ANO1

ano1
  • 文章类型: Journal Article
    潘氏细胞在肠道先天免疫反应中起着核心作用。这些细胞位于Lieberkuhn的小肠隐窝的底部。钙激活的氯离子通道TMEM16A和磷脂加扰酶TMEM16F控制细胞内Ca2+信号传导和胞吐作用。我们分析了TMEM16A和TMEM16F对Paneth细胞分泌的作用。
    产生具有Tmem16a(Tmem16a-/-)和Tmem16f(Tmem16f-/-)的肠上皮敲除的小鼠。分析了组织结构和Paneth细胞,并在体外小肠类器官中检查了Paneth细胞的胞吐作用。测量细胞内Ca2+信号并在野生型和Tmem16敲除小鼠之间进行比较。分析细菌定植和肠道凋亡。
    来自Tmem16a-/-和Tmem16f-/-小鼠的Lieberkuhn隐窝中的Paneth细胞表现出溶菌酶的积累。Tmem16a和Tmem16f位于野生型Paneth细胞中,但在来自敲除动物的细胞中不存在。在敲除动物的肠杯状细胞中,隐窝基部和粘液积累的潘氏细胞数量和大小增加。在线检查胆碱能和嘌呤能刺激下的颗粒融合和胞吐作用。两者在不存在Tmem16a或Tmem16f的情况下都受到强烈损害,并且也被Tmem16a/f的抑制所阻断。嘌呤能Ca2+信号在Tmem16a敲除小鼠中被大量抑制。敲除小鼠空肠细菌含量增加,而细胞凋亡被抑制。
    本数据证明了Tmem16在Paneth细胞中的胞吐作用。Tmem16a/f的抑制或激活可能影响小肠中存在的微生物含量和免疫功能。
    UNASSIGNED: Paneth cells play a central role in intestinal innate immune response. These cells are localized at the base of small intestinal crypts of Lieberkuhn. The calcium-activated chloride channel TMEM16A and the phospholipid scramblase TMEM16F control intracellular Ca2+ signaling and exocytosis. We analyzed the role of TMEM16A and TMEM16F for Paneth cells secretion.
    UNASSIGNED: Mice with intestinal epithelial knockout of Tmem16a (Tmem16a-/-) and Tmem16f (Tmem16f-/-) were generated. Tissue structures and Paneth cells were analyzed, and Paneth cell exocytosis was examined in small intestinal organoids in vitro. Intracellular Ca2+ signals were measured and were compared between wild-type and Tmem16 knockout mice. Bacterial colonization and intestinal apoptosis were analyzed.
    UNASSIGNED: Paneth cells in the crypts of Lieberkuhn from Tmem16a-/- and Tmem16f-/- mice demonstrated accumulation of lysozyme. Tmem16a and Tmem16f were localized in wild-type Paneth cells but were absent in cells from knockout animals. Paneth cell number and size were enhanced in the crypt base and mucus accumulated in intestinal goblet cells of knockout animals. Granule fusion and exocytosis on cholinergic and purinergic stimulation were examined online. Both were strongly compromised in the absence of Tmem16a or Tmem16f and were also blocked by inhibition of Tmem16a/f. Purinergic Ca2+ signaling was largely inhibited in Tmem16a knockout mice. Jejunal bacterial content was enhanced in knockout mice, whereas cellular apoptosis was inhibited.
    UNASSIGNED: The present data demonstrate the role of Tmem16 for exocytosis in Paneth cells. Inhibition or activation of Tmem16a/f is likely to affect microbial content and immune functions present in the small intestine.
