RARRES1

RARRES1
  • 文章类型: Journal Article
    皮肤黑素瘤(SKCM)是一种高度恶性的皮肤癌,以预后不良和死亡率升高而闻名。RARRES1,一种对视黄酸受体有反应的基因,在各种癌症类型中显示不同的功能。然而,RARRES1在SKCM中的具体作用和潜在机制尚不清楚。GSE15605用于分析RARRES1在SKCM中的表达。随后,TCGA和GEO数据库用于研究RARRES1与临床病理参数之间的关系,以及RARRES1在SKCM中的预后意义和诊断效能。GO,KEGG,进行GSEA分析以探索RARRES1的潜在功能。此外,研究了RARRES1与免疫浸润之间的关联.使用cBioPortal评估SKCM中RARRES1的基因组改变和启动子甲基化水平,UALCAN,和GEO数据库。最后,通过免疫组织化学验证SKCM中RARRES1的表达,通过体内和体外实验阐明了其在SKCM进展中的功能作用。我们发现与正常组织相比,RARRES1在SKCM中下调,这种低表达与SKCM的临床病理特征较差和预后不良有关。通过ROC分析确定RARRES1的诊断效能,是0.732。通过GO,KEGG,和GSEA富集分析,我们确定了30个主要富集在肿瘤免疫微环境中的相关基因和通路,扩散,凋亡,和自噬。此外,发现RARRES1表达与SKCM中各种免疫细胞的浸润呈正相关,特别是巨噬细胞和T辅助细胞,在其他人中。基因组改变和启动子甲基化的分析表明,RARRES1启动子的浅缺失和超甲基化可能导致RARRES1表达降低。IHC验证证实了SKCM中RARRES1的下调。此外,过表达RARRES1抑制A375细胞的增殖和迁移,促进细胞凋亡,并抑制自噬通量。在小鼠异种移植模型中,RARRES1过表达也抑制SKCM肿瘤生长。总的来说,这些研究结果表明,RARRES1可能起到抑制因子的作用,并可能作为SKCM的预后生物标志物和治疗靶点.
    Skin cutaneous melanoma (SKCM) is a highly malignant form of skin cancer, known for its unfavorable prognosis and elevated mortality rate. RARRES1, a gene responsive to retinoic acid receptors, displays varied functions in various cancer types. However, the specific role and underlying mechanisms of RARRES1 in SKCM are still unclear. GSE15605 was utilized to analyze the expression of RARRES1 in SKCM. Subsequently, the TCGA and GEO databases were employed to investigate the relationships between RARRES1 and clinicopathological parameters, as well as the prognostic implications and diagnostic efficacy of RARRES1 in SKCM. GO, KEGG, and GSEA analyses were conducted to explore the potential functions of RARRES1. Furthermore, the associations between RARRES1 and immune infiltration were examined. Genomic alterations and promoter methylation levels of RARRES1 in SKCM were assessed using cBioPortal, UALCAN, and the GEO database. Finally, RARRES1 expression in SKCM was validated through immunohistochemistry, and its functional role in SKCM progression was elucidated via in vivo and in vitro experiments. We found that RARRES1 was downregulated in SKCM compared with normal tissues, and this low expression was associated with worse clinicopathological features and poor prognosis of SKCM. The diagnostic efficacy of RARRES1, as determined by ROC analysis, was 0.732. Through GO, KEGG, and GSEA enrichment analysis, we identified 30 correlated genes and pathways that were mainly enriched in the tumor immune microenvironment, proliferation, apoptosis, and autophagy. Additionally, RARRES1 expression was found to be positively related to the infiltration of various immune cells in SKCM, particularly macrophages and T helper cells, among others. Analysis of genomic alterations and promoter methylation revealed that shallow deletion and hypermethylation of the RARRES1 promoter could lead to reduced RARRES1 expression. IHC validation confirmed the downregulation of RARRES1 in SKCM. Moreover, overexpression of RARRES1 inhibited the proliferation and migration of A375 cells, promoted apoptosis, and inhibited autophagic flux. In the mouse xenograft model, RARRES1 overexpression also suppressed SKCM tumor growth. Collectively, these findings suggest that RARRES1 may function as a suppressor and could potentially serve as a prognostic biomarker and therapeutic target for SKCM.
