argininosuccinate synthase 1

  • 文章类型: Journal Article
    柯萨奇病毒组B3(CVB3)属于小核糖核酸科肠道病毒属,是病毒性心肌炎(VMC)的主要病原体。对于这种情况没有特定的治疗方法。精氨酸琥珀酸合酶1(ASS1)是尿素循环中的关键酶,可将瓜氨酸和天冬氨酸转化为精氨酸琥珀酸。这里,我们发现CVB3及其衣壳蛋白VP2抑制ASS1的自噬降解,CVB3消耗瓜氨酸上调ASS1,触发尿素循环代谢重编程,然后激活巨噬细胞形成促炎极化,从而促进VMC的发生和发展。相反,补充瓜氨酸以防止耗竭可以下调ASS1,挽救巨噬细胞极化,减轻VMC的致病性。这些发现为VMC的发生和发展提供了新的视角,揭示ASS1是治疗这种疾病的潜在新靶点。
    目的:病毒性心肌炎(VMC)是一种常见且潜在危及生命的心肌炎性疾病,最常见的是CVB3感染。到目前为止,CVB3引起的VMC的发病机制主要集中在两个方面:一是感染早期病毒大量复制引起的直接心肌损伤,另一种是在适应性免疫反应阶段心肌局部免疫细胞浸润和炎症损伤。关于CVB3感染心肌组织早期固有免疫的研究较少,但巨噬细胞在CVB3感染早期的出现表明它们可以作为心肌组织中早期固有免疫反应细胞发挥调节作用。这里,我们发现了CVB3引起VMC的可能新机制,揭示了抗CVB3的新药物靶点,并发现了瓜氨酸对VMC的治疗潜力.
    Coxsackievirus group B3 (CVB3) belongs to the genus Enteroviruses of the family Picornaviridae and is the main pathogen underlying viral myocarditis (VMC). No specific therapeutic is available for this condition. Argininosuccinate synthase 1 (ASS1) is a key enzyme in the urea cycle that converts citrulline and aspartic acid to argininosuccinate. Here, we found that CVB3 and its capsid protein VP2 inhibit the autophagic degradation of ASS1 and that CVB3 consumes citrulline to upregulate ASS1, triggers urea cycle metabolic reprogramming, and then activates macrophages to develop pro-inflammatory polarization, thereby promoting the occurrence and development of VMC. Conversely, citrulline supplementation to prevent depletion can downregulate ASS1, rescue macrophage polarization, and alleviate the pathogenicity of VMC. These findings provide a new perspective on the occurrence and development of VMC, revealing ASS1 as a potential new target for treating this disease.
    OBJECTIVE: Viral myocarditis (VMC) is a common and potentially life-threatening myocardial inflammatory disease, most commonly caused by CVB3 infection. So far, the pathogenesis of VMC caused by CVB3 is mainly focused on two aspects: one is the direct myocardial injury caused by a large number of viral replication in the early stage of infection, and the other is the local immune cell infiltration and inflammatory damage of the myocardium in the adaptive immune response stage. There are few studies on the early innate immunity of CVB3 infection in myocardial tissue, but the appearance of macrophages in the early stage of CVB3 infection suggests that they can play a regulatory role as early innate immune response cells in myocardial tissue. Here, we discovered a possible new mechanism of VMC caused by CVB3, revealed new drug targets for anti-CVB3, and discovered the therapeutic potential of citrulline for VMC.
