Amino Acid Transport System ASC

氨基酸转运系统 ASC
  • 文章类型: Journal Article
    ASCT2是中性氨基酸的专性交换剂,有助于细胞氨基酸稳态。ASCT2与将谷氨酸浓缩在细胞溶质中的兴奋性氨基酸转运蛋白(EAAT)属于同一家族(SLC1)。使ASCT2成为交换器而不是浓缩器的机制仍然是神秘的。这里,我们使用低温电子显微镜和分子动力学模拟来阐明ASCT2交换机制的结构基础。我们确定ASCT2在每个运输的底物上结合三个Na离子,并访问一个可能作为防止Na离子泄漏的检查点的状态。这两个功能都与EAAT共享。然而,与EAAT相比,ASCT2即使在Na+耗尽的条件下也保留一个Na+离子。我们证明ASCT2不能经历TM7的结构转变,这对于EAAT的集中运输周期至关重要。这种结构刚性和高亲和力Na结合位点有效地将ASCT2限制为交换模式。
    ASCT2 is an obligate exchanger of neutral amino acids, contributing to cellular amino acid homeostasis. ASCT2 belongs to the same family (SLC1) as Excitatory Amino Acid Transporters (EAATs) that concentrate glutamate in the cytosol. The mechanism that makes ASCT2 an exchanger rather than a concentrator remains enigmatic. Here, we employ cryo-electron microscopy and molecular dynamics simulations to elucidate the structural basis of the exchange mechanism of ASCT2. We establish that ASCT2 binds three Na+ ions per transported substrate and visits a state that likely acts as checkpoint in preventing Na+ ion leakage, both features shared with EAATs. However, in contrast to EAATs, ASCT2 retains one Na+ ion even under Na+-depleted conditions. We demonstrate that ASCT2 cannot undergo the structural transition in TM7 that is essential for the concentrative transport cycle of EAATs. This structural rigidity and the high-affinity Na+ binding site effectively confine ASCT2 to an exchange mode.
    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
    肝细胞癌(HCC)是最常见的原发性肝脏恶性肿瘤,是世界上癌症相关死亡的主要原因。这项研究旨在表征与正常肝细胞相比,HCC中谷氨酰胺氨基酸转运蛋白的表达谱。使用qPCR研究了肝癌的体外和体内模型,而通过RNAseq分析患者肿瘤中谷氨酰胺转运体表达水平的预后意义。通过siRNA或γ-对硝基苯胺(GPNA)靶向溶质载体(SLC)1A5和SLC38A2。HCC细胞依赖于外源谷氨酰胺以实现最佳存活和生长。小鼠肝癌细胞的谷氨酰胺摄取率优于正常肝细胞(p<0.0001)。与正常肝脏相比,HCC表现出谷氨酰胺转运蛋白的全局重编程:SLC38A3水平降低,而SLC38A1、SLC7A6和SLC1A5水平升高。此外,SLC6A14和SLC38A3水平降低或SLC38A1,SLC7A6和SLC1A5水平升高预测生存结局恶化(均P<0.05).敲低人Huh7和Hep3B肝癌细胞中SLC1A5和/或SLC38A2的表达,以及GPNA介导的抑制,显著降低谷氨酰胺的摄取;联合SLC1A5和SLC38A2靶向作用最显著(均p<0.05)。这项研究表明,谷氨酰胺转运体重编程是HCC的新标志,并且这种表达谱具有临床意义。
    Hepatocellular carcinoma (HCC) is the most prevalent primary liver malignancy and is a major cause of cancer-related mortality in the world. This study aimed to characterize glutamine amino acid transporter expression profiles in HCC compared to those of normal liver cells. In vitro and in vivo models of HCC were studied using qPCR, whereas the prognostic significance of glutamine transporter expression levels within patient tumors was analyzed through RNAseq. Solute carrier (SLC) 1A5 and SLC38A2 were targeted through siRNA or gamma-p-nitroanilide (GPNA). HCC cells depended on exogenous glutamine for optimal survival and growth. Murine HCC cells showed superior glutamine uptake rate than normal hepatocytes (p < 0.0001). HCC manifested a global reprogramming of glutamine transporters compared to normal liver: SLC38A3 levels decreased, whereas SLC38A1, SLC7A6, and SLC1A5 levels increased. Also, decreased SLC6A14 and SLC38A3 levels or increased SLC38A1, SLC7A6, and SLC1A5 levels predicted worse survival outcomes (all p < 0.05). Knockdown of SLC1A5 and/or SLC38A2 expression in human Huh7 and Hep3B HCC cells, as well as GPNA-mediated inhibition, significantly decreased the uptake of glutamine; combined SLC1A5 and SLC38A2 targeting had the most considerable impact (all p < 0.05). This study revealed glutamine transporter reprogramming as a novel hallmark of HCC and that such expression profiles are clinically significant.
