Cre-loxP system

Cre - loxP 系统
  • 文章类型: Journal Article
    结节性皮肤病病毒(LSDV)是一种在亚洲迅速崛起的病原体,包括中国。LSDV的遗传操作对于阐明LSDV编码蛋白的致病机制和生物学功能至关重要。在这项研究中,我们在VACV的改良的早晚期H5启动子下建立了Cre-loxP重组系统的平台,以快速构建重组LSDV病毒。重组病毒,LSDV-EGFP-ΔTK,使用连续有限稀释和挑选单细胞方法纯化和获得。使用慢病毒包装系统,建立Cre重组酶稳定表达的MDBK细胞系以提供用于报告基因切除的Cre重组酶。一个基因稳定的,使用同源重组和Cre-loxP系统构建了安全的TK基因缺失的LSDV(LSDV-ΔTK)。在MDBK-Cre细胞系中使用有限稀释对其进行纯化。建立Cre-loxP重组系统将能够从LSDV基因组中顺序删除感兴趣的基因,并对LSDV基因组进行遗传操作。为开发减毒LSDV疫苗提供技术支持和平台。
    Lumpy skin disease virus (LSDV) is a rapidly emerging pathogen in Asia, including China. Genetic manipulation of the LSDV is essential for the elucidation of the pathogenic mechanism and biological function of the LSDV-encoded protein. In this study, we established a platform for the Cre-loxP recombination system under a modified early-late H5 promoter of the VACV for quick construction of the recombinant LSDV virus. The recombinant virus, LSDV-EGFP-ΔTK, was purified and obtained using serial limited dilution and picking the single cells methods. Using the lentiviral package system, a Cre recombinase enzyme stable expression MDBK cell line was established to supply the Cre recombinase for the reporter gene excision. A genetically stable, safe TK gene-deleted LSDV (LSDV-ΔTK) was constructed using homologous recombination and the Cre-loxP system. It was purified using limited dilution in the MDBK-Cre cell line. Establishing the Cre-loxP recombination system will enable sequential deletion of the interested genes from the LSDV genome and genetic manipulation of the LSDV genome, providing technical support and a platform for developing the attenuated LSDV vaccine.
    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
    血管疾病,包括动脉粥样硬化和腹部动脉瘤,是全球老年人死亡和发病的主要原因。由于不充分的治疗方法和有限的药物靶标,患者的生活质量显著受损。为了扩大我们对血管疾病的理解,基因敲除(KO)小鼠,尤其是条件性敲除(cKO)小鼠,被广泛用于研究基因功能和作用机制。Cre-loxP系统是产生cKO小鼠的最常用方法。已经建立了许多Cre驱动小鼠来研究组成血管的主要细胞类型,包括内皮细胞,平滑肌细胞,和成纤维细胞。这里,我们首先讨论了动脉壁各层的特征。接下来,我们概述了用于血管壁中每种主要细胞类型的代表性Cre驱动小鼠及其在血管生物学中的最新应用。然后,我们讨论Cre毒性,并讨论最小化Cre干扰实验结果的实用方法。最后,我们通过引入革命性的单细胞RNA测序和双重组酶系统来展望组织特异性Cre驱动的未来。
    Vascular diseases, including atherosclerosis and abdominal aneurysms, are the primary cause of mortality and morbidity among the elderly worldwide. The life quality of patients is significantly compromised due to inadequate therapeutic approaches and limited drug targets. To expand our comprehension of vascular diseases, gene knockout (KO) mice, especially conditional knockout (cKO) mice, are widely used for investigating gene function and mechanisms of action. The Cre-loxP system is the most common method for generating cKO mice. Numerous Cre driver mice have been established to study the main cell types that compose blood vessels, including endothelial cells, smooth muscle cells, and fibroblasts. Here, we first discuss the characteristics of each layer of the arterial wall. Next, we provide an overview of the representative Cre driver mice utilized for each of the major cell types in the vessel wall and their most recent applications in vascular biology. We then go over Cre toxicity and discuss the practical methods for minimizing Cre interference in experimental outcomes. Finally, we look into the future of tissue-specific Cre drivers by introducing the revolutionary single-cell RNA sequencing and dual recombinase system.
