gene modification

基因修饰
  • 文章类型: Journal Article
    嵌合抗原受体(CAR)T细胞疗法在过去十年中被证明是癌症治疗的突破。对血液恶性肿瘤产生前所未有的效果。所有批准的CAR-T细胞产品,以及许多正在临床试验中评估的人,使用病毒载体将外源遗传物质部署到T细胞中产生。病毒载体在基因传递中具有长期的临床历史,因此进行了迭代优化,以提高其效率和安全性。尽管如此,它们半随机整合到宿主基因组中的能力使它们可能通过插入诱变和关键细胞基因的失调而致癌。CART细胞施用后的继发性癌症似乎是罕见的不良事件。然而,过去几年记录的几起案件引起了人们对这个问题的关注,到目前为止可能被低估了,考虑到相对较新的CART细胞疗法的部署。此外,在血液恶性肿瘤中获得的最初成功尚未在实体瘤中复制。现在很明显,需要进一步的增强才能使CAR-T细胞增加长期持久性,克服疲惫,应对免疫抑制肿瘤微环境。为了这个目标,各种基因组工程策略正在评估中,最依赖CRISPR/Cas9或其他基因编辑技术。这些方法很容易引入意想不到的,产物细胞中不可逆的基因组改变。在本文的第一部分,我们将讨论用于产生CAR-T细胞的病毒和非病毒方法,而在第二部分,我们将专注于基因编辑和非基因编辑T细胞工程,特别是在优势方面,局限性,和安全。最后,我们将批判性地分析不同的基因部署和基因组工程组合,描述具有卓越安全性的下一代CAR-T细胞生产策略。
    Chimeric antigen receptor (CAR) T-cell therapy has proven a breakthrough in cancer treatment in the last decade, giving unprecedented results against hematological malignancies. All approved CAR T-cell products, as well as many being assessed in clinical trials, are generated using viral vectors to deploy the exogenous genetic material into T-cells. Viral vectors have a long-standing clinical history in gene delivery, and thus underwent iterations of optimization to improve their efficiency and safety. Nonetheless, their capacity to integrate semi-randomly into the host genome makes them potentially oncogenic via insertional mutagenesis and dysregulation of key cellular genes. Secondary cancers following CAR T-cell administration appear to be a rare adverse event. However several cases documented in the last few years put the spotlight on this issue, which might have been underestimated so far, given the relatively recent deployment of CAR T-cell therapies. Furthermore, the initial successes obtained in hematological malignancies have not yet been replicated in solid tumors. It is now clear that further enhancements are needed to allow CAR T-cells to increase long-term persistence, overcome exhaustion and cope with the immunosuppressive tumor microenvironment. To this aim, a variety of genomic engineering strategies are under evaluation, most relying on CRISPR/Cas9 or other gene editing technologies. These approaches are liable to introduce unintended, irreversible genomic alterations in the product cells. In the first part of this review, we will discuss the viral and non-viral approaches used for the generation of CAR T-cells, whereas in the second part we will focus on gene editing and non-gene editing T-cell engineering, with particular regard to advantages, limitations, and safety. Finally, we will critically analyze the different gene deployment and genomic engineering combinations, delineating strategies with a superior safety profile for the production of next-generation CAR T-cell.