    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
    镉(Cd)暴露显着增加患肺癌的风险。癌症对谷氨酰胺的需求正在增加,包括肺癌.在这项研究中,我们研究了谷氨酰胺代谢在Cd诱导的细胞生长和迁移中的作用。首先,我们发现2μMCd处理上调了ASCT2的表达(丙氨酸,丝氨酸,半胱氨酸优先转运蛋白2)和ASNS(天冬酰胺合成酶),同时下调A549细胞中的线粒体谷氨酰胺酶GLS1。在用0.5和1mgCd/kg体重处理的雄性BALB/c小鼠中获得了相同的结果。随后,谷氨酰胺剥夺和siASCT2转染均显示谷氨酰胺在Cd诱导的细胞生长和迁移中起作用。此外,使用4-PBA(5mM),内质网(ER)应激的抑制剂,Tm(0.1μg/ml),ER压力的诱导剂,siHMGA2和过表达HMGA2质粒,我们证明ER应激/HMGA2轴参与诱导ASCT2和ASNS,同时抑制GLS1。此外,使用HMGA2抗体的染色质免疫沉淀分析揭示了HMGA2与ASCT2、ASNS、和GLS1基因。最后,双荧光素酶报告基因测定确定HMGA2增加ASCT2和ASNS的转录,同时抑制GLS1的转录。总的来说,我们发现,ER应激诱导的HMGA2通过ASCT2,ASNS和GLS1的转录调节来控制谷氨酰胺代谢,从而在低浓度Cd暴露期间加速细胞生长和迁移。这项研究创新性地揭示了Cd诱导细胞生长的机制,为通过谷氨酰胺代谢预防Cd毒性提供了新的视角。
    Cadmium (Cd) exposure significantly increases the risk of lung cancer. The demand for glutamine is increasing in cancers, including lung cancer. In this study, we investigated the role of glutamine metabolism in Cd-induced cell growth and migration. Firstly, we found that 2 μM Cd-treatment up-regulated the expression of ASCT2 (alanine, serine, cysteine-preferring transporter 2) and ASNS (asparagine synthetase) while downregulating mitochondrial glutaminase GLS1 in A549 cells. The same results were obtained in male BALB/c mice treated with 0.5 and 1 mg Cd/kg body weight. Subsequently, both glutamine deprivation and transfection with siASCT2 revealed that glutamine played a role in Cd-induced cell growth and migration. Furthermore, using 4-PBA (5 mM), an inhibitor of endoplasmic reticulum (ER) stress, Tm (0.1 μg/ml), an inducer of ER stress, siHMGA2, and over-expressing HMGA2 plasmids we demonstrated that ER stress/HMGA2 axis was involved in inducing ASCT2 and ASNS, while inhibiting GLS1. Additionally, the chromatin immunoprecipitation assay using an HMGA2 antibody revealed the direct binding of the HMGA2 to the promoter sequences of the ASCT2, ASNS, and GLS1 genes. Finally, dual luciferase reporter assay determined that HMGA2 increased the transcription of ASCT2 and ASNS while inhibiting the transcription of GLS1. Overall, we found that ER stress-induced HMGA2 controls glutamine metabolism by transcriptional regulation of ASCT2, ASNS and GLS1 to accelerate cell growth and migration during exposure to Cd at low concentrations. This study innovatively revealed the mechanism of Cd-induced cell growth which offers a fresh perspective on preventing Cd toxicity through glutamine metabolism.
