[go: up one dir, main page]

CN116064597B - Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA - Google Patents

Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA Download PDF

Info

Publication number
CN116064597B
CN116064597B CN202310078321.2A CN202310078321A CN116064597B CN 116064597 B CN116064597 B CN 116064597B CN 202310078321 A CN202310078321 A CN 202310078321A CN 116064597 B CN116064597 B CN 116064597B
Authority
CN
China
Prior art keywords
rna
host cell
replicase
replace
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202310078321.2A
Other languages
Chinese (zh)
Other versions
CN116064597A (en
Inventor
马良
林一瀚
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Peking University
Original Assignee
Peking University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Peking University filed Critical Peking University
Priority to CN202310078321.2A priority Critical patent/CN116064597B/en
Publication of CN116064597A publication Critical patent/CN116064597A/en
Priority to PCT/CN2024/072737 priority patent/WO2024153118A1/en
Application granted granted Critical
Publication of CN116064597B publication Critical patent/CN116064597B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Plant Pathology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to an orthogonal alphavirus RNA replication system REPLACE system to evolve RNA-based elements, enabling sustained in vivo evolution of RNA replicase-assisted mammalian cells. The method of the present invention is capable of continuous diversification and selection of over 10 billion autonomously replicating RNA copies, RNA replication by replicase restriction patterns and powerful inducible RNA mutations. The present invention evolves new functions of fluorescent proteins, transcription factors, and mini-Cas proteins (dCasMINI) or improves upon existing functions. The present invention shows that cells equipped with replacement can adapt to challenges outside or inside the cell by continually evolving autonomously replicating RNAs that carry the key genes associated with cancer (i.e., MEK1 and KRAS). The novel RNA-based evolution platform of the invention provides a novel high-performance tool box for mammalian synthetic biology and facilitates the adaptive engineering of mammalian cells and tissues.

Description

通过自主复制RNA实现哺乳动物细胞中的定向进化和达尔文 适应Directed evolution and Darwinian adaptation in mammalian cells via autonomously replicating RNA

技术领域Technical Field

本发明涉及生物工程领域,具体涉及通过自主复制RNA实现哺乳动物细胞中的定向进化和达尔文适应。The present invention relates to the field of bioengineering, and in particular to directed evolution and Darwinian adaptation in mammalian cells achieved through autonomous replication of RNA.

背景技术Background Art

虽然哺乳动物合成生物学为解决人类健康问题带来了希望7–9,21–24,但它仍然需要克服两个主要障碍。第一,由于通常情况下基因序列到功能的对应关系的复杂性,使得为指定功能来优化合成装置是非常费力的7–9。第二,预设好的合成装置不仅难以无限地适应不断变化的环境,而且可能出现导致功能丧失的突变6,8,10,25,26。而合成装置在哺乳动物细胞内的定向进化可以帮助应对这些挑战:一方面,通过定向进化可以加速合成装置的构建,也即使基因或基因线路多样化(例如,通过诱导突变)再进行筛选27–30;另一方面,合成装置进化的能力,特别是在活细胞内连续的进化,将使得细胞进化并适应环境变化,或逆转合成装置出现的有害变异10,25,26Although mammalian synthetic biology holds promise for addressing human health issues7–9,21–24 , it still needs to overcome two major obstacles. First, due to the complexity of the correspondence between gene sequence and function, it is very laborious to optimize the synthetic machinery for a given function7–9 . Second, the pre-set synthetic machinery is not only difficult to adapt to the changing environment indefinitely, but also may undergo mutations that lead to loss of function6,8,10,25,26. The directed evolution of synthetic machinery in mammalian cells can help address these challenges: on the one hand, directed evolution can accelerate the construction of synthetic machinery, that is, diversify genes or gene circuits (for example, by inducing mutations) and then screen them27–30 ; on the other hand, the ability to evolve synthetic machinery, especially continuous evolution in living cells, will enable cells to evolve and adapt to environmental changes, or reverse harmful mutations in synthetic machinery10,25,26 .

正如以酵母为载体的定向进化方法OrthoRep那样31,一个理想的基于哺乳动物细胞的定向进化方法也应该将合成装置与宿主基因组隔离,以确保连续的、正交的和长期的进化。它还需要克服低效率的转染以及哺乳动物细胞增殖缓慢1而难以在哺乳动物细胞内形成大规模的突变体库的困难。进一步地,相对广泛的突变谱对于形成合成装置完备的序列-功能映射图谱是必要的,同时也将提高该方法的性能1,3。此外,实现对突变速度的可调节控制是非常有益的3,因为这将通过调整诱变时间和诱变程度以适用各类应用场景。然而现有的方法1,2,11–19,并不能满足上述所有需求。As with the yeast-based directed evolution method OrthoRep, 31 an ideal mammalian-based directed evolution method should isolate the synthetic machinery from the host genome to ensure continuous, orthogonal, and long-term evolution. It also needs to overcome the difficulties of inefficient transfection and slow proliferation of mammalian cells1 , which make it difficult to generate large mutant libraries in mammalian cells. Furthermore, a relatively broad mutation spectrum is necessary to form a complete sequence-function map of the synthetic machinery, which will also improve the performance of the method1,3 . In addition, it is very beneficial to achieve adjustable control over the mutation rate3 , because this will enable the mutagenesis time and degree to be adjusted to suit various application scenarios. However, existing methods1,2,11–19 do not meet all of the above requirements.