    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
    尽管越来越多的证据表明钙激活的氯离子通道anocamin1(ANO1)在癌症转移中,其对前列腺癌转移潜能的直接影响以及表观遗传改变在这一过程中的可能意义尚不完全清楚.这里,我们显示ANO1在LNCap和DU145前列腺癌细胞系中最低表达,具有低转移潜能,但在高转移PC3前列腺癌细胞系中过表达。用DNMT抑制剂5-氮杂-2'-脱氧胞苷(5-Aza-CdR)处理LNCap和DU145细胞可增强ANO1表达,提示DNA甲基化是控制ANO1表达的机制之一。与这个概念一致,在LNCap和DU145细胞中,在ANO1启动子区的CpG岛检测到高甲基化,和5-Aza-CdR处理导致启动子CpG甲基化位点的剧烈去甲基化。在5-Aza-CdR处理后,转移指数,如细胞运动性,入侵,和转移相关基因表达,在LNCap和DU145细胞中显著改变。这些5-Aza-CdR诱导的转移标志是,然而,几乎完全被稳定的ANO1击倒。我们的体内观察进一步支持了这些体外发现,即异种移植小鼠模型中的ANO1表达增强了前列腺癌细胞向胫骨骨中的转移性播散和溶骨性病变的发展。总的来说,我们的结果有助于阐明ANO1表达在前列腺癌骨转移中的关键作用,其通过启动子CpG甲基化进行表观遗传调节。
    Despite growing evidence implicating the calcium-activated chloride channel anoctamin1 (ANO1) in cancer metastasis, its direct impact on the metastatic potential of prostate cancer and the possible significance of epigenetic alteration in this process are not fully understood. Here, we show that ANO1 is minimally expressed in LNCap and DU145 prostate cancer cell lines with low metastatic potential but overexpressed in high metastatic PC3 prostate cancer cell line. The treatment of LNCap and DU145 cells with DNMT inhibitor 5-aza-2\'-deoxycytidine (5-Aza-CdR) potentiates ANO1 expression, suggesting that DNA methylation is one of the mechanisms controlling ANO1 expression. Consistent with this notion, hypermethylation was detected at the CpG island of ANO1 promoter region in LNCap and DU145 cells, and 5-Aza-CdR treatment resulted in a drastic demethylation at promoter CpG methylation sites. Upon 5-Aza-CdR treatment, metastatic indexes, such as cell motility, invasion, and metastasis-related gene expression, were significantly altered in LNCap and DU145 cells. These 5-Aza-CdR-induced metastatic hallmarks were, however, almost completely ablated by stable knockdown of ANO1. These in vitro discoveries were further supported by our in vivo observation that ANO1 expression in xenograft mouse models enhances the metastatic dissemination of prostate cancer cells into tibial bone and the development of osteolytic lesions. Collectively, our results help elucidate the critical role of ANO1 expression in prostate cancer bone metastases, which is epigenetically modulated by promoter CpG methylation.
    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
    肝病包括所有类型的病毒性肝炎,酒精性肝病(ALD),非酒精性脂肪性肝病(NAFLD),肝硬化,肝功能衰竭(LF)和肝细胞癌(HCC)。肝病现在是全球疾病和死亡的主要原因之一,这迫使我们更好地了解肝脏疾病发展的机制。阳极素1(ANO1),钙激活的氯化物通道(CaCC),在上皮细胞分泌中起重要作用,扩散和迁移。ANO1在转录调控以及许多信号通路中起关键作用。它参与了起源,发展,一些肿瘤和其他疾病包括肝脏疾病的进展和/或转移。本文就ANO1在多种肝脏疾病发生发展中的作用及分子机制作一综述。旨在为进一步研究ANO1在肝脏疾病中的作用提供参考,为通过调节ANO1改善肝脏疾病的治疗策略做出贡献。
    Liver diseases include all types of viral hepatitis, alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), cirrhosis, liver failure (LF) and hepatocellular carcinoma (HCC). Liver disease is now one of the leading causes of disease and death worldwide, which compels us to better understand the mechanisms involved in the development of liver diseases. Anoctamin 1 (ANO1), a calcium-activated chloride channel (CaCC), plays an important role in epithelial cell secretion, proliferation and migration. ANO1 plays a key role in transcriptional regulation as well as in many signalling pathways. It is involved in the genesis, development, progression and/or metastasis of several tumours and other diseases including liver diseases. This paper reviews the role and molecular mechanisms of ANO1 in the development of various liver diseases, aiming to provide a reference for further research on the role of ANO1 in liver diseases and to contribute to the improvement of therapeutic strategies for liver diseases by regulating ANO1.