    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
    泛素化在蛋白质稳定性中起着至关重要的作用,亚细胞定位,和互动。不同类型的泛素化之间的串扰导致蛋白质的不同生物学结果。然而,泛素化相关串扰在淋巴结(LN)转移中的作用以及控制这一过程的关键调节因素尚未确定.采用高通量测序,我们发现,泛素结合酶E2C(UBE2C)在膀胱癌(BCa)中过度表达,并与不良预后密切相关.UBE2C的过表达在体外和体内都增加了BCa淋巴管生成并促进了LN转移。机械上,UBE2C介导的钠偶联中性氨基酸转运体2(SNAT2)在赖氨酸59处的单泛素化,以抑制在SNAT2的赖氨酸33处的K63连接的多泛素化。单泛素化和K63连接的聚泛素化之间的串扰通过抑制epsin1介导的(EPN1介导的)内吞作用增加了SNAT2膜蛋白水平。SNAT2促进谷氨酰胺的摄取和代谢,促进VEGFC的分泌,最终导致BCa患者的淋巴管生成和LN转移。重要的是,在患者来源的异种移植模型中,抑制UBE2C显著减弱BCa淋巴管生成。我们的结果揭示了UBE2C介导SNAT2的单泛素化和K63连接的多泛素化之间的串扰以促进BCa转移并将UBE2C鉴定为治疗LN转移性BCa的有希望的靶标的机制。
    Ubiquitination plays an essential role in protein stability, subcellular localization, and interactions. Crosstalk between different types of ubiquitination results in distinct biological outcomes for proteins. However, the role of ubiquitination-related crosstalk in lymph node (LN) metastasis and the key regulatory factors controlling this process have not been determined. Using high-throughput sequencing, we found that ubiquitin-conjugating enzyme E2 C (UBE2C) was overexpressed in bladder cancer (BCa) and was strongly associated with an unfavorable prognosis. Overexpression of UBE2C increased BCa lymphangiogenesis and promoted LN metastasis both in vitro and in vivo. Mechanistically, UBE2C mediated sodium-coupled neutral amino acid transporter 2 (SNAT2) monoubiquitination at lysine 59 to inhibit K63-linked polyubiquitination at lysine 33 of SNAT2. Crosstalk between monoubiquitination and K63-linked polyubiquitination increased SNAT2 membrane protein levels by suppressing epsin 1-mediated (EPN1-mediated) endocytosis. SNAT2 facilitated glutamine uptake and metabolism to promote VEGFC secretion, ultimately leading to lymphangiogenesis and LN metastasis in patients with BCa. Importantly, inhibition of UBE2C significantly attenuated BCa lymphangiogenesis in a patient-derived xenograft model. Our results reveal the mechanism by which UBE2C mediates crosstalk between the monoubiquitination and K63-linked polyubiquitination of SNAT2 to promote BCa metastasis and identify UBE2C as a promising target for treating LN-metastatic BCa.
    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
    尽管许多队列研究报道长期暴露于颗粒物(PM)会导致肺癌,PM诱导肺癌进展增加的分子机制仍不清楚.我们应用肺癌细胞系A549(Parental;A549。Par)延长PM,以建立模拟PM暴露的肺癌细胞系,A549.下午。我们的结果表明,A549。与A549相比,PM表现出更高的细胞生长和增殖能力。Par细胞在体外和体内。RNA测序分析发现双调蛋白(AREG)在PM诱导的细胞增殖中起关键作用。我们观察到PM通过谷氨酰胺代谢增加AREG依赖性肺癌增殖。此外,EGFR/PI3K/AKT/mTOR信号通路参与PM诱导的溶质载体家族A1成员5(SLC1A5)表达和谷氨酰胺代谢。我们的发现提供了有关暴露于PM后肺癌增殖如何发展的重要见解。
    Although many cohort studies have reported that long-term exposure to particulate matter (PM) causes lung cancer, the molecular mechanisms underlying the PM-induced increases in lung cancer progression remain unclear. We applied the lung cancer cell line A549 (Parental; A549.Par) to PM for an extended period to establish a mimic PM-exposed lung cancer cell line, A549.PM. Our results indicate that A549.PM exhibits higher cell growth and proliferation abilities compared to A549.Par cells in vitro and in vivo. The RNA sequencing analysis found amphiregulin (AREG) plays a critical role in PM-induced cell proliferation. We observed that PM increases AREG-dependent lung cancer proliferation through glutamine metabolism. In addition, the EGFR/PI3K/AKT/mTOR signaling pathway is involved in PM-induced solute carrier family A1 member 5 (SLC1A5) expression and glutamine metabolism. Our findings offer important insights into how lung cancer proliferation develops upon exposure to PM.