    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
    CCN2 has been shown to be closely involved in the progression of renal fibrosis, indicating the potential of CCN2 inhibition as a therapeutic target. Although the examination of the renal disease phenotypes of adult CCN2 knockout mice has yielded valuable scientific insights, perinatal death has limited studies of CCN2 in vivo. Conditional knockout technology has become widely used to delete genes in the target cell populations or time points using cell-specific Cre recombinase-expressing mice. Therefore, several lines of CCN2-floxed mice have been developed to assess the functional role of CCN2 in adult mice.CCN2 levels are elevated in renal fibrosis and proliferative glomerulonephritis, making them suitable disease models for assessing the effects of CCN2 deletion on the kidney. Renal fibrosis is characterized by glomerulosclerosis and tubulointerstitial fibrosis and transforming growth factor-β. CCN2 is increased in fibrosis and modulates a number of downstream signaling pathways involved in the fibrogenic properties of TGF-β. Unilateral ureteral obstruction is one of the most widely used models of renal tubulointerstitial fibrosis. In addition, anti-glomerular basement membrane antibody glomerulonephritis has become the most widely used model for evaluating the effect of increased renal CCN2 expression. Herein, we describe the construction of CCN2-floxed mice and inducible systemic CCN2 conditional knockout mice and methods for the operation of unilateral ureteral obstruction and the induction of anti-glomerular basement membrane antibody glomerulonephritis.
    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
    Renal interstitial fibrosis is the final common pathway in the process of all kidney diseases, and it results in chronic kidney disease. CCN2 is an important factor in the pathogenesis of renal interstitial fibrosis, and analysis of its function can lead to treatments for chronic kidney disease. Since CCN2 knockout mice are developmentally lethal, generation of conditional knockout mice is essential for in vivo analysis. Since CCN2 is expressed in a variety of cells in the kidney, including podocytes, mesangial cells, pericytes, and tubular epithelial cells, it is necessary to perform cell-specific verification of the cells that play a central role in fibrosis. However, cell-specific validation using the Cre/loxP system in vivo has only been performed in mesangial cells. In our research program, we are focusing on the role of CCN2 in tubular epithelial cells in renal fibrogenesis. In this report, we introduce the creation of a tubular epithelial cell-specific knockout model and method of its analysis.
    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
    MRP8-Cre-ires/EGFP转基因小鼠(Mrp8creTg,在C57BL/6J遗传背景上)在免疫学和血液学研究中很受欢迎,用于在中性粒细胞中特异性表达Cre重组酶和EGFP报告基因。它通常与携带loxP侧翼基因的其他转基因品系杂交,以在嗜中性粒细胞中实现限制性基因敲除。然而,由于线条的创建方式,关于宿主基因组中MRP8-Cre-ires/EGFP转基因的基本知识,例如其整合位点和侧翼序列,在很大程度上仍然未知,妨碍稳健的实验设计和数据解释。在这里,我们使用了一种最近开发的技术,靶向基因座扩增(TLA)测序,填补这些知识空白。我们发现MRP8-Cre-ires/EGFP转基因被整合到宿主小鼠基因组的5号染色体(5qG2)中。这种整合导致宿主基因组序列的44kb缺失,导致Serpine1的完全缺失和Ap1s1的部分缺失。确定了转基因的侧翼序列,我们设计了一种新的基因分型方案,可以区分纯合子,杂合子,和野生型Mrp8creTg小鼠。令我们惊讶的是,杂交杂合小鼠没有产生纯合Mrp8creTg小鼠,很可能是由于Ap1s1基因表达中断导致的产前致死性。