    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
    附睾导管表现出自发性阶段性收缩(SPCs)以储存和运输精子。这里,我们探索了驱动尾附睾导管中SPCs的起搏器细胞的分子鉴定,并研究了SPCs基础上的起搏器电流的特性,重点是ANO1Ca2激活的Cl-通道(CaCCs)。进行免疫组织化学以可视化大鼠尾附睾导管中血小板衍生的生长因子受体α(PDGFRα)或ANO1阳性细胞的分布。穿孔全细胞膜片钳技术应用于酶分离的附睾细胞,而SPC是用视频边缘跟踪技术记录的。免疫组织化学显示共表达PDGFRα和ANO1的α-平滑肌肌动蛋白(α-SMA)阳性细胞在最内层的平滑肌层中的分布。大约三分之一的分离的附睾细胞在-60mV的保持电位下表现出自发的瞬态内向电流(STIC)。根据细胞内Cl-浓度,STIC的逆转潜力接近计算的氯化物当量潜力。Ani9(3µM),ANO1特异性抑制剂,降低了STIC的振幅和频率,而环吡唑酸(CPA,30µM),一种肌细胞/内质网Ca2+-ATP酶(SERCA)抑制剂,废除了STIC。Ani9(3或10µM)降低了SPCs的频率,而不改变其幅度。因此,PDGFRα+,ANO1特化平滑肌细胞(SMC)似乎充当起搏器细胞,通过产生ANO1依赖性STIC来电驱动附睾SPC。从细胞内Ca2存储中自发释放Ca2并随后打开ANO1引起的STIC会导致去极化,并扩散到相邻的SMC中,其中L型电压依赖性Ca2通道被激活以发展SPC。
    The epididymal duct exhibits spontaneous phasic contractions (SPCs) to store and transport sperm. Here, we explored molecular identification of pacemaker cells driving SPCs in the caudal epididymal duct and also investigated properties of pacemaker currents underlying SPCs focusing on ANO1 Ca2+-activated Cl- channels (CaCCs). Immunohistochemistry was performed to visualise the distribution of platelet-derived growth factor receptor α (PDGFRα)- or ANO1-positive cells in the rat caudal epididymal duct. Perforated whole-cell patch clamp technique was applied to enzymatically isolated epididymal cells, while SPCs were recorded with video edge-tracking technique. Immunohistochemistry revealed the distribution of α-smooth muscle actin (α-SMA)-positive cells co-expressing both PDGFRα and ANO1 in the innermost smooth muscle layer. Approximately one-third of isolated epididymis cells exhibited spontaneous transient inward currents (STICs) at the holding potential -60 mV. The reversal potential for STICs was close to the calculated chloride equivalent potential depending on intracellular Cl- concentrations. Ani9 (3 µM), the ANO1 specific inhibitor, decreased both amplitude and frequency of STICs, while cyclopiazonic acid (CPA, 30 µM), a sarco-/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor, abolished STICs. Ani9 (3 or 10 µM) reduced the frequency of SPCs without changing their amplitude. Thus, PDGFRα+, ANO1+ specialised smooth muscle cells (SMCs) appear to function as pacemaker cells to electrically drive epididymal SPCs by generating ANO1-dependnet STICs. STICs arising from spontaneous Ca2+ release from intracellular Ca2+ store and subsequent opening of ANO1 result in depolarisations that spread into adjacent SMCs where L-type voltage-dependent Ca2+ channels are activated to develop SPCs.