    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
    基因组不稳定性是对肿瘤异质性至关重要的癌症的标志,并且通常是细胞分裂和DNA损伤修复缺陷的结果。肿瘤耐受基因组不稳定性,但是遗传畸变的积累受到调节,以避免灾难性的染色体改变和细胞死亡。在卵巢癌肿瘤中,claudin-4经常上调,并与基因组不稳定和患者预后较差密切相关.然而,它与调节基因组不稳定性的生物学关联知之甚少。这里,我们使用CRISPR干扰和claudin模拟肽(CMP)在体外和体内调节claudin-4的表达及其功能。我们发现claudin-4通过在肿瘤细胞中产生微核来促进对基因组不稳定性的耐受机制。claudin-4的破坏增加了自噬,并与细胞质定位的DNA的吞噬有关。机械上,我们观察到claudin-4与调节mTOR上游自噬的氨基酸转运蛋白SLC1A5和LAT1建立了生物学轴.此外,claudin-4/SLC1A5/LAT1轴与氨基酸跨质膜转运相关,这是显著降低卵巢癌患者生存率的潜在细胞过程之一.一起,我们的研究结果表明,claudin-4的上调有助于通过自噬限制其积累,从而提高卵巢肿瘤细胞对基因组不稳定性的耐受阈值。
    Genome instability is a hallmark of cancer crucial for tumor heterogeneity and is often a result of defects in cell division and DNA damage repair. Tumors tolerate genomic instability, but the accumulation of genetic aberrations is regulated to avoid catastrophic chromosomal alterations and cell death. In ovarian cancer tumors, claudin-4 is frequently upregulated and closely associated with genome instability and worse patient outcomes. However, its biological association with regulating genomic instability is poorly understood. Here, we used CRISPR interference and a claudin mimic peptide to modulate the claudin-4 expression and its function in vitro and in vivo. We found that claudin-4 promotes a tolerance mechanism for genomic instability through micronuclei generation in tumor cells. Disruption of claudin-4 increased autophagy and was associated with the engulfment of cytoplasm-localized DNA. Mechanistically, we observed that claudin-4 establishes a biological axis with the amino acid transporters SLC1A5 and LAT1, which regulate autophagy upstream of mTOR. Furthermore, the claudin-4/SLC1A5/LAT1 axis was linked to the transport of amino acids across the plasma membrane as one of the potential cellular processes that significantly decreased survival in ovarian cancer patients. Together, our results show that the upregulation of claudin-4 contributes to increasing the threshold of tolerance for genomic instability in ovarian tumor cells by limiting its accumulation through autophagy.