    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
    动脉粥样硬化起因于高脂血症下的内皮细胞(EC)功能障碍和血管炎症。Sirtuin3(SIRT3)是一种线粒体脱乙酰酶。然而,内皮细胞SIRT3在动脉粥样硬化过程中的具体作用尚不清楚.本研究旨在研究SIRT3在动脉粥样硬化过程中在EC功能中的作用及机制。野生型Sirt3f/f小鼠和内皮选择性SIRT3敲除Sirt3f/f;Cdh5Cre/+(Sirt3EC-KO)小鼠注射腺相关病毒(AAV)以过表达PCSK9,并饲喂高胆固醇饮食(HCD)12周以诱导动脉粥样硬化。Sirt3EC-KO小鼠表现出增加的动脉粥样硬化斑块形成,随着巨噬细胞浸润的增加,血管炎症,和降低循环L-精氨酸水平。在人类ECs中,SIRT3抑制导致血管炎症加剧,减少一氧化氮(NO)的产生,增加活性氧(ROS),和减少L-精氨酸水平。SIRT3的沉默会导致精氨酸琥珀酸合酶1(ASS1)的高乙酰化和失活,一种参与L-精氨酸生物合成的限速酶,这种效应在突变型ASS1中消失。此外,在Sirt3EC-KO小鼠中,L-精氨酸补充减弱增强的斑块形成和血管炎症。这项研究提供了令人信服的证据支持内皮SIRT3在动脉粥样硬化中的保护作用,并且还表明SIRT3诱导的ECs对ASS1的去乙酰化对精氨酸合成的关键作用。
    Atherosclerosis is initiated with endothelial cell (EC) dysfunction and vascular inflammation under hyperlipidemia. Sirtuin 3 (SIRT3) is a mitochondrial deacetylase. However, the specific role of endothelial SIRT3 during atherosclerosis remains poorly understood. The present study aims to study the role and mechanism of SIRT3 in EC function during atherosclerosis. Wild-type Sirt3f/f mice and endothelium-selective SIRT3 knockout Sirt3f/f; Cdh5Cre/+ (Sirt3EC-KO) mice are injected with adeno-associated virus (AAV) to overexpress PCSK9 and fed with high-cholesterol diet (HCD) for 12 weeks to induce atherosclerosis. Sirt3EC-KO mice exhibit increased atherosclerotic plaque formation, along with elevated macrophage infiltration, vascular inflammation, and reduced circulating L-arginine levels. In human ECs, SIRT3 inhibition resulted in heightened vascular inflammation, reduced nitric oxide (NO) production, increased reactive oxygen species (ROS), and diminished L-arginine levels. Silencing of SIRT3 results in hyperacetylation and deactivation of Argininosuccinate Synthase 1 (ASS1), a rate-limiting enzyme involved in L-arginine biosynthesis, and this effect is abolished in mutant ASS1. Furthermore, L-arginine supplementation attenuates enhanced plaque formation and vascular inflammation in Sirt3EC-KO mice. This study provides compelling evidence supporting the protective role of endothelial SIRT3 in atherosclerosis and also suggests a critical role of SIRT3-induced deacetylation of ASS1 by ECs for arginine synthesis.
    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
    越来越多的证据表明ASS1与肿瘤密切相关。没有ASS1的泛癌症分析可用。
    在这里,我们基于TCGA(癌症基因组图谱)的数据集,探索了33种肿瘤中ASS1的基因表达和生存分析,GEO(基因表达综合),和GEPIA2(基因表达谱交互式分析,版本2)。
    ASS1在大多数正常组织中高表达,并且与某些肿瘤的进展有关。我们还报告了ASS1遗传改变及其与肿瘤预后的关系,并报告了肿瘤和对照正常组织之间ASS1磷酸化位点的差异。ASS1的表达与BRCA(乳腺浸润性癌)的TCGA肿瘤的癌相关成纤维细胞(CAFs)的浸润有关,CESC(宫颈鳞状细胞癌和宫颈腺癌),COAD(结肠腺癌),ESCA(食管癌),SKCM(皮肤皮肤黑色素瘤),SKCM-转移,TGCT(睾丸生殖细胞肿瘤),和BRCA肿瘤的内皮细胞,BRCA-基础,CESC,ESCA,KIRC(肾肾透明细胞癌),LUAD(肺腺癌),LUSC(肺鳞状细胞癌),SKCM,SKCM-转移,SKCM-Primary,STAD(胃腺癌),和TGCT。KEGG和GO分析用于分析ASS1相关信号通路。最后,我们用Huh7细胞系在体外验证ASS1的功能。在使用小干扰RNA(siRNA)进行ASS1敲低后,Huh7的增殖和侵袭增强,细胞周期蛋白D1上调,抗凋亡蛋白bax下调,提示ASS1是肝细胞癌的抑癌基因。
    我们的第一项泛癌症研究提供了对ASS1在不同肿瘤中的作用的相对全面的了解。
    UNASSIGNED: There is accumulating evidence indicating that ASS1 is closely related to tumors. No pan-cancer analysis of ASS1 was available.
    UNASSIGNED: Here we explored the gene expression and survival analysis of ASS1 across thirty-three tumors based on the datasets of the TCGA (Cancer Genome Atlas), the GEO (Gene Expression Omnibus), and the GEPIA2 (Gene Expression Profiling Interactive Analysis, version 2).