    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
    视黄酸受体反应蛋白-1(RARRES1)是一种富含足细胞的跨膜蛋白,其表达增加与人类肾小球疾病进展相关。RARRES1通过p53介导的足细胞凋亡促进足细胞减少和肾小球硬化。重要的是,RARRES1的细胞病变作用完全依赖于其蛋白水解裂解成可溶性蛋白(sRARRES1)和随后通过内吞作用摄取足细胞,作为切割突变体RARRES1在体外或体内没有作用。由于RARRES1表达在人类肾小球疾病中上调,在这里,我们研究了足细胞特异性过表达RARRES1在实验性局灶节段性肾小球硬化和糖尿病肾病小鼠中的功能后果。我们还研究了长期RARRES1过表达对缓慢发展的衰老诱导的肾损伤的影响。如预期,足细胞过度表达RARRES1(Pod-RARRES1WT)的诱导在所有三种模型中都显着恶化了肾小球损伤和肾功能恶化,而RARRES1切割突变体(Pod-RARRES1MT)的过表达则没有。值得注意的是,在受伤的Pod-RARRES1WT小鼠的近端小管中也观察到sRARRES1的直接摄取,并与加剧的肾小管损伤有关,空泡化,和脂质积累。小鼠肾脏的单细胞RNA序列分析表明,RARRES1导致近端小管亚群脂质代谢明显失调。我们进一步确定基质金属蛋白酶23(MMP23)是一种高度足细胞特异性的金属蛋白酶,并在疾病环境中负责RARRES1裂解。作为腺相关病毒9介导的MMP23敲低,体内肾小管细胞中sRARRES1的摄取。因此,我们的研究描述了一种以前未被认识到的机制,通过这种机制,足细胞来源的蛋白在肾小球疾病中直接促进足细胞和肾小管损伤,并提示RARRES1和MMP23的足细胞特异性功能可能是体内改善肾小球疾病进展的目标.
    Retinoic acid receptor responder protein-1 (RARRES1) is a podocyte-enriched transmembrane protein whose increased expression correlates with human glomerular disease progression. RARRES1 promotes podocytopenia and glomerulosclerosis via p53-mediated podocyte apoptosis. Importantly, the cytopathic actions of RARRES1 are entirely dependent on its proteolytic cleavage into a soluble protein (sRARRES1) and subsequent podocyte uptake by endocytosis, as a cleavage mutant RARRES1 exerted no effects in vitro or in vivo. As RARRES1 expression is upregulated in human glomerular diseases, here we investigated the functional consequence of podocyte-specific overexpression of RARRES1 in mice in the experimental focal segmental glomerulosclerosis and diabetic kidney disease. We also examined the effects of long-term RARRES1 overexpression on slowly developing aging-induced kidney injury. As anticipated, the induction of podocyte overexpression of RARRES1 (Pod-RARRES1WT) significantly worsened glomerular injuries and worsened kidney function in all three models, while overexpression of RARRES1 cleavage mutant (Pod-RARRES1MT) did not. Remarkably, direct uptake of sRARRES1 was also seen in proximal tubules of injured Pod-RARRES1WT mice and associated with exacerbated tubular injuries, vacuolation, and lipid accumulation. Single-cell RNA sequence analysis of mouse kidneys demonstrated RARRES1 led to a marked deregulation of lipid metabolism in proximal tubule subsets. We further identified matrix metalloproteinase 23 (MMP23) as a highly podocyte-specific metalloproteinase and responsible for RARRES1 cleavage in disease settings, as adeno-associated virus 9-mediated knockdown of MMP23 abrogated sRARRES1 uptake in tubular cells in vivo. Thus, our study delineates a previously unrecognized mechanism by which a podocyte-derived protein directly facilitates podocyte and tubular injury in glomerular diseases and suggests that podocyte-specific functions of RARRES1 and MMP23 may be targeted to ameliorate glomerular disease progression in vivo.