    UNASSIGNED: Autophagy regulation via claudin-4/SLC1A5/LAT1 has the potential to be a targetable mechanism to interfere with genomic instability in ovarian tumor 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
    背景:肝细胞癌(HCC)是一种常见的恶性肿瘤,谷氨酰胺对肿瘤细胞至关重要。谷氨酰胺转运体SLC1A5在肿瘤进展和经动脉化疗栓塞(TACE)疗效中的作用正在研究中。本研究旨在确定SLC1A5表达对HCC预后和TACE疗效的影响,并阐明其机制。
    方法:SLC1A5在肝癌中的表达,与患者预后的相关性,和对TACE的反应在开放获取肝癌数据集中进行了研究,并在我们的队列中得到证实。此外,SLC1A5表达与缺氧的相关性,血管生成和免疫浸润分析,并通过免疫组织化学验证,免疫荧光和转录组测序。构建SLC1A5表达敲低或过表达的肝癌细胞系,和细胞增殖,菌落形成,凋亡,测量了体内异种移植肿瘤的迁移和药物敏感性。进行基因集富集分析以确定受SLC1A5影响的信号传导途径,并进行蛋白质印迹分析以检测蛋白质表达改变。
    结果:SLC1A5在HCC组织中的表达更高,并且与低生存率和TACE耐药相关。缺氧可以刺激谷氨酰胺转运的上调,血管生成和SLC1A5表达。SLC1A5表达与缺氧和血管生成相关基因呈正相关,免疫检查点途径,巨噬细胞,Tregs,和其他免疫抑制细胞浸润。SLC1A5的敲减减少增殖,菌落形成,和移民,但细胞凋亡增加,对化疗药物的敏感性增加。SLC1A5的下调导致波形蛋白和N-钙黏着蛋白表达的减少,然而E-cadherin表达增加。SLC1A5的上调增加了波形蛋白和N-钙粘蛋白的表达,同时减少E-cadherin。在SLC1A5敲低肝癌细胞系中过表达β-连环蛋白可以增加波形蛋白和N-钙黏着蛋白的表达,抑制E-cadherin表达,并增加迁移和耐药性。
    结论:升高的SLC1A5与HCC患者的TACE抵抗和生存缩短有关。SLC1A5与缺氧呈正相关,血管生成,和免疫抑制。SLC1A5可能通过上皮-间质转化(EMT)途径介导HCC细胞迁移和耐药。
    BACKGROUND: Hepatocellular carcinoma (HCC) is a common malignant tumor, and glutamine is vital for tumor cells. The role of glutamine transporter SLC1A5 in tumor progression and transarterial chemoembolization (TACE) efficacy is under study. This research seeks to determine the impact of SLC1A5 expression on the prognosis and TACE efficacy of HCC and elucidate its mechanisms.
    METHODS: SLC1A5 expression in HCC, correlation with patient outcomes, and response to TACE were studied in an open access liver cancer dataset and confirmed in our cohort. Additionally, the correlation between SLC1A5 expression and hypoxia, angiogenesis and immune infiltration was analyzed and verified by immunohistochemistry, immunofluorescence and transcriptome sequencing. Liver cancer cell lines with SLC1A5 expression knockdown or overexpression were constructed, and cell proliferation, colony formation, apoptosis, migration and drug sensitivity as well as in vivo xenograft tumor were measured. A gene set enrichment analysis was conducted to determine the signaling pathway influenced by SLC1A5, and a western blot analysis was performed to detect protein expression alterations.
    RESULTS: SLC1A5 expression was higher in HCC tissue and associated with poor survival and TACE resistance. Hypoxia could stimulate the upregulation of glutamine transport, angiogenesis and SLC1A5 expression. The SLC1A5 expression was positively correlated with hypoxia and angiogenesis-related genes, immune checkpoint pathways, macrophage, Tregs, and other immunosuppressive cells infiltration. Knockdown of SLC1A5 decreased proliferation, colony formation, and migration, but increased apoptosis and increased sensitivity to chemotherapy drugs. Downregulation of SLC1A5 resulted in a decrease in Vimentin and N-cadherin expression, yet an increase in E-cadherin expression. Upregulation of SLC1A5 increased Vimentin and N-cadherin expression, while decreasing E-cadherin. Overexpression of β-catenin in SLC1A5-knockdown HCC cell lines could augment Vimentin and N-cadherin expression, suppress E-cadherin expression, and increase the migration and drug resistance.
    CONCLUSIONS: Elevated SLC1A5 was linked to TACE resistance and survival shortening in HCC patients. SLC1A5 was positively correlated with hypoxia, angiogenesis, and immunosuppression. SLC1A5 may mediate HCC cell migration and drug resistance via Epithelial-mesenchymal transition (EMT) pathway.
    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
    背景:放射治疗(RT)可以驱动癌细胞进入细胞衰老状态,其中细胞可以分泌衰老相关的分泌表型(SASP)并产生小的细胞外囊泡(sEV)以与肿瘤微环境(TME)中的细胞相互作用。被受体细胞吸收的肿瘤来源的sEV有助于癌细胞代谢可塑性,对抗癌治疗的抗性,适应TME。然而,辐射诱导的sEV如何支持口腔鳞状细胞癌(OSCC)进展尚不清楚.