    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
    瞬时受体电位香草酸4(TRPV4)是一种广泛表达的阳离子通道,在许多生理和病理过程中起重要作用。然而,大多数TRPV4药物都有副作用的风险。此外,现有的筛选方法不适用于药物的高通量筛选(HTS)。在这项研究中,基于钙激活的氯通道蛋白1Anoctamin1(ANO1)和黄色荧光蛋白(YFP)的双突变体(YFP-H148Q/I152L),建立了靶向TRPV4通道药物的细胞模型和HTS方法。采用膜片钳实验和荧光猝灭动力学实验验证了该模型能灵敏检测细胞内Ca2+浓度的变化。通过温度变化和不同浓度的TRPV4调节剂检查TRPV4细胞模型的功能。并对模型在HTS中的性能进行了评价。该模型能够敏感地检测细胞内Ca2+浓度的变化,并且在筛选TRPV4药物方面也很出色,该模型更适用于高温超导。我们成功构建了靶向TRPV4通道的药物细胞筛选模型,这为体外研究TRPV4的病理生理功能提供了工具。
    Transient receptor potential vanilloid 4 (TRPV4) is a widely expressed cation channel that plays an important role in many physiological and pathological processes. However, most TRPV4 drugs carry a risk of side effects. Moreover, existing screening methods are not suitable for the high-throughput screening (HTS) of drugs. In this study, a cell model and HTS method for targeting TRPV4 channel drugs were established based on a calcium-activated chloride channel protein 1 Anoctamin 1 (ANO1) and a double mutant (YFP-H148Q/I152L) of the yellow fluorescent protein (YFP). Patch-clamp experiments and fluorescence quenching kinetic experiments were used to verify that the model could sensitively detect changes in intracellular Ca2+ concentration. The functionality of the TRPV4 cell model was examined through temperature variations and different concentrations of TRPV4 modulators, and the performance of the model in HTS was also evaluated. The model was able to sensitively detect changes in the intracellular Ca2+ concentration and also excelled at screening TRPV4 drugs, and the model was more suitable for HTS. We successfully constructed a drug cell screening model targeting the TRPV4 channel, which provides a tool to study the pathophysiological functions of TRPV4 in vitro.
    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
    增强子-基因通讯取决于拓扑关联域(TAD)和由CCCTC结合因子(CTCF)绝缘体强制执行的边界,但是潜在的结构和机制仍然存在争议。这里,我们研究了在胃肠道间质瘤(GIST)中通常隔离成纤维细胞生长因子(FGF)癌基因但被DNA甲基化破坏的边界.边界包含一组CTCF站点,这些站点强制相邻的TAD,一个含有FGF基因,另一个含有ANO1及其推定的增强子,在GIST及其可能的起源细胞中特别活跃。我们表明,边界中四个CTCF基序的坐标破坏会融合相邻的TAD,允许ANO1增强子接触FGF3,并引起其强大的诱导。高分辨率micro-C图谱揭示了ANO1增强子和FGF3启动子中转录起始位点之间的特定接触,该启动子与FGF3诱导定量缩放,从而使接触频率的适度变化导致表达的强烈变化。符合因果关系。
    Enhancer-gene communication is dependent on topologically associating domains (TADs) and boundaries enforced by the CCCTC-binding factor (CTCF) insulator, but the underlying structures and mechanisms remain controversial. Here, we investigate a boundary that typically insulates fibroblast growth factor (FGF) oncogenes but is disrupted by DNA hypermethylation in gastrointestinal stromal tumors (GISTs). The boundary contains an array of CTCF sites that enforce adjacent TADs, one containing FGF genes and the other containing ANO1 and its putative enhancers, which are specifically active in GIST and its likely cell of origin. We show that coordinate disruption of four CTCF motifs in the boundary fuses the adjacent TADs, allows the ANO1 enhancer to contact FGF3, and causes its robust induction. High-resolution micro-C maps reveal specific contact between transcription initiation sites in the ANO1 enhancer and FGF3 promoter that quantitatively scales with FGF3 induction such that modest changes in contact frequency result in strong changes in expression, consistent with a causal relationship.
    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
    输卵管平滑肌表现出自发的节律性收缩(SRC),并在确切的时间控制卵子的通过,但其机械调控仍有待确定。在这项研究中,患有Ano1SMKO(Ano1的平滑肌特异性缺失)的雌性小鼠的生育力降低。小鼠中Ano1的缺乏导致输卵管中单个平滑肌细胞的输卵管SRC功能受损和钙信号传导降低。Ano1拮抗剂T16Ainh-A01剂量依赖性地抑制人和小鼠输卵管中的SRCs和[Ca2]i。硝苯地平治疗后,观察到SRCs和[Ca2]i的抑制作用相似。在我们的研究中,ANO1主要在[Ca2]i和输卵管平滑肌细胞的收缩中起激活剂或放大器的作用。我们发现异位妊娠患者的输卵管SRC明显减弱。然后,我们的研究旨在确定氯通道Ano1介导的平滑肌运动是否与输卵管SRC相关.我们的发现揭示了调节输卵管运动的新机制,该机制可能与异位妊娠等异常妊娠有关。
    Oviductal smooth muscle exhibits spontaneous rhythmic contraction (SRC) and controls the passage of the ova at the exact time, but its mechanistic regulation remains to be determined. In this study, female mice with Ano1SMKO (smooth muscle-specific deletion of Ano1) had reduced fertility. Deficiency of Ano1 in mice resulted in impaired oviductal SRC function and reduced calcium signaling in individual smooth muscle cells in the oviduct. The Ano1 antagonist T16Ainh-A01 dose-dependently inhibited SRCs and [Ca2+]i in the oviducts of humans and mice. A similar inhibitory effect of SRCs and [Ca2+]i was observed after treatment with nifedipine. In our study, ANO1 acted primarily as an activator or amplifier in [Ca2+]i and contraction of tubal smooth muscle cells. We found that tubal SRC was markedly attenuated in patients with ectopic pregnancy. Then, our study was designed to determine whether chloride channel Ano1-mediated smooth muscle motility is associated with tubal SRC. Our findings reveal a new mechanism for the regulation of tubal motility that may be associated with abnormal pregnancies such as ectopic pregnancies.