    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
    三阴性乳腺癌(TNBC)是乳腺癌的一种亚型,预后差,治疗选择有限。尽管免疫检查点抑制剂(ICIs)已被证明可以改善TNBC患者的预后,决定ICIs治疗反应的潜在机制和标志物仍不确定.揭示癌细胞与肿瘤微环境(TME)之间的关系和相互作用可能有助于预测治疗效果和开发新的治疗剂。通过分析单细胞RNA测序数据集,我们全面分析了细胞类型和亚群,并在TNBC的TME中鉴定了它们的特征。我们还提出了一种定量评估TME免疫谱的方法,并提供了通过整合分析鉴定与TME相关的癌细胞内在特征的框架。使用综合分析,RARRES1被鉴定为TME相关基因,其表达与TNBC的预后和对ICIs的反应呈正相关。总之,这项研究描述了TNBC患者TME中细胞成分的异质性,并为癌细胞和TME之间的关系带来了新的见解。此外,RARRES1被确定为TNBC中ICIs预后和反应的潜在预测因子。
    Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor prognosis and limited treatment options. Although immune checkpoint inhibitors (ICIs) have been proven to improve outcomes in TNBC patients, the potential mechanisms and markers that determine the therapeutic response to ICIs remains uncertain. Revealing the relationship and interaction between cancer cells and tumor microenvironment (TME) could be helpful in predicting treatment efficacy and developing novel therapeutic agents. By analyzing single-cell RNA sequencing dataset, we comprehensively profiled cell types and subpopulations as well as identified their signatures in the TME of TNBC. We also proposed a method for quantitatively assessment of the TME immune profile and provided a framework for identifying cancer cell-intrinsic features associated with TME through integrated analysis. Using integrative analyses, RARRES1 was identified as a TME-associated gene, whose expression was positively correlated with prognosis and response to ICIs in TNBC. In conclusion, this study characterized the heterogeneity of cellular components in TME of TNBC patients, and brought new insights into the relationship between cancer cells and TME. In addition, RARRES1 was identified as a potential predictor of prognosis and response to ICIs in TNBC.
    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:Lenvatinib是一种口服小分子抑制剂,在全球范围内被批准用于治疗不可切除的肝细胞癌(HCC)患者。增加细胞对乐伐替尼的敏感性将是提高治疗效果的有效方法。
    方法:采用高通量方法扫描肝癌细胞中与乐伐替尼敏感性相关的差异表达基因(DEGs)。使用增益和损失功能实验来探索这些DEG在HCC中的功能和lenvatinib敏感性。CO-IP测定和救援实验用于研究机制。
    结果:我们确定RAR应答蛋白1(RARRES1),足细胞特异性生长停滞基因,在lenvatinib治疗后,肝癌细胞中DEGs显著上调。功能分析显示,异位RARRES1表达在体外和体内降低HCC进展,以及提高肿瘤对乐伐替尼的敏感性,而RARRES1沉默可增加HCC细胞的增殖和迁移。机械上,免疫共沉淀实验表明,RARRES1与HCC细胞中的丝氨酸蛋白酶抑制剂Kazal-2型(SPINK2)相互作用。Further,SPINK2过表达抑制HCC细胞增殖和迁移,以及增加对乐伐替尼的敏感性,而SPINK2敲低促进细胞进展并降低乐伐替尼敏感性。肝癌组织标本中RARRES1和SPINK2的mRNA和蛋白水平较低,相对于正常肝组织。
    结论:我们的发现强调RARRES1可以通过与SPINK2相互作用来抑制HCC进展并调节HCC对乐伐替尼的敏感性,这代表了HCC中一种新的肿瘤抑制因子RARRES1/SPINK2轴,可调节对乐伐替尼的敏感性。
    Lenvatinib is an oral small molecule inhibitor approved for treating patients with unresectable hepatocellular carcinoma (HCC) worldwide. Increasing cell sensitivity to lenvatinib would be an effective method of improving therapeutic efficacy.
    High throughput methods was used to scan the differentially expressed genes (DEGs) related to lenvatinib sensitivity in HCC cells. Gain- and loss-function experiments were used to explore the functions of these DEGs in HCC and lenvatinib sensitivity. CO-IP assay and rescue experiments were utilized to investigate the mechanism.
    We identified that RAR responder protein 1 (RARRES1), a podocyte-specific growth arrest gene, was among significantly upregulated DEGs in HCC cells following lenvatinib treatment. Functional analysis showed that ectopic RARRES1 expression decreased HCC progression in vitro and in vivo, as well as improving tumor sensitivity to lenvatinib, while RARRES1 silencing increased HCC cell proliferation and migration. Mechanistically, co-immunoprecipitation assays demonstrated that RARRES1 interacted with serine protease inhibitor Kazal-type 2 (SPINK2) in HCC cells. Further, SPINK2 overexpression suppressed HCC cell proliferation and migration, as well as increasing sensitivity to lenvatinib whereas SPINK2 knockdown promoted cell progression and decreased lenvatinib sensitivity. The mRNA and protein levels of RARRES1 and SPINK2 were low in HCC tissue samples, relative to those in normal liver tissue.
    Our findings highlighted that RARRES1 can inhibit HCC progression and regulate HCC sensitivity to lenvatinib by interacting SPINK2, representing a new tumor suppressor RARRES1/SPINK2 axis in HCC that modulates sensitivity to lenvatinib.
