Phenotypic screen

表型筛选
  • 文章类型: Journal Article
    癌症仍然是全球死亡的主要原因,批准的肿瘤药物继续具有异质性的患者反应和伴随的不良反应(AE),限制了有效性。这里,我们使用转化的人类多能癌症干细胞(CSC)的替代模型,在干性的背景下检查了超过100种FDA批准的肿瘤药物能够区分异常自我更新和分化的健康干细胞(hSCs)。尽管这些药物中有一部分没有作用(无效),较大部分受影响的CSC(活性),和独特的子集优先影响CSC而不是hSC(选择性)。每种药物的FDA认可靶标的单细胞基因表达和蛋白质谱提供了CSC与CSC反应的分子相关性hSC。独特的,对CSCs具有选择性的药物表现出临床疗效,以总生存率衡量,并减少AE。我们的研究结果表明,虽然无意中,一半的抗癌药物对CSC具有活性,并与改善的临床结果相关.基于这些发现,我们建议将靶向CSC靶向的能力作为早期肿瘤治疗开发的一个特性.
    Cancer remains the leading cause of death worldwide with approved oncology drugs continuing to have heterogenous patient responses and accompanied adverse effects (AEs) that limits effectiveness. Here, we examined >100 FDA-approved oncology drugs in the context of stemness using a surrogate model of transformed human pluripotent cancer stem cells (CSCs) vs. healthy stem cells (hSCs) capable of distinguishing abnormal self-renewal and differentiation. Although a proportion of these drugs had no effects (inactive), a larger portion affected CSCs (active), and a unique subset preferentially affected CSCs over hSCs (selective). Single cell gene expression and protein profiling of each drug\'s FDA recognized target provided a molecular correlation of responses in CSCs vs. hSCs. Uniquely, drugs selective for CSCs demonstrated clinical efficacy, measured by overall survival, and reduced AEs. Our findings reveal that while unintentional, half of anticancer drugs are active against CSCs and associated with improved clinical outcomes. Based on these findings, we suggest ability to target CSC targeting should be included as a property of early onco-therapeutic development.
    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
    我们提出了一种新的小分子抗病毒化学型,通过非常规的无细胞蛋白质合成和基于组装的表型筛选鉴定,用于调节病毒衣壳组装。PAV-431,该系列的代表性化合物的活性,已在多种细胞培养模型中针对引起人类大多数呼吸道疾病的所有六个病毒家族的感染性病毒进行了验证。在动物中,这种化学型已被证明对猪流行性腹泻病毒(一种冠状病毒)和呼吸道合胞病毒(一种副粘病毒)有效。PAV-431显示与蛋白质14-3-3(一种已知的变构调节剂)结合。然而,它似乎只针对14-3-3的一小部分,该部分存在于动态多蛋白复合物中,该复合物的成分包括与病毒生命周期和先天免疫有关的蛋白质。这种靶多蛋白复合物的组成似乎在病毒感染后被修饰,并通过PAV-431治疗在很大程度上恢复。先进的模拟,PAV-104被证明对病毒修饰的靶标具有选择性,从而避免宿主毒性。我们的发现提出了一种新的理解范式,和下药,主机-病毒接口,从而为呼吸道病毒性疾病的治疗提供了一种新的临床治疗策略。
    We present a novel small molecule antiviral chemotype that was identified by an unconventional cell-free protein synthesis and assembly-based phenotypic screen for modulation of viral capsid assembly. Activity of PAV-431, a representative compound from the series, has been validated against infectious viruses in multiple cell culture models for all six families of viruses causing most respiratory diseases in humans. In animals, this chemotype has been demonstrated efficacious for porcine epidemic diarrhoea virus (a coronavirus) and respiratory syncytial virus (a paramyxovirus). PAV-431 is shown to bind to the protein 14-3-3, a known allosteric modulator. However, it only appears to target the small subset of 14-3-3 which is present in a dynamic multi-protein complex whose components include proteins implicated in viral life cycles and in innate immunity. The composition of this target multi-protein complex appears to be modified upon viral infection and largely restored by PAV-431 treatment. An advanced analog, PAV-104, is shown to be selective for the virally modified target, thereby avoiding host toxicity. Our findings suggest a new paradigm for understanding, and drugging, the host-virus interface, which leads to a new clinical therapeutic strategy for treatment of respiratory viral disease.