    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
    背景:本研究旨在探讨环状RNA(circircRNA)基质金属肽酶1(circleMMP1)在食管鳞状细胞癌(ESCC)进展中的功能和机制。
    方法:通过定量实时PCR(qRT-PCR)检测CircMMP1的表达,通过Kaplan-Meier分析评价其与ESCC患者预后的关系。使用相应的质粒转染细胞,和细胞增殖活性,评估了体外迁移和侵袭能力。使用蛋白质印迹分析组织和细胞中的蛋白质水平。RNA下拉,在ESCC细胞中进行双荧光素酶报告基因测定和RNA免疫沉淀测定,以检测circMMP1和miR-671-5p之间的相互作用,或miR-671-5p与ANO1之间的相关性。进行异种移植肿瘤实验以揭示circMMP1在体内的功能。
    结果:肿瘤组织中circMMP1的高水平与ESCC患者的不良预后相关。circMMP1敲除抑制ESCC细胞增殖,体外迁移和侵袭。MiR-671-5p是circMMP1的靶标,并介导circMMP1对ESCC细胞的抑制作用。CircMMP1靶向miR-671-5p调节ANO1表达,它位于miR-671-5p的下游。ANO1的过表达削弱了ESCC细胞中circMMP1敲低的肿瘤抑制功能。此外,circMMP1的沉默阻碍了体内ESCC肿瘤的生长。
    结论:我们的研究提供了新的证据,表明circMMP1通过作为miR-671-5p海绵增强ANO1表达来加速ESCC进展。
    The aim of this study was to explore the function and mechanism of circular RNA (circRNA) matrix metallopeptidase 1 (circMMP1) in the progression of esophageal squamous cell carcinoma (ESCC).
    CircMMP1 expression was detected by quantitative real-time PCR (qRT-PCR), and its relationship with the prognosis of ESCC patients was evaluated by Kaplan-Meier analysis. Cells were transfected using corresponding plasmids, and the cell proliferation activity, migration and invasion capabilities in vitro were assessed. The protein level in tissues and cells was analyzed using western blotting. RNA pulldown, dual-luciferase reporter assay and RNA immunoprecipitation assay were performed in ESCC cells to detect the interaction between circMMP1 and miR-671-5p, or the correlation between miR-671-5p and ANO1. Xenograft tumor experiment was carried out to uncover the function of circMMP1 in vivo.
    The high level of circMMP1 in tumor tissues was associated with poor prognoses of ESCC patients. Knockdown of circMMP1 suppressed ESCC cell proliferation, migration and invasion in vitro. MiR-671-5p was the target of circMMP1 and mediated the inhibition effect of circMMP1 on ESCC cells. CircMMP1 targeted miR-671-5p to regulate ANO1 expression, which was downstream of miR-671-5p. Overexpression of ANO1 weakened tumor-repressive function of circMMP1 knockdown in ESCC cells. Moreover, silencing of circMMP1 impeded ESCC tumor growth in vivo.
