VIPR1

VIPR1
  • 文章类型: Journal Article
    背景:宣威肺癌(XWLC)以其高发病率和高死亡率而闻名。然而,分子机制尚不清楚。
    方法:我们进行了全面的转录组学,蛋白质组学,和来自三名患有肺腺癌(LUAD)的XWLC患者的肿瘤和匹配的正常邻近组织的磷酸化蛋白质组学表征。
    结果:综合转录组和蛋白质组分析揭示了肿瘤中失调的分子和途径,并确定了增强的代谢-疾病偶联。非编码RNA广泛参与转录后调控机制,以协调LUAD的进展,并部分解释了RNA和蛋白质表达模式之间的分子差异。磷酸化蛋白质组为新的磷酸盐位点提供了证据支持,报告核心激酶家族成员和参与代谢的关键激酶途径的潜在作用,豁免权,和稳态。此外,通过与以前的LUAD研究相比,我们强调宣威LUAD患者的氧化磷酸化程度较高,并指出VIPR1缺乏会加重代谢功能障碍。
    结论:我们的综合多组学分析为系统了解XWLC的分子结构提供了强大的资源,并提出了基于氧化还原代谢的治疗机会。
    Xuanwei lung cancer (XWLC) is well-known for its high incidence and mortality. However, the molecular mechanism is still unclear.
    We performed a comprehensive transcriptomic, proteomic, and phosphoproteomic characterization of tumors and matched normal adjacent tissues from three XWLC patients with lung adenocarcinoma (LUAD).
    Integrated transcriptome and proteome analysis revealed dysregulated molecules and pathways in tumors and identified enhanced metabolic-disease coupling. Non-coding RNAs were widely involved in post-transcriptional regulatory mechanisms to coordinate the progress of LUAD and partially explained the molecular differences between RNA and protein expression patterns. Phosphoproteome provided evidence support for new phosphate sites, reporting the potential roles of core kinase family members and key kinase pathways involved in metabolism, immunity, and homeostasis. In addition, by comparing with the previous LUAD researches, we emphasized the higher degree of oxidative phosphorylation in Xuanwei LUAD and pointed that VIPR1 deficiency aggravated metabolic dysfunction.
    Our integrated multi-omics analysis provided a powerful resource for a systematic understanding of the molecular structure of XWLC and proposed therapeutic opportunities based on redox metabolism.
    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
    Williams-Beuren综合征(WBS)是由~27个连续基因的半合子微缺失引起的一种罕见疾病。尽管存在神经发育和认知缺陷,患有WBS的人幸免或增强了音乐和听觉能力,可能提供对听觉感知的遗传基础的洞察。这里,我们报道,WBS小鼠模型在听觉皮层(ACx)具有天生增强的频率辨别敏锐度和改善的频率编码.化学遗传学挽救显示,频率辨别过强是由ACx中过度兴奋的中间神经元引起的。一个WBS基因的单倍体不足,Gtf2ird1,通过下调神经肽受体VIPR1复制WBS表型。VIPR1在患有WBS的个体的ACx中和在源自具有WBS微缺失的人诱导多能干细胞的脑类器官中降低。在ACx中间神经元中,Vipr1缺失或过表达被模仿或逆转,分别,WBS小鼠的细胞和行为表型。因此,ACx中间神经元中的Gtf2ird1-Vipr1机制可能是WBS中上听觉敏锐度的基础。
    Williams-Beuren syndrome (WBS) is a rare disorder caused by hemizygous microdeletion of ∼27 contiguous genes. Despite neurodevelopmental and cognitive deficits, individuals with WBS have spared or enhanced musical and auditory abilities, potentially offering an insight into the genetic basis of auditory perception. Here, we report that the mouse models of WBS have innately enhanced frequency-discrimination acuity and improved frequency coding in the auditory cortex (ACx). Chemogenetic rescue showed frequency-discrimination hyperacuity is caused by hyperexcitable interneurons in the ACx. Haploinsufficiency of one WBS gene, Gtf2ird1, replicated WBS phenotypes by downregulating the neuropeptide receptor VIPR1. VIPR1 is reduced in the ACx of individuals with WBS and in the cerebral organoids derived from human induced pluripotent stem cells with the WBS microdeletion. Vipr1 deletion or overexpression in ACx interneurons mimicked or reversed, respectively, the cellular and behavioral phenotypes of WBS mice. Thus, the Gtf2ird1-Vipr1 mechanism in ACx interneurons may underlie the superior auditory acuity in WBS.