    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
    RARRES1是一种肿瘤抑制蛋白,并且其在各种肿瘤细胞中的表达被抑制。然而,是否参与肾透明细胞癌(KIRC)的免疫反应尚不清楚,定义的机制不清楚。因此,RARRES1在KIRC中的作用机制值得研究。
    我们用癌症基因组图谱(TCGA)数据库分析了RARRES1的表达和功能。采用Kaplan-Meier曲线估计生存期。从UALCAN数据库获得RARRES1相关基因,并进行基因本体论(GO)富集和蛋白质-蛋白质相互作用(PPI)网络分析。使用TIMER数据库进行肿瘤浸润免疫细胞与所选基因之间的相关性分析。我们还通过将Caki-1细胞与THP-1细胞共培养,研究了RARRES1在KIRC中的可能功能。免疫荧光法检测不同分级KIRC组织中RARRES1的表达。
    在KIRC患者中,RARRES1的表达与生存呈负相关。与RARRES1相关基因最显著富集的GO生物学过程术语是细胞粘附的调节。与RARRES1表现出相对最高的相关性的ICAM1与巨噬细胞的浸润水平正相关。RARRES1可以增强Caki-1细胞中ICAM1的表达,然后诱导THP-1M1细胞的活化,从而降低Caki-1细胞的活力并诱导其凋亡。
    RARRES1通过促进ICAM1表达和诱导M1巨噬细胞活化而发挥抗肿瘤作用。我们提供了对KIRC分子机制的见解,并揭示了潜在的治疗靶标。
    RARRES1 is a tumor suppressor protein, and its expression is suppressed in various tumor cells. However, whether it participates in the immune response in kidney renal clear cell carcinoma (KIRC) is unknown, and the defined mechanism is not clear. Therefore, the mechanism of RARRES1 in KIRC is worthy of investigation.
    We analysed the expression and function of RARRES1 with The Cancer Genome Atlas (TCGA) database. The Kaplan-Meier curve was adopted to estimate survival. RARRES1-correlated genes were obtained from the UALCAN database and subjected to Gene Ontology (GO) enrichment and protein-protein interaction (PPI) network analyses. The correlation analysis between tumor-infiltrating immune cells and selected genes were performed with TIMER database. We also investigated the possible function of RARRES1 in KIRC by coculturing Caki-1 cells with THP-1 cells. Immunofluorescence assay was performed to study the RARRES1 expression in difference grade KIRC tissues.
    The expression of RARRES1 was negatively correlated with survival in KIRC patients. The GO biological process term most significantly enriched with the RARRES1-correlated genes was regulation of cell adhesion. ICAM1, which exhibited a relatively highest correlation with RARRES1, is positively correlated with the infiltration level of macrophages. RARRES1 could enhance the expression of ICAM1 in Caki-1 cells and then induce the activation of M1 THP-1 cells to decrease the viability and induce the apoptosis of Caki-1 cells.
    RARRES1 plays an antitumor role by promoting ICAM1 expression and inducing the activation of M1 macrophages. We offer insights into the molecular mechanism of KIRC and reveal a potential therapeutic target.
    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:在胰腺导管腺癌(PDAC)中,循环肿瘤细胞(CTC)的表征为癌症转移作为癌症相关死亡的主要原因提供了新的见解.这里,我们重点研究了视黄酸受体应答者1(RARRES1)在CTC上的表达,作为治疗失败和早期复发的新标志物.
    方法:细胞培养中氨基酸的稳定同位素标记(SILAC)方法用于鉴定和定量PDAC细胞系HPDE及其化学抗性对应物中的新生物标志物蛋白,L3.6pl-Res.55个基线和36个随访(FUP)外周血样品通过使用RARRES1作为另外的标记物的基于标记物的非依赖性微流体的CTC检测方法进行处理。
    结果:基于SILAC的蛋白质组学将RARRES1鉴定为在更具侵袭性的化学抗性PDAC细胞中大量表达的蛋白质。在基线,所有PDAC患者中检测到25.5%的CTC,而FUP分析(中位数:11个月FUP)显示45.5%的切除患者中检测到CTC.FUP时CTC阳性(≥3CTC)与无复发生存期短相关(p=0.002)。此外,检测到RARRES1阳性CTC提示术后复发更早(p=0.001).