    The MRP8-Cre-ires/EGFP transgenic mouse (Mrp8creTg, on C57BL/6J genetic background) is popular in immunological and hematological research for specifically expressing Cre recombinase and an EGFP reporter in neutrophils. It is often crossed with other transgenic lines carrying loxP-flanked genes to achieve restricted gene knockout in neutrophils. However, due to the way in which the line was created, basic knowledge about the MRP8-Cre-ires/EGFP transgene in the host genome, such as its integration site(s) and flanking sequences, remains largely unknown, hampering robust experimental design and data interpretation. Here we used a recently developed technique, targeted locus amplification (TLA) sequencing, to fill these knowledge gaps. We found that the MRP8-Cre-ires/EGFP transgene was integrated into chromosome 5 (5qG2) of the host mouse genome. This integration led to a 44 kb deletion of the host genomic sequence, resulting in complete deletion of Serpine1 and partial deletion of Ap1s1. Having determined the flanking sequences of the transgene, we designed a new genotyping protocol that can distinguish homozygous, heterozygous, and wildtype Mrp8creTg mice. To our surprise, crossing heterozygous mice produced no homozygous Mrp8creTg mice, most likely due to prenatal lethality resulting from disrupted Ap1s1 gene 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: Journal Article
    源自人诱导多能干细胞(hiPSC)的肝细胞和肝类器官(HO)是有前途的基于细胞的肝病疗法。重编程转基因的去除可以影响hiPSC向三个胚层的分化潜力,但不会影响晚期发育阶段的肝细胞和肝类器官。在这里,我们使用基于Cre重组酶介导的loxP侧翼重编程盒去除的可切除多顺反子慢病毒载体,从正常人成纤维细胞中产生了hiPSCs.比较具有相同遗传背景的转基因携带和无转基因hiPSCs的特性,所有hiPSC的多能状态非常相似,如多能标记的表达所示,胚胎身体形成,和体外三谱系分化。然而,体外分化为肝细胞后,无转基因的hiPSC优于转基因残留的hiPSC。有趣的是,通过从hiPSCs中消除转基因,可以显着增强人肝类器官(hHOs)的生成和肝分化,如通过上调胎儿肝脏(CK19,SOX9和ITGA6)和功能性肝细胞(白蛋白,ASGR1,HNF4α,CYP1A2,CYP3A4和AAT)在分化培养基中培养后的标记。因此,消除重编程转基因有助于hiPSC分化为肝细胞样细胞和具有肝祖细胞特性的肝类器官。因此,我们的发现为iPSC衍生的肝类器官的特征提供了重要的见解。
    Hepatocytes and hepatic organoids (HOs) derived from human induced pluripotent stem cells (hiPSCs) are promising cell-based therapies for liver diseases. The removal of reprogramming transgenes can affect hiPSC differentiation potential into the three germ layers but not into hepatocytes and hepatic organoids in the late developmental stage. Herein, we generated hiPSCs from normal human fibroblasts using an excisable polycistronic lentiviral vector based on the Cre recombinase-mediated removal of the loxP-flanked reprogramming cassette. Comparing the properties of transgene-carrying and transgene-free hiPSCs with the same genetic background, the pluripotent states of all hiPSCs were quite similar, as indicated by the expression of pluripotent markers, embryonic body formation, and tri-lineage differentiation in vitro. However, after in vitro differentiation into hepatocytes, transgene-free hiPSCs were superior to the transgene-residual hiPSCs. Interestingly, the generation and hepatic differentiation of human hepatic organoids (hHOs) were significantly enhanced by transgene elimination from hiPSCs, as observed by the upregulated fetal liver (CK19, SOX9, and ITGA6) and functional hepatocyte (albumin, ASGR1, HNF4α, CYP1A2, CYP3A4, and AAT) markers upon culture in differentiation media. Thus, the elimination of reprogramming transgenes facilitates hiPSC differentiation into hepatocyte-like cells and hepatic organoids with properties of liver progenitor cells. Our findings thus provide significant insights into the characteristics of iPSC-derived hepatic organoids.