    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
    精确切割肺切片(PCLS),离体3D肺组织模型,已广泛用于肺部研究中的各种应用。PCLS充当体外和体内模型之间的极好中介,因为它们将所有驻留细胞类型保留在其天然生态位内,同时保留细胞外基质环境。该方案描述了TReATS(组织切片中TAT-Cre重组酶介导的Floxed等位基因修饰)方法,该方法能够在源自成年Floxed动物的PCLS中进行快速有效的基因修饰。这里,我们提出了TReATS方法的详细协议,由两个简单步骤组成:PCLS生成和在TAT-Cre重组酶溶液中孵育。基因修饰的后续验证涉及PCLS的活染色和成像,实时定量PCR,和细胞活力评估。这个为期四天的协议消除了复杂的Cre繁殖的需要,规避与基因突变相关的过早致死问题,并大大减少了动物的使用。TReATS方法为复杂的离体组织模型中的基因修饰提供了简单且可重复的解决方案,加快基因功能的研究,疾病机制,以及药物靶点的发现。关键特征•在四天内在复杂的基于组织的模型中实现永久性离体基因修饰。•可应用于诱导基因缺失或激活的高度适应性基因修饰方法。•允许在受控离体设置中进行简单的Cre剂量测试,其优点是使用从与真实对照相同的动物产生的PCLS。•通过优化,该方法可应用于其他器官的精确切割组织切片。
    Precision-cut lung slices (PCLS), ex vivo 3D lung tissue models, have been widely used for various applications in lung research. PCLS serve as an excellent intermediary between in vitro and in vivo models because they retain all resident cell types within their natural niche while preserving the extracellular matrix environment. This protocol describes the TReATS (TAT-Cre recombinase-mediated floxed allele modification in tissue slices) method that enables rapid and efficient gene modification in PCLS derived from adult floxed animals. Here, we present detailed protocols for the TReATS method, consisting of two simple steps: PCLS generation and incubation in a TAT-Cre recombinase solution. Subsequent validation of gene modification involves live staining and imaging of PCLS, quantitative real-time PCR, and cell viability assessment. This four-day protocol eliminates the need for complex Cre-breeding, circumvents issues with premature lethality related to gene mutation, and significantly reduces the use of animals. The TReATS method offers a simple and reproducible solution for gene modification in complex ex vivo tissue-based models, accelerating the study of gene function, disease mechanisms, and the discovery of drug targets. Key features • Achieve permanent ex vivo gene modifications in complex tissue-based models within four days. • Highly adaptable gene modification method that can be applied to induce gene deletion or activation. • Allows simple Cre dosage testing in a controlled ex vivo setting with the advantage of using PCLS generated from the same animal as true controls. • With optimisation, this method can be applied to precision-cut tissue slices of other organs.
    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
    Neurodegenerative diseases (NDs) are a group of debilitating neurological disorders that primarily affect elderly populations and include Alzheimer\'s disease (AD), Parkinson\'s disease (PD), Huntington\'s disease (HD), and amyotrophic lateral sclerosis (ALS). Currently, there are no therapies available that can delay, stop, or reverse the pathological progression of NDs in clinical settings. As the population ages, NDs are imposing a huge burden on public health systems and affected families. Animal models are important tools for preclinical investigations to understand disease pathogenesis and test potential treatments. While numerous rodent models of NDs have been developed to enhance our understanding of disease mechanisms, the limited success of translating findings from animal models to clinical practice suggests that there is still a need to bridge this translation gap. Old World non-human primates (NHPs), such as rhesus, cynomolgus, and vervet monkeys, are phylogenetically, physiologically, biochemically, and behaviorally most relevant to humans. This is particularly evident in the similarity of the structure and function of their central nervous systems, rendering such species uniquely valuable for neuroscience research. Recently, the development of several genetically modified NHP models of NDs has successfully recapitulated key pathologies and revealed novel mechanisms. This review focuses on the efficacy of NHPs in modeling NDs and the novel pathological insights gained, as well as the challenges associated with the generation of such models and the complexities involved in their subsequent analysis.
    神经退行性疾病 (Neurodegenerative diseases,ND) 是一类主要影响老年人群的神经疾病,包括阿尔茨海默病 (Alzheimer’s disease,AD)、帕金森病 (Parkinson’s disease,PD)、亨廷顿病 (Huntington’s disease,HD) 和肌萎缩侧索硬化症 (Amyotrophic lateral sclerosis,ALS)。目前,临床上尚无可以延迟、阻止或逆转 ND 病理进展的疗法。随着人口老龄化,ND 给公共卫生系统和患者家庭带来巨大负担。动物模型是临床前研究了解疾病发病机制和测试潜在治疗方法的重要工具。虽然已经开发出许多神经退行性疾病的啮齿类动物模型,并极大地增强了我们对疾病机制的理解,但动物模型的研究结果临床转化效果低下,这表明仍然需要弥合临床前研究和临床研究之间的差距。旧大陆非人类灵长类动物(non-human primates,NHPs),如恒河猴、食蟹猴和黑长尾猴,在系统发育、生理、生化和行为上,特别是在中枢神经系统结构和功能方面与人类最为接近,这使得它们对神经科学的研究具有重要作用。最近,已经有几种神经退行性疾病的转基因 NHP 模型报道,它们复刻了关键病理学指征并揭示了新的分子机制。在这篇综述中,我们描述了 NHP 模拟 ND 的能力以及来自这些 NHP 模型的新病理学见解。我们还讨论了模型制备及后续研究中面临的挑战。.