    结论:在FUP期间切除的PDAC患者中检测到CTC与预后较差相关,和RARRES1表达可能鉴定出值得进一步研究的侵袭性CTC亚型。
    BACKGROUND: In pancreatic ductal adenocarcinoma (PDAC), the characterization of circulating tumor cells (CTCs) opens new insights into cancer metastasis as the leading cause of cancer-related death. Here, we focused on the expression of retinoic acid receptor responder 1 (RARRES1) on CTCs as a novel marker for treatment failure and early relapse.
    METHODS: The stable isotope labeling of amino acids in cell culture (SILAC)-approach was applied for identifying and quantifying new biomarker proteins in PDAC cell lines HPDE and its chemoresistant counterpart, L3.6pl-Res. Fifty-five baseline and 36 follow-up (FUP) peripheral blood samples were processed via a marker-independent microfluidic-based CTC detection approach using RARRES1 as an additional marker.
    RESULTS: SILAC-based proteomics identified RARRES1 as an abundantly expressed protein in more aggressive chemoresistant PDAC cells. At baseline, CTCs were detected in 25.5% of all PDAC patients, while FUP analysis (median: 11 months FUP) showed CTC detection in 45.5% of the resected patients. CTC positivity (≥3 CTC) at FUP was significantly associated with short recurrence-free survival (p = 0.002). Furthermore, detection of RARRES1 positive CTCs was indicative of an even earlier relapse after surgery (p = 0.001).
    CONCLUSIONS: CTC detection in resected PDAC patients during FUP is associated with a worse prognosis, and RARRES1 expression might identify an aggressive subtype of CTCs that deserves further investigation.
    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
    维甲酸受体反应者1(RARRES1)是多种癌症中最常见的甲基化位点之一。RARRES1调节线粒体和脂肪酸代谢,干细胞分化,和永生化细胞系在体外的存活。这里,我们创建了组成型Rarres1敲除(Rarres1-/-)小鼠模型来研究RARRES1的体内功能。Rarres1-/-胚胎成纤维细胞调节微管蛋白谷氨酸化,细胞代谢,和生存,在永生化细胞系中重述RARRES1功能。在两种小鼠品系中,Rarres1缺失导致滤泡性淋巴瘤(FL)的剂量依赖性发病率显著增加.在淋巴瘤形成之前,Rarres1-/-B细胞激活受损,成熟,分化为分泌抗体的浆细胞,和细胞周期进程。Rarres1消融增加了B细胞的存活率,并导致未折叠蛋白反应(UPR)和热休克反应(HSR)的激活。Rarres1缺乏对细胞代谢有不同的影响,随着成纤维细胞生物能量能力的增加,对B细胞的生物能学和代谢影响较小。这些发现表明,RARRES1在体内是一种真正的肿瘤抑制因子,在小鼠体内的缺失促进细胞存活,并减少B细胞分化,具有B细胞自主和非自主功能。
    Retinoic acid receptor responder 1 (RARRES1) is among the most commonly methylated loci in multiple cancers. RARRES1 regulates mitochondrial and fatty acid metabolism, stem cell differentiation, and survival of immortalized cell lines in vitro. Here, we created constitutive Rarres1 knockout (Rarres1 -/-) mouse models to study RARRES1 function in vivo. Rarres1 -/- embryonic fibroblasts regulated tubulin glutamylation, cell metabolism, and survival, recapitulating RARRES1 function in immortalized cell lines. In two mouse strains, loss of Rarres1 led to a markedly increased dose-dependent incidence of follicular lymphoma (FL). Prior to lymphoma formation, Rarres1 -/- B cells have compromised activation, maturation, differentiation into antibody-secreting plasma cells, and cell cycle progression. Rarres1 ablation increased B cell survival and led to activation of the unfolded protein response (UPR) and heat shock response (HSR). Rarres1 deficiency had differential effects on cellular metabolism, with increased bioenergetic capacity in fibroblasts, and minor effects on bioenergetics and metabolism in B cells. These findings reveal that RARRES1 is a bona fide tumor suppressor in vivo and the deletion in mice promotes cell survival, and reduces B cell differentiation with B cell autonomous and non-autonomous functions.