    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
    增殖细胞核抗原(PCNA)是一种同源三聚体蛋白复合物,在复制过程中会将DNA周围的DNA与模板束缚在一起,并充当许多其他相互作用蛋白的枢纽。它通过结合具有短线性基序(SLiMs)的蛋白质来调节DNA代谢过程和其他重要的细胞功能,例如PIP盒(PCNA相互作用蛋白盒)或APIM(AlkB同源物2PCNA相互作用基序)在SLiMs结合的疏水口袋中。然而,致病性原生锥虫中过量生产TbPCNA或人类PCNA(hPCNA)会引发增殖停滞的显性阴性表型。阻止布氏木霉增殖的机制需要过度产生的PCNA直向同源物具有功能完整的SLiM结合袋。定点突变研究表明,布鲁氏菌过度生产具有破坏的SLiM结合口袋的PCNA变体正常生长。我们假设化学破坏SLiM结合口袋会恢复布鲁切T.Brucei的增殖,过度生产PCNA直系同源物。测试该假设是基于布鲁氏T.brucei的PCNA筛选测定的概念证明。分析设计是为了发现生物活性小分子,恢复过度产生PCNA直向同源物的布鲁氏菌菌株的增殖,可能是通过破坏SLiM结合口袋中的相互作用。该测定的试验筛选发现了与预定PCNA靶标连接的两种命中化合物。化合物#1,一种已知的hPCNA抑制剂,对布鲁氏菌过量生产的hPCNA具有选择性生物活性,验证分析。化合物#6对hPCNA和TbPCNA具有混杂的生物活性,但是是第一个发现的具有抑制TbPCNA的生物活性的化合物。
    Proliferating cell nuclear antigen (PCNA) is a homo-trimeric protein complex that clamps around DNA to tether DNA polymerases to the template during replication and serves as a hub for many other interacting proteins. It regulates DNA metabolic processes and other vital cellar functions through the binding of proteins having short linear motifs (SLiMs) like the PIP-box (PCNA-interacting protein-box) or the APIM (AlkB homolog 2 PCNA-interacting motif) in the hydrophobic pocket where SLiMs bind. However, overproducing TbPCNA or human PCNA (hPCNA) in the pathogenic protist Trypanosoma brucei triggers a dominant-negative phenotype of arrested proliferation. The mechanism for arresting T. brucei proliferation requires the overproduced PCNA orthologs to have functional intact SLiM-binding pocket. Sight-directed mutagenesis studies showed that T. brucei overproducing PCNA variants with disrupted SLiM-binding pockets grew normally. We hypothesized that chemically disrupting the SLiM-binding pocket would restore proliferation in T. brucei, overproducing PCNA orthologs. Testing this hypothesis is the proof-of-concept for a T. brucei-based PCNA screening assay. The assay design is to discover bioactive small molecules that restore proliferation in T. brucei strains that overproduce PCNA orthologs, likely by disrupting interactions in the SLiM-binding pocket. The pilot screen for this assay discovered two hit compounds that linked to predetermined PCNA targets. Compound #1, a known hPCNA inhibitor, had selective bioactivity to hPCNA overproduced in T. brucei, validating the assay. Compound #6 had promiscuous bioactivity for hPCNA and TbPCNA but is the first compound discovered with bioactivity for inhibiting TbPCNA.
    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
    在药物发现过程的临床前阶段,在体外药物敏感性筛选后,细胞活力测定是研究细胞表型特性和总体健康的基本工具。因此,重要的是要优化您选择的活力测定,以获得可重复和可复制的结果,以及使用相关的药物反应指标(例如,IC50,AUC,GR50和GRmax)以确定候选药物,以进行体内进一步评估。在这里,我们使用了刃天青素减少测定法,这是一种快速的方法,成本效益高,简单易用,和敏感的方法来检查细胞的表型特性。使用MCF7乳腺癌细胞系,我们提供了一个详细的分步方案,用于使用刃天青测定法优化药物敏感性筛选.
    During the preclinical stages of the drug discovery process, cell viability assays are fundamental tools for studying the phenotypic properties and overall health of cells following in vitro drug sensitivity screens. Therefore, it is important to optimize your viability assay of choice to obtain reproducible and replicable results, as well as use relevant drug response metrics (e.g., IC50, AUC, GR50, and GRmax) to identify candidate drugs for further evaluation in vivo. Herein, we used the resazurin reduction assay which is a quick, cost-effective, simple-to-use, and sensitive method for examining the phenotypic properties of cells. Using the MCF7 breast cancer cell line, we provide a detailed step-by-step protocol for optimizing drug sensitivity screens using the resazurin assay.