    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
    CRB1(面包屑同源物1)中的突变会引起罕见的视网膜疾病,例如12型色素性视网膜炎(RP12)和Leber先天性黑蒙8型(LCA8)。RP12导致周边视力逐渐恶化,而LCA8在出生时或早期会导致严重的视力障碍。虽然已经针对RP12提出了几种小鼠模型,但很少复制人类LCA8病理学的全谱,比如无序的视网膜分层,异常视网膜增厚,色素缺陷,过度反射病变,出生时视网膜电图反应严重减弱。已经提出了利用Cre-loxP系统删除特定视网膜细胞类型和发育阶段中的候选基因的六个模型。从眼睛发育开始烧蚀Crb1及其同系物Crb2(使用mRx-Cre)的模型是最完整的,因为它在睁眼阶段显示失明,RPE中的色素缺陷,神经节细胞层异位症,视网膜层片的破坏,和无细胞斑块。LCA8代表LCA亚型中一种独特的视网膜营养不良类型,由眼睛发育过程中功能失调的视网膜祖细胞驱动。相比之下,其他LCA类型和RP12是由感光体缺陷引起的。因此,最准确的LCA8样小鼠模型必须针对视囊泡中或更早的Crb1和Crb2基因的等位基因。
    Mutations in the CRB1 (Crumbs homolog 1) cause rare retinal diseases like retinitis pigmentosa type 12 (RP12) and Leber congenital amaurosis type 8 (LCA8). RP12 results in progressively worsening peripheral vision, whereas LCA8 causes severe visual impairment at birth or in early life. While several mouse models have been proposed for RP12, few replicate the full spectrum of human LCA8 pathology, such as disorganized retinal layering, abnormal retinal thickening, pigmentary defects, hyperreflective lesions, and severely attenuated electroretinogram responses at birth. Six models have been proposed utilizing the Cre-loxP system to delete candidate genes in specific retinal cell types and developmental stages. The model ablating Crb1 and its homolog Crb2 (using mRx-Cre) from the beginning of the eye development is the most complete as it shows blindness during the eye-opening stage, pigmentary defects in the RPE, ganglion cell layer heterotopia, disruption of retinal lamination, and acellular patches. LCA8 represents a unique type of retinal dystrophy among LCA subtypes, driven by dysfunctional retinal progenitor cells during eye development. In contrast, other LCA types and RP12 are caused by photoreceptor defects. Therefore, the most accurate LCA8-like mouse model must target both alleles of the Crb1 and Crb2 genes in the optic vesicle or earlier.
    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
    哺乳动物肾脏由30多种不同类型的细胞组成。具有挑战性的任务是鉴定和表征在胚胎和新生儿肾脏中的肾发生期间以及在成熟肾脏中的组织稳态和/或损伤修复期间在这些细胞元件之间建立谱系关系的干/祖细胞亚群。此外,干/祖细胞的潜在临床效用为开发新的治疗肾脏疾病的再生医学方法提供了希望。干细胞由无限的自我更新能力和多潜能定义。祖细胞具有多能性,但没有或有限的自我更新潜力。基于Cre-LoxP的体内遗传谱系追踪是在其天然环境中鉴定干/祖细胞的有力工具。假设,这项技术使研究人员能够准确地追踪单个细胞或一组细胞的后代。Cre/LoxP系统已广泛用于揭示各种哺乳动物组织中基因的功能,并通过体内谱系追踪分析鉴定干/祖细胞。在这次审查中,我们总结了各种Cre驱动因子的开发和表征以及它们在鉴定发育中和成熟小鼠肾脏中潜在的肾脏干/祖细胞方面的最新进展。
    Mammalian kidneys consist of more than 30 different types of cells. A challenging task is to identify and characterize the stem/progenitor subpopulations that establish the lineage relationships among these cellular elements during nephrogenesis in the embryonic and neonate kidneys and during tissue homeostasis and/or injury repair in the mature kidney. Moreover, the potential clinical utility of stem/progenitor cells holds promise for the development of new regenerative medicine approaches for the treatment of renal diseases. Stem cells are defined by unlimited self-renewal capacity and pluripotentiality. Progenitor cells have pluripotentiality but no or limited self-renewal potential. Cre-LoxP-based in vivo genetic lineage tracing is a powerful tool to identify stem/progenitor cells in their native environment. Hypothetically, this technique enables investigators to accurately track the progeny of a single cell or a group of cells. The Cre/LoxP system has been widely used to uncover the function of genes in various mammalian tissues and to identify stem/progenitor cells through in vivo lineage tracing analyses. In this review, we summarize the recent advances in the development and characterization of various Cre drivers and their use in identifying potential renal stem/progenitor cells in both developing and mature mouse kidneys.