    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
    在大多数肾小球疾病中,炎症途径被激活,但在肾脏组织中驱动炎症途径的分子机制尚不清楚。我们确定视黄酸受体反应者1(Rarres1)是健康肾脏中高度富含足细胞的蛋白质。在足细胞特异性敲除动物中的研究表明,在肾小球肾炎模型中,Rarres1对于肾小球滤过屏障的正常发育或维持是不需要的,并且不调节肾脏疾病的结果。有趣的是,我们在IgA和糖尿病肾病的肾小球和肾小管周围毛细血管内皮细胞中检测到Rarres1表达的诱导,以及ANCA相关血管炎。对可公开获得的RNA数据集的分析表明,诱导Rarres1表达是慢性肾脏疾病中的常见分子机制。有条件的敲入鼠标行,在内皮细胞中特异性过表达Rarres1,未显示任何明显的肾脏表型。然而,这种过表达促进了肾小球肾炎模型中肾脏损害的进展。与此相符,条件性敲除小鼠,内皮细胞中缺乏Rarres1,在疾病模型中受到部分保护。机械上,Rarres1通过激活受体酪氨酸激酶Axl,通过转录因子核因子κB信号通路促进炎症和纤维化。因此,在内皮细胞中诱导Rarres1表达是人类肾小球疾病中普遍的分子机制,这似乎在通过核因子-κB信号通路驱动炎症和纤维化中具有致病作用。
    Inflammatory pathways are activated in most glomerular diseases but molecular mechanisms driving them in kidney tissue are poorly known. We identified retinoic acid receptor responder 1 (Rarres1) as a highly podocyte-enriched protein in healthy kidneys. Studies in podocyte-specific knockout animals indicated that Rarres1 was not needed for the normal development or maintenance of the glomerulus filtration barrier and did not modulate the outcome of kidney disease in a model of glomerulonephritis. Interestingly, we detected an induction of Rarres1 expression in glomerular and peritubular capillary endothelial cells in IgA and diabetic kidney disease, as well as in ANCA-associated vasculitis. Analysis of publicly available RNA data sets showed that the induction of Rarres1 expression was a common molecular mechanism in chronic kidney diseases. A conditional knock-in mouse line, overexpressing Rarres1 specifically in endothelial cells, did not show any obvious kidney phenotype. However, the overexpression promoted the progression of kidney damage in a model of glomerulonephritis. In line with this, conditional knock-out mice, lacking Rarres1 in endothelial cells, were partially protected in the disease model. Mechanistically, Rarres1 promoted inflammation and fibrosis via transcription factor Nuclear Factor-κB signaling pathway by activating receptor tyrosine kinase Axl. Thus, induction of Rarres1 expression in endothelial cells is a prevalent molecular mechanism in human glomerulopathies and this seems to have a pathogenic role in driving inflammation and fibrosis via the Nuclear Factor-κB signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • DOI:
    文章类型: Journal Article
    Immunotherapy is a promising route for the treatment of glioblastoma (GBM). Researchers have conducted a large number of studies on the pathogenesis of GBM; however, these studies are not comprehensive. High-throughput sequence analysis allows for insights into the pathogenesis of GBM. In this study, we used The Cancer Genome Atlas dataset to identify the function of RARRES1 enriched in GBM, especially in the WHO grade-IV cases. We discovered that RARRES1 is highly expressed in patients with mesenchymal subtype, unmethylated MGMT, IDH1 wild type, and non-G-CIMP, all of which are molecular characteristics of malignant GBM. Results of the immune microenvironment analysis showed that RARRES1 is strongly correlated with dendritic cells PD1, PDL2, TIM3, and CTLA4, which are the immune checkpoints in GBM. Furthermore, according to the overall survival and status analysis, a high expression of RARRES1 was found to be an unfavorable factor for prognosis. This indicates that RARRES1 may participate in the pathogenesis and immune-related processes in GBM, and may serve as a therapeutic target.
    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
    RARRES1, a retinoic acid regulated carboxypeptidase inhibitor associated with fatty acid metabolism, stem cell differentiation and tumorigenesis is among the most commonly methylated loci in multiple cancers but has no known mechanism of action. Here we show that RARRES1 interaction with cytoplasmic carboxypeptidase 2 (CCP2) inhibits tubulin deglutamylation, which in turn regulates the mitochondrial voltage dependent anion channel (VDAC1), mitochondrial membrane potential, AMPK activation, energy balance and metabolically reprograms cells and zebrafish to a more energetic and anabolic phenotype. Depletion of RARRES1 also increases expression of stem cell markers, promotes anoikis, anchorage independent growth and insensitivity to multiple apoptotic stimuli. As depletion of CCP2 or inhibition of VDAC1 reverses the effects of RARRES1 depletion on energy balance and cell survival we conclude that RARRES1 modulation of CCP2-modulated tubulin-mitochondrial VDAC1 interactions is a fundamental regulator of cancer and stem cell metabolism and survival.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号