    方法:β-半乳糖苷酶染色和SASPmRNA表达分析用于评估照射后OSCC细胞的衰老相关活性。进行纳米粒子跟踪分析以鉴定辐射诱导的sEV。液相色谱-串联质谱(LC-MS)用于探索辐射诱导的sEV中蛋白质水平的变化。进行细胞计数试剂盒-8和集落形成测定以研究体外辐射诱导的SASP和sEV的功能。建立异种移植肿瘤模型以研究辐射诱导的sEV和V-9302在体内的功能以及潜在的机制。进行生物信息学分析以确定谷氨酰胺代谢与OSCC复发之间的关系。
    结果:我们确定辐射诱导的SASP触发了OSCC细胞增殖。此外,辐射诱导的sEV加剧了OSCC细胞的恶性程度。LC-MS/MS和生物信息学分析显示,SLC1A5是一种参与谷氨酰胺摄取的细胞受体,在辐射诱导的sEV中显著富集。在体外和体内,抑制SLC1A5可以阻断辐射诱导的sEV在OSCC中的致癌作用。
    结论:辐射诱导的sEV可能通过增强谷氨酰胺代谢促进未照射癌细胞的增殖;这可能是OSCC患者辐射抵抗的新分子机制。
    BACKGROUND: Radiotherapy (RT) can drive cancer cells to enter a state of cellular senescence in which cells can secrete senescence-associated secretory phenotype (SASP) and produce small extracellular vesicles (sEVs) to interact with cells in the tumor microenvironment (TME). Tumor-derived sEVs that are taken up by recipient cells contribute to cancer cell metabolic plasticity, resistance to anticancer therapy, and adaptation to the TME. However, how radiation-induced sEVs support oral squamous cell carcinoma (OSCC) progression remains unclear.
    METHODS: Beta-galactosidase staining and SASP mRNA expression analysis were used to evaluate the senescence-associated activity of OSCC cells after irradiation. Nanoparticle tracking analysis was performed to identify radiation-induced sEVs. Liquid chromatography-tandem mass spectrometry (LC-MS) was used to explore changes in the levels of proteins in radiation-induced sEVs. Cell Counting Kit-8 and colony formation assays were performed to investigate the function of radiation-induced SASP and sEVs in vitro. A xenograft tumor model was established to investigate the functions of radiation-induced sEVs and V-9302 in vivo as well as the underlying mechanisms. Bioinformatics analysis was performed to determine the relationship between glutamine metabolism and OSCC recurrence.
    RESULTS: We determined that the radiation-induced SASP triggered OSCC cell proliferation. Additionally, radiation-induced sEVs exacerbated OSCC cell malignancy. LC-MS/MS and bioinformatics analyses revealed that SLC1A5, which is a cellular receptor that participates in glutamine uptake, was significantly enriched in radiation-induced sEVs. In vitro and in vivo, inhibiting SLC1A5 could block the oncogenic effects of radiation-induced sEVs in OSCC.
    CONCLUSIONS: Radiation-induced sEVs might promote the proliferation of unirradiated cancer cells by enhancing glutamine metabolism; this might be a novel molecular mechanism underlying radiation resistance in OSCC patients.