    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
    只有三类当代抗真菌药物常规用于临床以对抗丝状真菌病原体如烟曲霉。高通量表型筛选以鉴定具有抗丝状真菌活性的小分子仍然具有挑战性,因为菌丝,这些重要生物的生物膜样生长形态。在这一章中,我们描述了利用生物发光的烟曲霉菌株鉴定增强三唑抗真菌药物氟康唑活性的小分子的方案。该测定法对于鉴定具有针对丝状真菌病原体的活性的小分子具有很大的希望。
    Only three classes of contemporary antifungal drugs are routinely utilized in the clinic against filamentous fungal pathogens such as Aspergillus fumigatus. High-throughput phenotypic screens to identify small molecules with activity against filamentous fungi remain challenging due to the hyphal, biofilm-like growth morphology of these important organisms. In this chapter, we describe a protocol for utilizing a bioluminescent A. fumigatus strain for identifying small molecules that potentiate the activity of the triazole antifungal drug fluconazole. The assay holds great promise for identifying small molecules with activity against filamentous fungal pathogens.
    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:我们提出了一种针对结直肠癌(CRC)患者设计个性化治疗的新方法,通过将体外类器官功效测试与结果的数学建模相结合。
    方法:经过验证的表型方法称为治疗指导多药优化(TGMO),用于在对一线CRC化疗(FOLFOXIRI)敏感或耐药的3D人类CRC细胞模型中鉴定四种低剂量协同优化药物组合(ODC)。我们的发现是使用二阶线性回归和自适应套索获得的。
    结果:所有ODC的活性均在原发性或转移性CRC患者来源的类器官(PDO)上得到验证。使用全外显子组测序和RNAseq对CRC材料进行分子表征。在鉴别为CMS4/CRIS-A的肝转移(IV期)患者的PDO中,我们的ODC由雷戈拉非尼[1mM]组成,vemurafenib[11mM],palbociclib[1mM]和拉帕替尼[0.5mM]抑制细胞活力达88%,显着优于临床剂量的FOLFOXIRI。此外,我们确定了基于患者特异性TGMO的ODC,其疗效优于当前化疗标准的治疗效果,FolfOXIRI.
    结论:我们的方法允许在临床相关的时间范围内优化患者定制的协同多药组合。
    BACKGROUND: We propose a new approach for designing personalized treatment for colorectal cancer (CRC) patients, by combining ex vivo organoid efficacy testing with mathematical modeling of the results.
    METHODS: The validated phenotypic approach called Therapeutically Guided Multidrug Optimization (TGMO) was used to identify four low-dose synergistic optimized drug combinations (ODC) in 3D human CRC models of cells that are either sensitive or resistant to first-line CRC chemotherapy (FOLFOXIRI). Our findings were obtained using second order linear regression and adaptive lasso.
    RESULTS: The activity of all ODCs was validated on patient-derived organoids (PDO) from cases with either primary or metastatic CRC. The CRC material was molecularly characterized using whole-exome sequencing and RNAseq. In PDO from patients with liver metastases (stage IV) identified as CMS4/CRIS-A, our ODCs consisting of regorafenib [1 mM], vemurafenib [11 mM], palbociclib [1 mM] and lapatinib [0.5 mM] inhibited cell viability up to 88%, which significantly outperforms FOLFOXIRI administered at clinical doses. Furthermore, we identified patient-specific TGMO-based ODCs that outperform the efficacy of the current chemotherapy standard of care, FOLFOXIRI.
    CONCLUSIONS: Our approach allows the optimization of patient-tailored synergistic multi-drug combinations within a clinically relevant timeframe.