    UNASSIGNED: ASS1 is highly expressed in most normal tissues and is related to the progression of some tumors. We also report ASS1 genetic alteration and their association with tumor prognosis and report differences in ASS1 phosphorylation sites between tumors and control normal tissues. ASS1 expression was associated with the infiltration of cancer-associated fibroblasts (CAFs) for the TCGA tumors of BRCA (Breast invasive carcinoma), CESC (Cervical squamous cell carcinoma and endocervical adenocarcinoma), COAD (Colon adenocarcinoma), ESCA (Esophageal carcinoma), SKCM (Skin cutaneous melanoma), SKCM-Metastasis, TGCT (Testicular germ cell tumors), and endothelial cell for the tumors of BRCA, BRCA-Basal, CESC, ESCA, KIRC (Kidney renal clear cell carcinoma), LUAD (Lung adenocarcinoma), LUSC (Lung squamous cell carcinoma), SKCM, SKCM-Metastasis, SKCM-Primary, STAD (Stomach adenocarcinoma), and TGCT. The KEGG and GO analysis were used to analyze ASS1-related signaling pathways. Finally, we used Huh7 cell line to verify the function of ASS1 in vitro. After ASS1 knockdown using small interfering RNA (siRNA), the proliferation and invasion of Huh7 were enhanced, cyclin D1 was up-regulated, and anti-apoptotic protein bax was down-regulated, suggesting that ASS1 is a tumor suppressor gene in hepatocellular carcinoma.
    UNASSIGNED: Our first pan-cancer study offers a relatively comprehensive understanding of the roles of ASS1 in different tumors.
    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
    精氨酸剥夺作为一种新颖且安全的抗代谢策略,已获得越来越多的吸引力,用于治疗几种难以治疗的癌症,其特征是严重依赖精氨酸。小细胞肺癌(SCLC)由于限速酶精氨酸琥珀酸合成酶1(ASS1)的失活而表现出明显的精氨酸营养缺陷型,因此,可以用聚乙二醇化的精氨酸脱亚胺酶或ADI-PEG20(peggargiminase)和人重组聚乙二醇化的精氨酸酶(rhArgPEG,BCT-100和聚乙二醇精氨酸酶)。尽管临床前研究表明ASS1缺陷型SCLC细胞系对精氨酸降解酶高度敏感,到目前为止,与诊所的活动明显脱节,这可能部分归因于患者的选择。最近的研究已经探索了针对肿瘤适应的精氨酸耗竭的抗性机制,如ASS1再表达和自噬,基质细胞输入,包括巨噬细胞浸润,和肿瘤异质性。这里,我们探讨了精氨酸剥夺如何与新型药物战略性结合,通过调节耐药性和提高现有药物的疗效来改善SCLC管理.此外,最近的工作已经确定了靶向精氨酸联合PD-1/PD-L1免疫检查点抑制剂的有趣作用,并且临床试验正在进行中.因此,未来的精氨酸消耗药物与化学免疫疗法的研究,SCLC的现行标准,可能导致疾病控制增强和患者长期生存急需改善。
    Arginine deprivation has gained increasing traction as a novel and safe antimetabolite strategy for the treatment of several hard-to-treat cancers characterised by a critical dependency on arginine. Small cell lung cancer (SCLC) displays marked arginine auxotrophy due to inactivation of the rate-limiting enzyme argininosuccinate synthetase 1 (ASS1), and as a consequence may be targeted with pegylated arginine deiminase or ADI-PEG20 (pegargiminase) and human recombinant pegylated arginases (rhArgPEG, BCT-100 and pegzilarginase). Although preclinical studies reveal that ASS1-deficient SCLC cell lines are highly sensitive to arginine-degrading enzymes, there is a clear disconnect with the clinic with minimal activity seen to date that may be due in part to patient selection. Recent studies have explored resistance mechanisms to arginine depletion focusing on tumor adaptation, such as ASS1 re-expression and autophagy, stromal cell inputs including macrophage infiltration, and tumor heterogeneity. Here, we explore how arginine deprivation may be combined strategically with novel agents to improve SCLC management by modulating resistance and increasing the efficacy of existing agents. Moreover, recent work has identified an intriguing role for targeting arginine in combination with PD-1/PD-L1 immune checkpoint inhibitors and clinical trials are in progress. Thus, future studies of arginine-depleting agents with chemoimmunotherapy, the current standard of care for SCLC, may lead to enhanced disease control and much needed improvements in long-term survival for 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
    背景:肾癌是美国常见的成人恶性肿瘤。透明细胞肾细胞癌(ccRCC),肾癌的主要亚型,以广泛的代谢变化为特征。尿素代谢是ccRCC中的一种这样的改变的途径。这项研究的目的是阐明尿素循环酶的贡献,精氨酸琥珀酸合酶1(ASS1),和精氨酸琥珀酸裂解酶(ASL)对ccRCC进展的影响。
    方法:我们采用了计算,遗传,和代谢组学工具以及体内动物模型,以建立ASS1和ASL在ccRCC中的肿瘤抑制作用。