    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
    已经研究了转录变异,但尚未在疟原虫中研究由于RNA编辑引起的转录后修饰。我们研究了恶性疟原虫3D7中选定基因的发育阶段特异性RNA编辑。我们在紧密同步的疟原虫中在8、16、24、32、40和46h检测到广泛的氨基和脱氨基类型的RNA编辑。大多数编辑事件在8和16h环期寄生虫中观察到。在16h环阶段(25%)比8h环阶段(20%)检测到更多的A到G脱氨类型编辑。在16h环阶段(31%)比8h环阶段(22%)检测到更多的U到C胺化类型编辑。在28S,rRNA编辑将环结构转化为茎结构。PF3D7_0216900的血红蛋白结合活性也由于RNA编辑而改变。在表达的28SrRNA基因中,PF3D7_0532000和PF3D7_0726000表达量较高。发现这两个基因的转录量增加,特别是环阶段的PF3D7_0726000和滋养体和裂殖体阶段的PF3D7_0532000。腺苷脱氨酶(ADA)表达与编辑水平无关。RNA编辑的第一个实验报告将有助于确定可能对抗疟药物发现和疟疾控制有用的编辑机制。
    Transcriptional variation has been studied but post-transcriptional modification due to RNA editing has not been investigated in Plasmodium. We investigated developmental stage-specific RNA editing in selected genes in Plasmodium falciparum 3D7. We detected extensive amination- and deamination-type RNA editing at 8, 16, 24, 32, 40, and 46 h in tightly synchronized Plasmodium. Most of the editing events were observed in 8 and 16 h ring-stage parasites. Extensive A-to-G deamination-type editing was detected more during the 16 h ring stage (25%) than the 8 h ring stage (20%). Extensive U-to-C amination-type editing was detected more during the 16 h ring stage (31%) than the 8 h ring stage (22%). In 28S, rRNA editing converted the loop structure to the stem structure. The hemoglobin binding activity of PF3D7_0216900 was also altered due to RNA editing. Among the expressed 28S rRNA genes, PF3D7_0532000 and PF3D7_0726000 expression was higher. Increased amounts of the transcripts of these two genes were found, particularly PF3D7_0726000 in the ring stage and PF3D7_0532000 in the trophozoite and schizont stages. Adenosine deaminase (ADA) expression did not correlate with the editing level. This first experimental report of RNA editing will help to identify the editing machinery that might be useful for antimalarial drug discovery and malaria control.
    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:SARS-CoV-2的出现对老年人和免疫功能低下的个体造成生命威胁,而这些人群的治疗有限。据报道,间充质基质细胞(MSC)可用于SARS-CoV-2治疗并减少SARS-CoV-2相关后遗症。
    结果:在这项研究中,我们开发了一种自主的细胞机器,以临床级MSCs为基础,不断在体内秘密中和抗体,对抗SARS-CoV-2感染.首先,构建各种修饰的重组质粒,并通过电穿孔转染临床级MSCs,用于中和抗SARS-CoV-2抗体的组装和表达。第二,通过假病毒中和试验筛选出稳定的分泌抗体的MSCs克隆。最后,我们研究了工程化MSCs分泌的中和抗体在体内的药代动力学和生物分布。稳定的临床级MSCs克隆,表达XGv347-10和LY-CoV1404-5中和抗体,显示了它们对SARS-CoV-2感染的可行性和保护作用。移植的工程化临床级MSCs有效地将SARS-CoV-2抗体递送至肺部,由COVID-19引起的免疫高反应由MSC克隆通过抑制CD4T细胞分化为Th1和Th17亚群来协调。
    结论:我们的数据表明,作为细胞生产机器分泌有效中和抗体的工程化临床级MSCs具有对抗SARS-CoV-2感染的潜力,这为有效治疗老年和免疫功能低下的COVID-19患者提供了新的途径。
    BACKGROUND: The emergence of SARS-CoV-2 becomes life-threatening for the older and immunocompromised individuals, whereas limited treatment is available on these populations. Mesenchymal stromal cells (MSCs) have been reported to be useful in SARS-CoV-2 treatment and reduce SARS-CoV-2-related sequelae.