    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
    Cullin-RING泛素连接酶(CRL)调节心脏中的许多生物过程,并与调节心脏肥大有关。本研究旨在鉴定心肌细胞(CM)中新型的肥大调节CRLs。采用使用siRNA介导的耗竭和自动显微镜的功能基因组方法来筛选新生大鼠CM中的细胞大小调节CRL。通过3H-异亮氨酸掺入确认筛选命中。在筛选的43个目标中,siRNA介导的Fbxo6,Fbxo45和Fbxl14的消耗导致细胞大小减小,而在基础条件下,Fbxo9,Fbxo25,Fbxo30,Fbxo32,Fbxo33,Cullin1,Roc1,Ddb1,Fbxw4和Fbxw5的消耗导致细胞大小显着增加。在用去氧肾上腺素(PE)刺激的CM中,Fbxo6,Fbxo25,Fbxo33,Fbxo45和Fbxw4的消耗进一步增加了PE诱导的肥大。作为一个概念证明,通过横向主动脉缩窄术(TAC)分析CRLFbox25,导致与对照动物相比,Fbxo25蛋白浓度增加4.5倍.在细胞培养中,siRNA介导的Fbxo25耗竭导致CM细胞大小增加约37%,3H-异亮氨酸掺入增加约41%。消耗Fbxo25导致Anp和Bnp上调。总之,我们鉴定出13个新的CRLs为CM肥大的正或负调节因子.其中,CRLFbox25进一步表征,作为心脏肥大的潜在调节剂。
    Cullin-RING ubiquitin ligases (CRL) regulate numerous biological processes in the heart and have been implicated in regulating cardiac hypertrophy. This study aimed to identify novel hypertrophy-modulating CRLs in cardiomyocytes (CM). A functional genomic approach using siRNA-mediated depletion and automated microscopy was employed to screen for cell size-modulating CRLs in neonatal rat CM. Screening hits were confirmed by 3H-isoleucine incorporation. Of 43 targets screened, siRNA-mediated depletion of Fbxo6, Fbxo45, and Fbxl14 resulted in decreased cell size, whereas depletion of Fbxo9, Fbxo25, Fbxo30, Fbxo32, Fbxo33, Cullin1, Roc1, Ddb1, Fbxw4, and Fbxw5 led to a markedly increased cell size under basal conditions. In CM stimulated with phenylephrine (PE), depletion of Fbxo6, Fbxo25, Fbxo33, Fbxo45, and Fbxw4 further augmented PE-induced hypertrophy. As a proof-of-concept, the CRLFbox25 was analysed by transverse aortic constriction (TAC) resulting in a 4.5-fold increase in Fbxo25 protein concentrations compared to control animals. In cell culture, siRNA-mediated depletion of Fbxo25 resulted in a ∼ 37% increase in CM cell size and ∼41% increase in 3H-isoleucine incorporation. Depleting Fbxo25 resulted in upregulation of Anp and Bnp. In summary, we identified 13 novel CRLs as positive or negative regulators of CM hypertrophy. Of these, CRLFbox25 was further characterized, as a potential modulator of cardiac hypertrophy.
    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
    感觉神经元兴奋过度是病理性疼痛的关键驱动因素,可能由轴突损伤引起,炎症,或神经元应激。G蛋白偶联受体(GPCR)信号可以通过调节Trp家族离子型受体和电压门控离子通道的激活来诱导疼痛放大。这里,我们试图利用钙成像技术来鉴定介导感觉神经元致敏并导致过度兴奋的细胞内通路的新型抑制剂.我们确定了一种由SST2R激动剂L-054,264组成的新型刺激混合物,和CYM5541,一种S1PR3激动剂,在体外引起小鼠初级感觉神经元的钙反应以及在体内引起小鼠的疼痛和热超敏反应。我们筛选了906种生物活性化合物的文库,并鉴定了由L-054,264/CYM5541引起的降低钙通量的24个命中。在这些热门歌曲中,水飞蓟素,从水飞蓟中提取的天然产物,强烈减少了刺激鸡尾酒的激活,以及含有缓激肽和前列腺素E2的独特炎症混合物。水飞蓟素在基线时对感觉神经元兴奋性没有影响,但是通过Orai通道和磷脂酶C信号传导的下游介质减少了钙通量。在体内,水飞蓟素预处理阻断佐剂介导的热超敏反应的发展,表明作为抗炎镇痛药的潜在用途。
    Sensory neuron hyperexcitability is a critical driver of pathological pain and can result from axon damage, inflammation, or neuronal stress. G-protein coupled receptor signaling can induce pain amplification by modulating the activation of Trp-family ionotropic receptors and voltage-gated ion channels. Here, we sought to use calcium imaging to identify novel inhibitors of the intracellular pathways that mediate sensory neuron sensitization and lead to hyperexcitability. We identified a novel stimulus cocktail, consisting of the SSTR2 agonist L-054,264 and the S1PR3 agonist CYM5541, that elicits calcium responses in mouse primary sensory neurons in vitro as well as pain and thermal hypersensitivity in mice in vivo. We screened a library of 906 bioactive compounds and identified 24 hits that reduced calcium flux elicited by L-054,264/CYM5541. Among these hits, silymarin, a natural product derived from milk thistle, strongly reduced activation by the stimulation cocktail, as well as by a distinct inflammatory cocktail containing bradykinin and prostaglandin E2. Silymarin had no effect on sensory neuron excitability at baseline, but reduced calcium flux via Orai channels and downstream mediators of phospholipase C signaling. In vivo, silymarin pretreatment blocked development of adjuvant-mediated thermal hypersensitivity, indicating potential use as an anti-inflammatory analgesic.