    Our study provided novel evidence that circMMP1 accelerated ESCC progression by acting as a miR-671-5p sponge to enhance ANO1 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    TMEM16A钙激活的氯离子通道是各种疾病的有希望的治疗靶标。氯硝柳胺,一种驱虫药,已被认为是治疗哮喘和慢性阻塞性肺疾病的TMEM16A抑制剂,但最近发现具有广谱脱靶效应。在这里我们展示,在生理条件下,氯硝柳胺可急剧增强TMEM16A,而不会产生任何抑制作用。我们的计算和功能表征在TMEM16A的胞外侧确定了推定的氯硝柳胺结合位点。该位点中的突变减弱了增强作用。此外,氯硝柳胺增强血管平滑肌细胞中的内源性TMEM16A,引发细胞内钙增加,并收缩小鼠肠系膜动脉.我们的研究结果建议谨慎考虑氯硝柳胺作为TMEM16A抑制剂治疗哮喘等疾病时,COPD,和高血压。推定的氯硝柳胺结合位点的鉴定提供了对TMEM16A药理调节机制的见解,为开发特定的TMEM16A调节剂以治疗人类疾病而发光。
    The TMEM16A calcium-activated chloride channel is a promising therapeutic target for various diseases. Niclosamide, an anthelmintic medication, has been considered as a TMEM16A inhibitor for treating asthma and chronic obstructive pulmonary disease, but was recently found to possess broad-spectrum off-target effects. Here we show that, under physiological conditions, niclosamide acutely potentiates TMEM16A without having any inhibitory effect. Our computational and functional characterizations pinpoint a putative niclosamide binding site on the extracellular side of TMEM16A. Mutations in this site attenuate the potentiation. Moreover, niclosamide potentiates endogenous TMEM16A in vascular smooth muscle cells, triggers intracellular calcium increase, and constricts the murine mesenteric artery. Our findings advise caution when considering niclosamide as a TMEM16A inhibitor to treat diseases such as asthma, COPD, and hypertension. The identification of the putative niclosamide binding site provides insights into the mechanism of TMEM16A pharmacological modulation, shining light on developing specific TMEM16A modulators to treat human 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
    免疫疗法在胃肠(GI)癌症中的应用由于有限的应答率和新出现的治疗抗性而仍然具有挑战性。结合临床队列,多组学研究,和功能/分子实验,发现ANO1扩增或高表达预示着胃肠道肿瘤患者对免疫治疗的不良结局和耐药性.敲低或抑制ANO1抑制多种胃肠道癌细胞系的生长/转移/侵袭,细胞来源的异种移植物,和患者来源的异种移植模型。ANO1有助于免疫抑制肿瘤微环境,并诱导抗PD-1免疫疗法的获得性耐药,而ANO1敲低或抑制可增强免疫治疗效果并克服对免疫治疗的耐药性。机械上,通过以PI3K-Akt信号依赖的方式抑制癌症铁蛋白死亡,ANO1通过促进TGF-β释放促进肿瘤进展并促进癌症相关成纤维细胞募集,从而削弱CD8+T细胞介导的抗肿瘤免疫,并产生对免疫疗法的抵抗。这项工作强调了ANO1在介导肿瘤免疫微环境重塑和免疫治疗抵抗中的作用。并介绍了ANO1作为胃肠道癌症精准治疗的有希望的靶标。
    The application of immunotherapy in gastrointestinal (GI) cancers remains challenging because of the limited response rate and emerging therapeutic resistance. Combining clinical cohorts, multi-omics study, and functional/molecular experiments, it is found that ANO1 amplification or high-expression predicts poor outcomes and resistance to immunotherapy for GI cancer patients. Knocking-down or inhibiting ANO1 suppresses the growth/metastasis/invasion of multiple GI cancer cell lines, cell-derived xenograft, and patient-derived xenograft models. ANO1 contributes to an immune-suppressive tumor microenvironment and induces acquired resistance to anti-PD-1 immunotherapy, while ANO1 knockdown or inhibition enhances immunotherapeutic effectiveness and overcomes resistance to immunotherapy. Mechanistically, through inhibiting cancer ferroptosis in a PI3K-Akt signaling-dependent manner, ANO1 enhances tumor progression and facilitates cancer-associated fibroblast recruitment by promoting TGF-β release, thus crippling CD8+ T cell-mediated anti-tumor immunity and generating resistance to immunotherapy. This work highlights ANO1\'s role in mediating tumor immune microenvironment remodeling and immunotherapeutic resistance, and introduces ANO1 as a promising target for GI cancers\' precision treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号