    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
    作为一种构象受限的氨基酸,羟基-1-脯氨酸是用于合成多种多官能化吡咯烷以探测生物靶标的配体结合位点的通用支架。为了开发广泛表达的氨基酸转运蛋白SLC1A4和SLC1A5(也称为ASCT1和ASCT2)的新抑制剂,我们使用电生理和放射性标记的摄取方法对来自SLC1,SLC7和SLC38溶质载体家族的氨基酸转运蛋白进行了合成和功能筛选。我们发现了一类新型的烷氧基羟基吡咯烷羧酸(AHPC),可作为SLC1家族中性氨基酸转运蛋白SLC1A4和SLC1A5的选择性高亲和力抑制剂。将AHPC计算对接到同源性模型中,并评估预测的分子取向和功能活性。本文鉴定的一系列羟脯氨酸类似物代表了在药理学上调节SLC1A4和SLC1A5氨基酸交换剂的有前途的新试剂,这些试剂与许多病理生理过程如癌症和神经系统疾病有关。
    As a conformationally restricted amino acid, hydroxy-l-proline is a versatile scaffold for the synthesis of diverse multi-functionalized pyrrolidines for probing the ligand binding sites of biological targets. With the goal to develop new inhibitors of the widely expressed amino acid transporters SLC1A4 and SLC1A5 (also known as ASCT1 and ASCT2), we synthesized and functionally screened synthetic hydroxy-l-proline derivatives using electrophysiological and radiolabeled uptake methods against amino acid transporters from the SLC1, SLC7, and SLC38 solute carrier families. We have discovered a novel class of alkoxy hydroxy-pyrrolidine carboxylic acids (AHPCs) that act as selective high-affinity inhibitors of the SLC1 family neutral amino acid transporters SLC1A4 and SLC1A5. AHPCs were computationally docked into a homology model and assessed with respect to predicted molecular orientation and functional activity. The series of hydroxyproline analogs identified here represent promising new agents to pharmacologically modulate SLC1A4 and SLC1A5 amino acid exchangers which are implicated in numerous pathophysiological processes such as cancer and neurological diseases.
    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
    胰腺癌是一种高度致命的疾病,现有的治疗方法无效,因此,迫切需要开发新的有效治疗策略。胰腺癌细胞对葡萄糖和谷氨酰胺的高度依赖性表明,破坏这种依赖性可以作为胰腺癌治疗的替代策略。我们通过生物信息学分析确定了重要基因葡萄糖转运蛋白1(GLUT1)和丙氨酸-丝氨酸-半胱氨酸转运蛋白2(ASCT2),调节胰腺癌中葡萄糖和谷氨酰胺的代谢,分别。用于递送GLUT1和ASCT2抑制剂的人血清白蛋白纳米颗粒(HSANP),BAY-876/V-9302@HSANP,是通过自组装过程制备的。这种纳米药物通过释放的BAY-876和V-9302抑制胰腺癌细胞的葡萄糖和谷氨酰胺摄取,导致营养剥夺和氧化应激。谷氨酰胺的抑制导致谷胱甘肽合成的抑制,这进一步加剧了氧化应激。两者都导致活性氧的显著增加,激活caspase1和GSDMD,最终诱导焦亡。本研究为通过双重饥饿诱导的焦亡治疗原位胰腺癌提供了新的有效策略。利用生物信息学分析筛选代谢靶标,然后构建负载抑制剂的纳米药物的研究将激发未来胰腺癌的靶向代谢治疗。
    Pancreatic cancer is a highly fatal disease, and existing treatment methods are ineffective, so it is urgent to develop new effective treatment strategies. The high dependence of pancreatic cancer cells on glucose and glutamine suggests that disrupting this dependency could serve as an alternative strategy for pancreatic cancer therapy. We identified the vital genes glucose transporter 1 (GLUT1) and alanine-serine-cysteine transporter 2 (ASCT2) through bioinformatics analysis, which regulate glucose and glutamine metabolism in pancreatic cancer, respectively. Human serum albumin nanoparticles (HSA NPs) for delivery of GLUT1 and ASCT2 inhibitors, BAY-876/V-9302@HSA NPs, were prepared by a self-assembly process. This nanodrug inhibits glucose and glutamine uptake of pancreatic cancer cells through the released BAY-876 and V-9302, leading to nutrition deprivation and oxidative stress. The inhibition of glutamine leads to the inhibition of the synthesis of the glutathione, which further aggravates oxidative stress. Both of them lead to a significant increase in reactive oxygen species, activating caspase 1 and GSDMD and finally inducing pyroptosis. This study provides a new effective strategy for orthotopic pancreatic cancer treatment by dual starvation-induced pyroptosis. The study for screening metabolic targets using bioinformatics analysis followed by constructing nanodrugs loaded with inhibitors will inspire future targeted metabolic therapy for pancreatic cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号