    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
    抗疟药治疗的一个主要挑战是出现对青蒿素类联合疗法(ACTs)成分的耐药性,以及迫切需要通过新的作用机制发挥作用的新药。在过去的十年里,在表型高通量筛选(HTS)中鉴定的化合物为目前在疟疾药物风险(MMV)临床开发组合中的六种候选药物提供了起点。然而,已发布的筛选数据为疟疾药物发现项目提供了许多新的化学物质,这些数据已被广泛挖掘。在这里,我们提出了一种新的筛选和选择级联反应,用于产生对恶性疟原虫血液阶段有活性的命中化合物。此外,我们通过测试141786种化合物的文库来验证我们的方法,该文库之前没有报告过用于抗疟疾的测试.HitGenerationLibrary1(HGL1)旨在最大化具有与进一步开发潜力相关的物理化学性质的化合物的化学多样性和新颖性。包含正交功效和细胞毒性测定的稳健HTS级联,包括新开发和验证的基于纳米荧光素酶的测定法用于分析化合物。确定了75种化合物(筛选活性命中率为0.05%),符合我们对药物敏感(NF54)和耐药(Dd2)寄生虫菌株(IC50≤2µM)效力的严格选择标准,在HepG2细胞中快速的作用速度和细胞活力(IC50≥10µM)。在进一步剖析之后,鉴定了33种化合物,其符合MMV确认的活性谱,并且是新的抗疟药物发现项目的高质量起点。
    A central challenge of antimalarial therapy is the emergence of resistance to the components of artemisinin-based combination therapies (ACTs) and the urgent need for new drugs acting through novel mechanism of action. Over the last decade, compounds identified in phenotypic high throughput screens (HTS) have provided the starting point for six candidate drugs currently in the Medicines for Malaria Venture (MMV) clinical development portfolio. However, the published screening data which provided much of the new chemical matter for malaria drug discovery projects have been extensively mined. Here we present a new screening and selection cascade for generation of hit compounds active against the blood stage of Plasmodium falciparum. In addition, we validate our approach by testing a library of 141,786 compounds not reported earlier as being tested against malaria. The Hit Generation Library 1 (HGL1) was designed to maximise the chemical diversity and novelty of compounds with physicochemical properties associated with potential for further development. A robust HTS cascade containing orthogonal efficacy and cytotoxicity assays, including a newly developed and validated nanoluciferase-based assay was used to profile the compounds. 75 compounds (Screening Active hit rate of 0.05%) were identified meeting our stringent selection criteria of potency in drug sensitive (NF54) and drug resistant (Dd2) parasite strains (IC50 ≤ 2 µM), rapid speed of action and cell viability in HepG2 cells (IC50 ≥ 10 µM). Following further profiling, 33 compounds were identified that meet the MMV Confirmed Active profile and are high quality starting points for new antimalarial drug discovery projects.
    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
    Tay-Sachs病(TSD)是一种常染色体隐性遗传疾病,具有进行性神经退行性表现。它影响了每100,000个活产中的一个。目前,没有批准的治疗或治愈方法。这篇综述总结了TSD的多种药物开发策略,包括酶替代疗法,药物伴侣疗法,底物减少疗法,基因治疗,和造血干细胞替代疗法。描述了体外和体内系统以评估上述治疗策略的功效。此外,我们讨论了使用MALDI质谱对化合物库进行高通量筛选。这使得能够发现减少GM2的化合物并且可以导致TSD疗法的进一步发展。
    Tay-Sachs disease (TSD) is an autosomal recessive disease that features progressive neurodegenerative presentations. It affects one in 100,000 live births. Currently, there is no approved therapy or cure. This review summarizes multiple drug development strategies for TSD, including enzyme replacement therapy, pharmaceutical chaperone therapy, substrate reduction therapy, gene therapy, and hematopoietic stem cell replacement therapy. In vitro and in vivo systems are described to assess the efficacy of the aforementioned therapeutic strategies. Furthermore, we discuss using MALDI mass spectrometry to perform a high throughput screen of compound libraries. This enables discovery of compounds that reduce GM2 and can lead to further development of a TSD therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号