    RESULTS: In this study, we developed an autonomous cellular machine to secret neutralizing antibody in vivo constantly based on the clinical-grade MSCs, to combat SARS-CoV-2 infections. First, various modified recombinant plasmids were constructed and transfected into clinical-grade MSCs by electroporation, for assembly and expression of neutralizing anti-SARS-CoV-2 antibodies. Second, the stable antibody secreting MSCs clones were screened through pseudovirus neutralization assay. Finally, we investigated the pharmacokinetics and biodistribution of neutralizing antibody secreted by engineered MSCs in vivo. The stable clinical-grade MSCs clones, expressing XGv347-10 and LY-CoV1404-5 neutralizing antibodies, exhibited their feasibility and protective efficacy against SARS-CoV-2 infection. Transplanted engineered clinical-grade MSCs effectively delivered the SARS-CoV-2 antibodies to the lung, and the immune hyperresponsiveness caused by COVID-19 was coordinated by MSC clones through inhibiting the differentiation of CD4 + T cells into Th1 and Th17 subpopulations.
    CONCLUSIONS: Our data suggested that engineered clinical-grade MSCs secreting effective neutralizing antibodies as cellular production machines had the potential to combat SARS-CoV-2 infection, which provided a new avenue for effectively treating the older and immunocompromised COVID-19 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
    基因修饰疗法处于HIV-1治愈策略的最前沿。嵌合抗原受体(CAR)-T细胞在抗逆转录病毒疗法期间或在分析治疗中断(ATI)之后构成靶向感染细胞的潜在方法。然而,在慢病毒CAR基因递送的背景下,在HIV-1感染细胞和CAR-T细胞的定量以及在表达靶抗原的细胞的鉴定方面存在技术挑战。首先,缺乏有效的技术来鉴定和表征ART抑制和病毒血症个体中表达高变HIVgp120的细胞.第二,基于慢病毒的CAR-T基因修饰载体与HIV-1保守区之间的紧密序列同源性对HIV-1和慢病毒载体水平产生了定量挑战.需要考虑在CAR-T细胞和其他基于慢病毒载体的疗法中标准化HIV-1DNA/RNA测定,以避免这些混杂的相互作用。最后,随着在CAR-T细胞中引入HIV-1抗性基因,需要具有单细胞分辨率的测定法来确定基因插入物防止CAR-T细胞在体内被感染的能力。随着新疗法在HIV-1治愈领域的不断出现,解决CAR-T细胞治疗中的这些挑战将至关重要。
    Gene-modification therapies are at the forefront of HIV-1 cure strategies. Chimeric antigen receptor (CAR)-T cells pose a potential approach to target infected cells during antiretroviral therapy or following analytical treatment interruption (ATI). However, there are technical challenges in the quantification of HIV-1-infected and CAR-T cells in the setting of lentiviral CAR gene delivery and also in the identification of cells expressing target antigens. First, there is a lack of validated techniques to identify and characterize cells expressing the hypervariable HIV gp120 in both ART-suppressed and viremic individuals. Second, close sequence homology between lentiviral-based CAR-T gene modification vectors and conserved regions of HIV-1 creates quantification challenges of HIV-1 and lentiviral vector levels. Consideration needs to be taken into standardizing HIV-1 DNA/RNA assays in the setting of CAR-T cell and other lentiviral vector-based therapies to avoid these confounding interactions. Lastly, with the introduction of HIV-1 resistance genes in CAR-T cells, there is a need for assays with single-cell resolution to determine the competence of the gene inserts to prevent CAR-T cells from becoming infected in vivo. As novel therapies continue to arise in the HIV-1 cure field, resolving these challenges in CAR-T-cell therapy will be crucial.