    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
    肠神经系统(ENS)和肠上皮之间的相互作用被认为在肠道稳态中起着至关重要的作用。ENS如何在维持上皮功能的同时监测共生和病原微生物的前沿尚不清楚。这里,通过将膈下迷走神经切断术与转录组学相结合,化学遗传学策略,和肠神经元-肠类器官的共培养,我们显示表达VIP的肠神经元塑造肠上皮细胞(IECs)的α1,2-岩藻糖基化。机械上,神经肽VIP通过IECs上的VIPR1受体通过Erk1/2-c-Fos途径激活fut2表达。我们进一步证明,肠神经元的扰动在稳态下通过α1,2-岩藻糖基化导致肠道菌群失调,并导致对酒精相关肝病(ALD)的易感性增加。这归因于ALD中有益双歧杆菌和机会致病性粪肠球菌之间的不平衡。此外,双歧杆菌α1,2-岩藻糖苷酶可促进双歧杆菌粘附于粘膜表面,限制粪肠球菌过度生长并防止ALD进展。
    Interactions between the enteric nervous system (ENS) and intestinal epithelium are thought to play a vital role in intestinal homeostasis. How the ENS monitors the frontier with commensal and pathogenic microbes while maintaining epithelial function remains unclear. Here, by combining subdiaphragmatic vagotomy with transcriptomics, chemogenetic strategy, and coculture of enteric neuron-intestinal organoid, we show that enteric neurons expressing VIP shape the α1,2-fucosylation of intestinal epithelial cells (IECs). Mechanistically, neuropeptide VIP activates fut2 expression via the Erk1/2-c-Fos pathway through the VIPR1 receptor on IECs. We further demonstrate that perturbation of enteric neurons leads to gut dysbiosis through α1,2-fucosylation in the steady state and results in increased susceptibility to alcohol-associated liver disease (ALD). This was attributed to an imbalance between beneficial Bifidobacterium and opportunistic pathogenic Enterococcus faecalis in ALD. In addition, Bifidobacterium α1,2-fucosidase may promote Bifidobacterium adhesion to the mucosal surface, which restricts Enterococcus faecalis overgrowth and prevents ALD progression.
    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
    背景和目的:已经报道了血管活性肠多肽I型受体(VIPR1)在许多类型的恶性肿瘤中的过表达,并用于开发新的靶向疗法和基于放射性标记的VIP类似物的肿瘤成像技术。但其在肝癌发生中的作用尚未被探索。在目前的研究中,我们研究了VIP/VIPR1信号在控制肝细胞癌(HCC)进展中的作用。方法和结果:通过分析临床样本,我们发现VIPR1的表达水平在人肝癌组织中下调,这与晚期临床阶段相关,肿瘤生长,复发,和肝癌的临床不良结果。体外和体内研究表明,VIP激活VIPR1可显着抑制HCC的生长和转移。有趣的是,转录组测序分析表明,VIP对VIPR1的激活调节了精氨酸的生物合成。培养的肝癌细胞的力学研究表明,VIP治疗部分恢复精氨酸合成代谢关键酶精氨酸琥珀酸合酶(ASS1)的表达,在某种程度上,通过下调CAD(氨基甲酰磷酸合成酶2,天冬氨酸转碳淀粉酶,和二氢乳清酶)。VIP治疗上调ASS1并随后以mTOR/p70S6K信号依赖性方式抑制CAD磷酸化。临床上,我们发现人类肝癌样本与ASS1下调相关,但CAD磷酸化上调,VIPR1水平与ASS1水平和血清尿素水平呈正相关,肝癌中尿素循环和精氨酸代谢的最终产物。结论:HCC中VIPR1表达的缺失促进CAD磷酸化和肿瘤进展,VIPR1的恢复和VIPR1激动剂的治疗可能是肝癌治疗的一种有希望的方法。
    Background and aims: Vasoactive intestinal polypeptide type-I receptor (VIPR1) overexpression has been reported in numerous types of malignancies and utilized to develop novel target therapeutics and radiolabeled VIP analogue-based tumor imaging technology, but its role in liver carcinogenesis has not been explored. In the current study, we investigated the role of the VIP/VIPR1 signaling in controlling hepatocellular carcinoma (HCC) progression. Approach and results: By analyzing clinical samples, we found the expression level of VIPR1 was downregulated in human HCC tissues, which was correlated with advanced clinical stages, tumor growth, recurrence, and poor outcomes of HCC clinically. In vitro and in vivo studies revealed that activation of VIPR1 by VIP markedly inhibited HCC growth and metastasis. Intriguingly, transcriptome sequencing analyses revealed that activation of VIPR1 by VIP regulated arginine biosynthesis. Mechanistical studies in cultured HCC cells demonstrated that VIP treatment partially restored the expression of arginine anabolic key enzyme argininosuccinate synthase (ASS1), and to some extent, inhibited de novo pyrimidine synthetic pathway by downregulating the activation of CAD (carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase). VIP treatment upregulated ASS1 and subsequently suppressed CAD phosphorylation in an mTOR/p70S6K signaling dependent manner. Clinically, we found human HCC samples were associated with downregulation of ASS1 but upregulation of CAD phosphorylation, and that VIPR1 levels positively correlated with ASS1 levels and serum levels of urea, the end product of the urea cycle and arginine metabolism in HCC. Conclusions: Loss of VIPR1 expression in HCC facilitates CAD phosphorylation and tumor progression, and restoration of VIPR1 and treatment with the VIPR1 agonist may be a promising approach for HCC treatment.