    结果:我们显示,与正常肾脏相比,ccRCC肿瘤中尿素循环酶ASS1和ASL的mRNA和蛋白表达降低。此外,HK-2细胞(永生化的肾上皮细胞)中ASL的丢失促进了2D和3D生长测定中的生长,虽然ASS1和ASL在ccRCC细胞系中的联合再表达抑制了2D的生长,3D,和体内异种移植模型。我们确定这种抑制取决于它们的酶活性。最后,我们证明了细胞天冬氨酸的保守性,一氧化氮合成的调节,嘧啶的产生在ASS1+ASL介导的ccRCC生长抑制中起关键作用。
    结论:ccRCC肿瘤下调尿素循环的组分,包括精氨酸琥珀酸合酶1(ASS1)和精氨酸琥珀酸裂解酶(ASL)。这些细胞溶质酶位于细胞中的关键代谢中心,并参与天冬氨酸分解代谢以及精氨酸和一氧化氮的生物合成。ASS1和ASL的缺失有助于细胞将天冬氨酸重定向到嘧啶合成并支持增强的增殖。此外,降低ASS1和ASL水平可能有助于调节一氧化氮(NO)的产生并减轻其细胞毒性作用。总的来说,我们的工作增加了对尿素循环酶的理解,它们在ccRCC进展中的作用,并发现ccRCC中新的潜在代谢漏洞。
    BACKGROUND: Kidney cancer is a common adult malignancy in the USA. Clear cell renal cell carcinoma (ccRCC), the predominant subtype of kidney cancer, is characterized by widespread metabolic changes. Urea metabolism is one such altered pathway in ccRCC. The aim of this study was to elucidate the contributions of urea cycle enzymes, argininosuccinate synthase 1 (ASS1), and argininosuccinate lyase (ASL) towards ccRCC progression.
    METHODS: We employed a combination of computational, genetic, and metabolomic tools along with in vivo animal models to establish a tumor-suppressive role for ASS1 and ASL in ccRCC.
    RESULTS: We show that the mRNA and protein expression of urea cycle enzymes ASS1 and ASL are reduced in ccRCC tumors when compared to the normal kidney. Furthermore, the loss of ASL in HK-2 cells (immortalized renal epithelial cells) promotes growth in 2D and 3D growth assays, while combined re-expression of ASS1 and ASL in ccRCC cell lines suppresses growth in 2D, 3D, and in vivo xenograft models. We establish that this suppression is dependent on their enzymatic activity. Finally, we demonstrate that conservation of cellular aspartate, regulation of nitric oxide synthesis, and pyrimidine production play pivotal roles in ASS1+ASL-mediated growth suppression in ccRCC.
    CONCLUSIONS: ccRCC tumors downregulate the components of the urea cycle including the enzymes argininosuccinate synthase 1 (ASS1) and argininosuccinate lyase (ASL). These cytosolic enzymes lie at a critical metabolic hub in the cell and are involved in aspartate catabolism and arginine and nitric oxide biosynthesis. Loss of ASS1 and ASL helps cells redirect aspartate towards pyrimidine synthesis and support enhanced proliferation. Additionally, reduced levels of ASS1 and ASL might help regulate nitric oxide (NO) generation and mitigate its cytotoxic effects. Overall, our work adds to the understanding of urea cycle enzymes in a context-independent of ureagenesis, their role in ccRCC progression, and uncovers novel potential metabolic vulnerabilities in ccRCC.
    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
    Snail is a dedicated transcriptional repressor and acts as a master inducer of EMT and metastasis, yet the underlying signaling cascades triggered by Snail still remain elusive. Here, we report that Snail promotes colorectal cancer (CRC) migration by preventing non-coding RNA LOC113230-mediated degradation of argininosuccinate synthase 1 (ASS1). LOC113230 is a novel Snail target gene, and Snail binds to the functional E-boxes within its proximal promoter to repress its expression in response to TGF-β induction. Ectopic expression of LOC113230 potently suppresses CRC cell growth, migration, and lung metastasis in xenograft experiments. Mechanistically, LOC113230 acts as a scaffold to facilitate recruiting LRPPRC and the TRAF2 E3 ubiquitin ligase to ASS1, resulting in enhanced ubiquitination and degradation of ASS1 and decreased arginine synthesis. Moreover, elevated ASS1 expression is essential for CRC growth and migration. Collectively, these findings suggest that TGF-β and Snail promote arginine synthesis via inhibiting LOC113230-mediated LRPPRC/TRAF2/ASS1 complex assembly and this complex can serve as potential target for the development of new therapeutic approaches to treat CRC.