    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
    对于治疗选择有限的缺血性心肌病(ICM),动脉生成的诱导有可能通过主要的血流恢复来改善心脏功能。我们假设Notch信号修饰的间充质干细胞(SB623细胞)贴片的移植将在缺血性病变中诱导血管生成和动脉生成,导致大鼠ICM模型中左心室(LV)功能的改善。诱导缺血两周后,SB623细胞补片移植入ICM大鼠(SB组,n=10)或假手术(无治疗组,进行n=10)。SB623细胞补片移植后6周LV射血分数显著提高(P<0.001)。组织学发现,vonWillebrand因子(vWF)阳性毛细血管(P<0.01)和直径大于20µm的α平滑肌肌动蛋白(αSMA)和vWF阳性小动脉(P=0.002)的数量显着增加SB组,提示血管生成和动脉生成的诱导。此外,SB623细胞补片移植大鼠心肌细胞显示ephrin-B2(P=0.03)和EphB4(P=0.01)基因表达上调,指示动脉生成诱导。总之,SB623细胞补片移植通过诱导大鼠ICM模型中的血管生成和动脉生成来改善LV功能。
    For ischemic cardiomyopathy (ICM) with limited therapeutic options, the induction of arteriogenesis has the potential to improve cardiac function through major restoration of blood flow. We hypothesized that transplantation of a Notch signaling-modified mesenchymal stem cell (SB623 cell) patch would induce angiogenesis and arteriogenesis in ischemic lesions, leading to improvement of left ventricular (LV) function in a rat ICM model. Two weeks after the induction of ischemia, SB623 cell patch transplantation into ICM rats (SB group, n = 10) or a sham operation (no-treatment group, n = 10) was performed. The LV ejection fraction was significantly improved at 6 weeks after SB623 cell patch transplantation (P < 0.001). Histological findings revealed that the number of von Willebrand factor (vWF)-positive capillary vessels (P < 0.01) and alpha smooth muscle actin (αSMA)- and vWF-positive arterioles with a diameter greater than 20 µm (P = 0.002) was significantly increased in the SB group, suggesting the induction of angiogenesis and arteriogenesis. Moreover, rat cardiomyocytes treated with SB623 cell patch transplantation showed upregulation of ephrin-B2 (P = 0.03) and EphB4 (P = 0.01) gene expression, indicating arteriogenesis induction. In conclusion, SB623 cell patch transplantation improved LV function by inducing angiogenesis and arteriogenesis in a rat ICM model.
    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
    相关目标的识别和表征对于开发非激素男性避孕药是必要的。分子必须证明它们是繁殖所必需的。因此,需要一种复杂的技术来确定非激素男性避孕药的分子靶标。遗传修饰(GM)技术是可以应用的一种方法。该技术已广泛用于研究影响男性生育能力的基因功能,并发现了许多非激素男性避孕靶分子。我们研究了转基因技术和方法,用于研究与男性生育力有关的基因作为非激素避孕药的潜在目标。通过使用转基因技术增加了非激素避孕候选分子的发现,特别是集群定期间隔短回文重复/Cas9方法。候选非激素避孕分子的发现可以是开发非激素男性避孕药的广泛研究。因此,我们相信有一天非荷尔蒙的男性避孕药会被释放。
    The identification and characterization of relevant targets are necessary for developing nonhormonal male contraceptives. The molecules must demonstrate that they are necessary for reproduction. As a result, a sophisticated technique is required to identify the molecular targets for nonhormonal male contraceptives. Genetic modification (GM) techniques are one method that can be applied. This technique has been widely used to study gene function that effected male fertility and has resulted in the discovery of numerous nonhormonal male contraceptive target molecules. We examined GM techniques and approaches used to investigate genes involved in male fertility as potential targets for nonhormonal contraceptives. The discovery of nonhormonal contraceptive candidate molecules was increased by using GM techniques, especially the Clustered Regularly Interspaced Short Palindromic Repeats/Cas9 method. The discovery of candidate nonhormonal contraceptive molecules can be a wide-open research for the development of nonhormonal male contraceptives. Therefore, we are believing that one day nonhormonal male contraceptives will be released.