    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
    The vasoactive intestinal peptide receptor-1(VIPR1) has prominent growth effects on a number of common neoplasms. However, there were contradictions in the effect cross different cancers. We aimed to explore the effect of VIPR1 overexpression on a human lung adenocarcinoma cell line H1299. GEO dataset was used to screen differentially expressed genes in lung adenocarcinoma tissues. The expression of VIPR1 mRNA was determined in the cancer Genome Atlas (TCGA). Immunohistochemical analysis was performed to determine VIPR1 protein expression in lung adenocarcinoma and corresponding adjacent tissues (n = 22). Fluorescence real-time quantitative PCR detected the expression of VIPR1 in human normal lung epithelial cell line BEAS-2B and lung adenocarcinoma cell line H1299. Overexpression strategies were employed to assess functions of VIPR1 expression on several malignant phenotypes in H1299. The expression of VIPR1 was lower in lung adenocarcinoma tissues than that in adjacent tissues. Compared with the normal lung epithelial cells BEAS-2B, VIPR1 was down-regulated in lung cancer cells H1299 (P < 0.05). After the overexpression of VIPR1, we found that VIPR1 significantly inhibited growth, migration, and invasion of H1299 cells (P < 0.05). Our findings point out the tumor suppressor roles of VIPR1 in human LUAD pathogenesis.
    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
    Vasoactive intestinal peptide receptor 1 (VPAC1) is a member of a secretin-like subfamily of G protein-coupled receptors. Its endogenous neuropeptide (VIP), secreted by neurons and immune cells, modulates various physiological functions such as exocrine and endocrine secretions, immune response, smooth muscles relaxation, vasodilation, and fetal development. As a drug target, VPAC1 has been selected for therapy of inflammatory diseases but drug discovery is still hampered by lack of its crystal structure. In this study we presented the homology model of this receptor constructed with the well-known web service GPCRM. The VPAC1 model is composed of extracellular and transmembrane domains that form a complex with an endogenous hormone VIP. Using the homology model of VPAC1 the mechanism of action of potential drug candidates for VPAC1 was described. Only two series of small-molecule antagonists of confirmed biological activity for VPAC1 have been described thus far. Molecular docking and a series of molecular dynamics simulations were performed to elucidate their binding to VPAC1 and resulting antagonist effect. The presented work provides the basis for the possible binding mode of VPAC1 antagonists and determinants of their molecular recognition in the context of other class B GPCRs. Until the crystal structure of VPAC1 will be released, the presented homology model of VPAC1 can serve as a scaffold for drug discovery studies and is available from the author upon request.
    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
    Vasoactive intestinal peptide receptor 1 (VIPR1) is observed to express differently in human malignancies. Here, we aim to reveal clinical significance and transcriptional regulation mechanism of VIPR1 in hepatocellular carcinoma (HCC). Using immunohistochemistry, pyrosequencing, quantitative real-time PCR (qPCR), decitabine (DAC)/4-phenylbutyricacid (PBA) treatment and chromatin immunoprecipitation (ChIP), we found the low expression of VIPR1 was correlated with poor histological differentiation and poor survival. The promoter region of VIPR1 was methylated and DNA methylation inhibited VIPR1 gene transcription. Deacetylation of H3K27 in the promoter of VIPR1 inhibited the transcription of VIPR1 in HCC. In conclusion, low expression of VIPR1 had an adverse prognostic impact on HCC, and such expression is at least partially mediated by epigenetic modification.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号