation ability, along with increased expression of the vascular formation marker CD31. Additionally, miR-126-3p downregulated the protein expression of SLC7A5 and LC3 in HUVECs, while upregulating the protein expression of mTOR, P62, PPARγ, and CPT-1. However, the effects of miR-126-3p on HUVECs were counteracted by mTOR inhibitors and enhanced by mTOR activators. The results indicated that exosomal miR-126-3p has the potential to protect against vascular injury in SSc by regulating the SLC7A5/mTOR signalling pathway in HUVECs.
    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
    侵袭性自然杀伤细胞白血病(ANKL)是一种罕见的血液恶性肿瘤,具有暴发性临床病程。我们先前的研究表明,ANKL细胞主要在肝窦中增殖,并且强烈依赖于转铁蛋白的补充。此外,我们证明了肝脏驻留的ANKL细胞对PPMX-T003敏感,PPMX-T003是一种抗人转铁蛋白受体1抑制性抗体,而脾脏驻留的ANKL细胞对转铁蛋白受体1抑制具有抗性。然而,调节ANKL细胞铁依赖性的微环境因素尚不清楚.在这项研究中,我们首先揭示了PPMX-T003的抗肿瘤作用以DNA复制依赖性方式的DNA双链断裂为特征,类似于传统的细胞毒性剂。我们还发现,通过LAT1的细胞外氨基酸流入刺激了对PPMX-T003的敏感性。一起来看,我们发现通过LAT1的细胞外氨基酸流入量是通过调节其mTOR/Myc活性决定ANKL细胞铁依赖性的关键环境因素,这为肝脏和脾脏驻留的ANKL细胞对PPMX-T003的不同敏感性提供了很好的解释,因为肝窦含有从肠道吸收的丰富氨基酸。
    Aggressive natural killer cell leukemia (ANKL) is a rare hematological malignancy with a fulminant clinical course. Our previous study revealed that ANKL cells proliferate predominantly in the liver sinusoids and strongly depend on transferrin supplementation. In addition, we demonstrated that liver-resident ANKL cells are sensitive to PPMX-T003, an anti-human transferrin receptor 1 inhibitory antibody, whereas spleen-resident ANKL cells are resistant to transferrin receptor 1 inhibition. However, the microenvironmental factors that regulate the iron dependency of ANKL cells remain unclear. In this study, we first revealed that the anti-neoplastic effect of PPMX-T003 was characterized by DNA double-strand breaks in a DNA replication-dependent manner, similar to conventional cytotoxic agents. We also found that the influx of extracellular amino acids via LAT1 stimulated sensitivity to PPMX-T003. Taken together, we discovered that the amount of extracellular amino acid influx through LAT1 was the key environmental factor determining the iron dependency of ANKL cells via adjustment of their mTOR/Myc activity, which provides a good explanation for the different sensitivity to PPMX-T003 between liver- and spleen-resident ANKL cells, as the liver sinusoid contains abundant amino acids absorbed from the gut.
    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
    上呼吸道鳞状细胞癌(UASCC)是一种常见的侵袭性恶性肿瘤,几乎没有有效的治疗选择。这里,我们研究了这种癌症的氨基酸代谢,令人惊讶的是,UASCC在所有人类癌症中表现出最高的蛋氨酸水平,由其运输车LAT1驱动。我们表明LAT1在UASCC中也以最高水平表达,由UASCC特异性启动子和增强子转录激活,它们由SCC主调节剂TP63/KLF5/SREBF1直接共同调节。出乎意料的是,无偏生物信息学筛选将EZH2鉴定为LAT1-蛋氨酸途径下游的最重要靶标,直接将蛋氨酸代谢与表观基因组重编程联系起来。重要的是,这种级联反应对于UASCC患者来源的肿瘤类器官的存活和增殖是必不可少的。此外,LAT1表达与细胞对LAT1-甲硫氨酸-EZH2轴抑制的敏感性密切相关。值得注意的是,这种独特的LAT1-蛋氨酸-EZH2级联反应可以通过体内药理学方法或饮食干预有效靶向。总之,这项工作映射了表观基因组重编程与蛋氨酸代谢之间的独特机制串扰,确立了其在UASCC生物学中的生物学意义,并确定了一种独特的肿瘤特异性脆弱性,可以在药理和饮食上利用。
    Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.