    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
    放射治疗(RT)是多发性胸部恶性肿瘤的有效治疗选择,包括肺癌,胸腺肿瘤,和气管癌。放射性肺损伤(RILI)是放射治疗的严重并发症。辐射引起肺细胞和组织的损伤。多种因素导致放射性肺损伤的进展,包括基因改变,氧化应激,和炎症反应。尤其是,辐射源通过水分子的直接激发和电离促进氧化应激的发生,这导致水分子的分解和活性氧(ROS)的产生,活性氮物种(RNS)。随后,活性氧和活性氮的过量产生可引起DNA的氧化损伤。免疫细胞和多种信号分子在整个过程中起着重要作用。间充质干细胞(MSCs)是具有多向分化潜能的多能干细胞,正在研究治疗放射性肺损伤。间充质干细胞可以通过靶向多种信号分子来调节免疫细胞并控制抗氧化剂和促氧化剂之间的平衡来保护正常肺细胞免受损伤。从而抑制炎症和纤维化。基因修饰的间充质干细胞可以提高间充质干细胞的自然功能,包括细胞存活,组织再生,和归巢。这些重编程的间充质干细胞可以产生所需的产物,包括细胞因子,受体,和酶,这有助于间充质干细胞治疗应用的进一步发展。这里,本文综述了放射性肺损伤的分子机制,并讨论了间充质干细胞在预防和治疗放射性肺损伤方面的潜力。这些关键问题的澄清将使骨髓间充质干细胞成为临床上治疗放射性肺损伤的更奇妙的新策略。读者可以在这个领域有一个全面的了解。
    Radiotherapy (RT) is an effective treatment option for multiple thoracic malignant tumors, including lung cancers, thymic cancers, and tracheal cancers. Radiation-induced lung injury (RILI) is a serious complication of radiotherapy. Radiation causes damage to the pulmonary cells and tissues. Multiple factors contribute to the progression of Radiation-induced lung injury, including genetic alterations, oxidative stress, and inflammatory responses. Especially, radiation sources contribute to oxidative stress occurrence by direct excitation and ionization of water molecules, which leads to the decomposition of water molecules and the generation of reactive oxygen species (ROS), reactive nitrogen species (RNS). Subsequently, reactive oxygen species and reactive nitrogen species overproduction can induce oxidative DNA damage. Immune cells and multiple signaling molecules play a major role in the entire process. Mesenchymal stem cells (MSCs) are pluripotent stem cells with multiple differentiation potentials, which are under investigation to treat radiation-induced lung injury. Mesenchymal stem cells can protect normal pulmonary cells from injury by targeting multiple signaling molecules to regulate immune cells and to control balance between antioxidants and prooxidants, thereby inhibiting inflammation and fibrosis. Genetically modified mesenchymal stem cells can improve the natural function of mesenchymal stem cells, including cellular survival, tissue regeneration, and homing. These reprogrammed mesenchymal stem cells can produce the desired products, including cytokines, receptors, and enzymes, which can contribute to further advances in the therapeutic application of mesenchymal stem cells. Here, we review the molecular mechanisms of radiation-induced lung injury and discuss the potential of Mesenchymal stem cells for the prevention and treatment of radiation-induced lung injury. Clarification of these key issues will make mesenchymal stem cells a more fantastic novel therapeutic strategy for radiation-induced lung injury in clinics, and the readers can have a comprehensive understanding in this fields.
    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
    特异性靶向核酸酶是高精度介导基因组替代表达的有力工具。来自微生物成簇规则间隔短回文重复(CRISPR)适应性免疫系统的RNA序列引导的Cas9核酸酶可用于通过使用20-nt靶向序列促进细胞中的基因组工程。在这一章中,我们描述了通过非同源末端连接(NHEJ)或同源定向修复(HDR)在下游功能研究的修饰细胞系的产生中用于Cas9介导的基因组编辑的一组工具。该协议为目标位点的选择提供了实验得出的指南,切割效率的评估,和脱靶活动分析。从目标设计开始,我们将涵盖基因修饰和修饰的克隆细胞系。
    A specific targeting nuclease is a powerful tool for mediating genome alternative expression with high precision. The RNA sequence-guided Cas9 nuclease from the microbial clustered regularly interspaced short palindromic repeats (CRISPR) adaptive immune system can be used to facilitate genome engineering in cells by using a 20-nt targeting sequence. In this chapter, we describe a set of tools for Cas9-mediated genome editing via non-homologous end joining (NHEJ) or homology-directed repair (HDR) in the generation of modified cell lines for downstream functional studies. This protocol provides experimentally derived guidelines for the selection of target sites, evaluation of cleavage efficiency, and analysis of off-target activity. Beginning with target design, we will cover gene modifications and modified clonal cell lines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号