    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
    The use of Chlamydomonas for biofuel and biopharmaceutical production has been anticipated. However, the genetic engineering technology for Chlamydomonas is not as advanced as that for other organisms. Here, we established transgenic Chlamydomonas strains capable of high and stable transgene expression. The established cells exhibited stable reporter gene expression at a high level throughout long-term culture (∼60 days), even in the absence of drug pressure. The transgene insertion sites in the cell genome that may be suitable for exogenous gene expression were identified. Because the transgene contains a loxP site, the cells can be used as founders for retargeting other transgenes using the Cre-loxP system to generate transgenic Chlamydomonas producing useful substances. As a model biopharmaceutical gene, an interferon expression cassette was integrated into the genomic locus of the cells using Cre recombinase. The transgenic cells stably produced interferon protein in medium for 12 passages under non-selective conditions. These results indicate that the Chlamydomonas cells established in this study can serve as valuable and powerful tools not only for basic research on microalgae but also for the rapid establishment of cell lines expressing exogenous genes.
    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
    在老鼠身上,使用Cre-loxP系统的条件敲除策略可用于各种类型的研究。具有孕激素受体启动子的Cre小鼠系(PgrCre)已被广泛用于生产特定的子宫基因缺陷小鼠,但在Cre线上,内源性Pgr基因被Cre重组酶基因取代,这使得纯合小鼠(PgrCre/Cre)的繁殖变得困难,因为它们是不育的。Yangetal.(2013,https://10.1016/j。cell.2013.04.017)报道了另一个PrriresCre小鼠品系的产生,该品系仍然具有内源性Pgr基因,他们通过内部核糖体进入位点(IRES)将Cre重组酶插入Pgr基因的下游。这种新的PrriresCre系可能对子宫研究有用,因为小鼠可以作为纯合子(PrriresCre/iresCre)进行繁殖。在这里,我们证实了PrriresCre小鼠在雌性生殖道中有效地定向重组,并且能够在子宫内膜中进行遗传改变,从而能够研究其子宫功能.我们的发现表明,新的PrriresCre小鼠品系也可用于产生子宫特异性敲除小鼠。使用PrriresCre小鼠的发现将有助于理解人类和家畜的生殖系统和疾病。
    In mice, the conditional knockout strategy using the Cre-loxP system is useful for various types of research. The Cre mouse line with progesterone receptor promoter (PgrCre ) has been widely used to produce specific uterine gene-deficient mice, but in the Cre line, endogenous Pgr gene is replaced by Cre recombinase gene, which makes the breeding of homozygous mice (PgrCre/Cre ) difficult because they are infertile. Yang et al. (2013, https://10.1016/j.cell.2013.04.017) reported the generation of another PgriresCre mouse line that still has endogenous Pgr gene, and they inserted Cre recombinase downstream of the Pgr gene via an internal ribosome entry site (IRES). It is possible that this new PgriresCre line would be useful for uterine research as the mice can be bred as homozygotes (PgriresCre/iresCre ). Herein, we confirmed the PgriresCre mice effectively directed recombination in the female reproductive tract and was capable of genetic alteration in the endometrium that enables the studies of its uterine function. Our findings demonstrate that the new PgriresCre mouse line is also useful for the generation of uterine-specific knockout mice. The findings using PgriresCre mouse will contribute to the understanding of reproductive systems and diseases in humans and domestic animals.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号