    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
    Cancer cells are required to rewire existing metabolic pathways to support their abnormal proliferation. We have previously shown that, unlike glucose-addicted cancers, Kaposi\'s sarcoma-associated herpesvirus (KSHV)-transformed cells depend on glutamine rather than glucose for energy production and amino acid and nucleotide syntheses. High-level consumption of glutamine is tightly regulated and often coupled with the citrulline-nitric oxide (NO) cycle. We have found that KSHV infection accelerates nitrogen efflux by upregulating the expression of argininosuccinate synthase 1 (ASS1), a key enzyme in the citrulline-NO cycle. KSHV utilizes multiple microRNAs to upregulate ASS1 expression. Depletion of either ASS1 or inducible nitric oxide synthase (iNOS) in KSHV-transformed cells suppresses growth proliferation, abolishes colony formation in soft agar, and decreases NO generation. Furthermore, by maintaining intracellular NO levels, ASS1 expression facilitates KSHV-mediated activation of the STAT3 pathway, which is critical for virus-induced transformation. These results illustrate a novel mechanism by which an oncogenic virus hijacks a key metabolic pathway to promote growth transformation and reveal a potential novel therapeutic target for KSHV-induced malignancies.IMPORTANCE We have previously shown that Kaposi\'s sarcoma-associated herpesvirus (KSHV)-transformed cells depend on glutamine rather than glucose for energy production and amino acid and nucleotide syntheses. In this study, we have further examined how the KSHV-reprogramed metabolic pathways are regulated and discovered that KSHV hijacks the citrulline-nitric oxide (NO) cycle to promote growth proliferation and transformation. Multiple KSHV-encoded microRNAs upregulate argininosuccinate synthase 1 (ASS1), a key enzyme in the citrulline-NO cycle. ASS1 is required for KSHV-induced proliferation, colony formation in soft agar, and NO generation of KSHV-transformed cells, which also depends on inducible nitric oxide synthase. By maintaining intracellular NO levels, ASS1 mediates KSHV activation of the STAT3 pathway, which is essential for KSHV-induced abnormal cell proliferation and transformation. These results illustrate a novel mechanism by which an oncogenic virus hijacks a key metabolic pathway to promote growth transformation and reveal a potential novel therapeutic target for KSHV-induced malignancies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The significant disparities in metabolism between tumor and normal cells have inspired the development of metabolism-based anti-tumor therapeutics. Arginine is a semi-essential amino acid because normal cells can not only synthesize arginine de novo but also take up extracellular arginine. Several types of tumors have abnormalities in arginine metabolism enzymes and completely rely on extracellular arginine to support necessary biological processes. This property is referred to as arginine auxotrophy. Taking advantage of characteristic arginine auxotrophy in tumors, arginine deprivation, which is generally induced by the use of arginine deiminase (ADI) and arginase I, has been investigated as a novel strategy for cancer therapy. Arginine deprivation demonstrated promising efficacy against arginine-auxotrophic tumors. By integrating perspectives from both clinical oncologists and laboratory scientists, this article reviews the important aspects of arginine deprivation as a promising anticancer therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    L-Arginine (L-Arg), a conditional essential amino acid in adults, has been shown to enhance pregnancy outcome. Argininosuccinate synthase (Ass1) and argininosuccinate lyase (Asl) are the key enzyme for L-Arginine (L-Arg) biosynthesis. Based our microarray analysis, Ass1 expression is upregulated significantly at implantation site on day 5 of pregnancy compared to that at inter-implantation site. However, the expression, regulation and function of Ass1 during early pregnancy remain unknown. Here we found that Ass1 is highly expressed in mouse decidua and uterine stromal cells undergoing decidualization, and Asl is weakly expressed in mouse decidua and uterine stromal cells undergoing decidualization. α-Methyl-DL-aspartic acid (MDLA), a specific inhibitor for Ass1, can significantly increase the rate of embryonic reabsorption. Under in vitro induced decidualization, MDLA clearly inhibits the expression of decidual/trophoblast prolactin-related protein (Dtprp), a marker for decidualization in mice. Only Ass1 expression is induced by cAMP through PKA/p-Creb signaling pathway. Results from our cell culture models further indicates that the high level of L-Arg enhances stromal proliferation, while enzymatic activity or Ass1 expression level is essential to determine the magnitude of both mouse and human decidualization. Interestingly, L-Arg at high concentration down-regulates Ass1 and Asl expression by negative feedback to maintain L-Arg homeostasis. These findings highlight that cAMP-induced Ass1 expression is important in controlling the magnitude of decidualization through regulating L-Arg level.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号