    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
    乳腺癌是世界范围内最常见的恶性肿瘤之一。SLC7A2在多种癌症中异常表达。然而,其在三阴性乳腺癌(TNBC)中的潜力仍不清楚。目的探讨SLC7A2在TNBC中的作用及其分子机制。通过RT-qPCR检测mRNA的表达。蛋白质印迹法检测蛋白质表达。使用FISH测定法测定ACOX1和TCF1的共定位。使用体外组蛋白巴豆化测定进行组蛋白巴豆化。使用CCK-8和流式细胞术测定进行功能分析。进行异种移植物测定以进一步验证SLC7A2在TNBC中的作用。使用免疫组织化学检测CD8A表达。我们发现SLC7A2在TNBC肿瘤中下调。低水平与晚期分期和淋巴结转移有关。SLC7A2表达与CD8A呈正相关。SLC7A2介导的赖氨酸分解代谢驱动CD8+T细胞的活化。此外,SLC7A2通过上调ACOX1促进组蛋白巴豆化。它还促进ACOX1和TCF1之间的相互作用,从而促进抗肿瘤T细胞免疫。此外,SLC7A2的过表达激活CD8+T细胞并增强体内抗PD-1疗法的化学敏感性。总之,SLC7A2可能通过激活抗肿瘤免疫而在TNBC中发挥抗肿瘤基因的作用,提示SLC7A2/ACOX1/TCF1信号传导是一种有前途的治疗策略。
    Breast cancer is one of the most common malignant tumors worldwide. SLC7A2 is abnormally expressed in multiple cancers. However, its potential in triple negative breast cancer (TNBC) is still unclear. The purpose of this study was to investigate the roles of SLC7A2 and its underlying molecular mechanisms in TNBC. mRNA expression was detected by RT-qPCR. Protein expression was detected by western blot. Co-localization of ACOX1 and TCF1 was determined using FISH assay. Histone crotonylation was performed using in vitro histone crotonylation assay. Functional analysis was performed using CCK-8 and flow cytometry assays. Xenograft assay was conducted to further verify the role of SLC7A2 in TNBC. CD8A expression was detected using immunohistochemistry. We found that SLC7A2 is downregulated in TNBC tumors. Low levels are associated with advanced stages and lymph node metastasis. SLC7A2 expression is positively correlated with CD8A. SLC7A2-mediated lysine catabolism drives the activation of CD8+ T cells. Moreover, SLC7A2 promotes histone crotonylation via upregulating ACOX1. It also promotes interaction between ACOX1 and TCF1, thus promoting antitumor T cell immunity. Additionally, overexpression of SLC7A2 activates CD8+ T cells and enhances the chemosensitivity of anti-PD-1 therapies in vivo. In conclusion, SLC7A2 may function as an antitumor gene in TNBC by activating antitumor immunity, suggesting SLC7A2/ACOX1/TCF1 signaling as a promising therapeutic strategy.
    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
    血管生成,从预先存在的脉管系统中生长新血管,对新器官系统的发展至关重要,但是血管生成的转录控制仍未完全了解。在这里,我们表明FOXC1对于视网膜血管生成是必不可少的。Foxc1的内皮细胞(EC)特异性丢失会损害视网膜血管的生长以及Slc3a2和Slc7a5的表达,Slc3a2和Slc7a5编码异二聚体CD98(LAT1/4F2hc)氨基酸转运蛋白,并调节必需氨基酸的细胞内转运和哺乳动物靶蛋白的激活雷帕霉素(mTOR)。EC-Foxc1缺乏会降低mTOR活性,而mTOR激动剂MHY-1485的给药挽救了受干扰的视网膜血管生成。在氧诱导的视网膜病变模型中,EC-Foxc1表达是视网膜血运重建和新生血管簇消退所必需的。Foxc1对于周细胞也是不可或缺的,视网膜血管生成过程中血-视网膜屏障的关键组成部分。我们的发现确立了FOXC1作为视网膜血管的关键调节因子,并确定了治疗视网膜血管疾病的治疗靶点。
    Angiogenesis, the growth of new blood vessels from pre-existing vasculature, is essential for the development of new organ systems, but transcriptional control of angiogenesis remains incompletely understood. Here we show that FOXC1 is essential for retinal angiogenesis. Endothelial cell (EC)-specific loss of Foxc1 impairs retinal vascular growth and expression of Slc3a2 and Slc7a5, which encode the heterodimeric CD98 (LAT1/4F2hc) amino acid transporter and regulate the intracellular transport of essential amino acids and activation of the mammalian target of rapamycin (mTOR). EC-Foxc1 deficiency diminishes mTOR activity, while administration of the mTOR agonist MHY-1485 rescues perturbed retinal angiogenesis. EC-Foxc1 expression is required for retinal revascularization and resolution of neovascular tufts in a model of oxygen-induced retinopathy. Foxc1 is also indispensable for pericytes, a critical component of the blood-retina barrier during retinal angiogenesis. Our findings establish FOXC1 as a crucial regulator of retinal vessels and identify therapeutic targets for treating retinal vascular disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号