发明内容Summary of the invention

不同于进化基于DNA的装置的通常方法,出于对多方面因素的考量,本发明人选择在哺乳动物细胞内进化RNA。第一,基于RNA的治疗方法的前景非常乐观:由于其进入哺乳动物的细胞和组织中的便捷性,以及极低的对于宿主基因组的干扰32,33,RNA可以正交地进行进化。第二,哺乳动物细胞可以稳定地承载许多外来RNA的拷贝34,提供了一种大型变体库的构建并在其中筛选的方式。第三,RNA可以通过RNA病毒的复制酶在哺乳动物细胞内进行复制35,36,并且其复制的精确性可以被调控。通过对于上述RNA特点的考量,本发明人在此开发了REPLACE(RNA replicase-assisted continuous in vivo evolution)技术,一种基于自主复制RNA的用哺乳动物细胞作为底盘细胞的定向进化技术。它能确保正交性,构建相对大规模的突变体库,提供一个相对平衡和广泛的突变谱,允许控制诱变率,并以连续或非连续模式运行。本发明人在此展示了REPLACE的一系列应用场景。它将成为一个通用的、强大的合成生物学工具,用于直接适用于哺乳动物的生物大分子工程改造以及在哺乳动物细胞和组织中提供合成进化的能力。Different from the usual method of evolving DNA-based devices, the inventors chose to evolve RNA in mammalian cells for a number of reasons. First, the prospects for RNA-based therapeutic approaches are very promising: RNA can be evolved orthogonally due to its ease of entry into mammalian cells and tissues and minimal interference with the host genome32,33 . Second, mammalian cells can stably carry many copies of foreign RNA34 , providing a way to construct and screen large variant libraries. Third, RNA can be replicated in mammalian cells by RNA viral replicases35,36 , and the accuracy of its replication can be regulated. By considering the above characteristics of RNA, the inventors here developed REPLACE ( RNA replicated continuous in vivo evolution ) technology, a directed evolution technology based on autonomously replicating RNA using mammalian cells as chassis cells. It can ensure orthogonality, construct a relatively large mutant library, provide a relatively balanced and broad mutation spectrum, allow control of the mutagenesis rate, and operate in continuous or discontinuous mode. The inventors here demonstrate a series of application scenarios of REPLACE, which will become a general and powerful synthetic biology tool for directly applicable mammalian biomacromolecule engineering and providing synthetic evolution capabilities in mammalian cells and tissues.

一方面,本发明提供了一种RNA复制酶辅助的连续体内进化(REPLACE)系统,其包含自主复制RNA和宿主细胞,任选地还包含诱导突变的诱变剂。In one aspect, the present invention provides an RNA replicase-assisted continuous in vivo evolution (REPLACE) system, which comprises an autonomously replicating RNA and a host cell, and optionally further comprises a mutagen that induces mutations.

在一个实施方案中,所述自主复制RNA缺失RNA复制酶,并且所述宿主细胞组成型表达所述缺失的RNA复制酶。In one embodiment, the autonomously replicating RNA lacks RNA replicase, and the host cell constitutively expresses the lacked RNA replicase.

在另一个实施方案中,所述自主复制RNA衍生自正链RNA病毒,优选地衍生自甲病毒,更优选地衍生自Sindbis病毒,更优选地衍生自repRNA-v1,更优选地衍生自repRNA-v2。In another embodiment, the autonomously replicating RNA is derived from a positive-strand RNA virus, preferably from an alphavirus, more preferably from a Sindbis virus, more preferably from repRNA-v1, more preferably from repRNA-v2.

在又一个实施方案中,所述自主复制RNA选自repRNA-v3、repRNA-v4中的任一种或衍生自repRNA-v3、repRNA-v4中的任一种,优选地所述自主复制RNA为repRNA-v4或衍生自repRNA-v4,更优选地所述自主复制RNA为repRNA-v4。In another embodiment, the autonomously replicating RNA is selected from any one of repRNA-v3, repRNA-v4 or is derived from any one of repRNA-v3, repRNA-v4, preferably the autonomously replicating RNA is repRNA-v4 or is derived from repRNA-v4, more preferably the autonomously replicating RNA is repRNA-v4.

在另一个实施方案中,所述宿主细胞为真核生物宿主细胞,优选地所述宿主细胞为动物宿主细胞,更优选地所述宿主细胞为哺乳动物宿主细胞,更优选地所述宿主细胞选自仓鼠成纤维细胞系或衍生自仓鼠成纤维细胞系,更优选地所述宿主细胞选自BHK-21(ATCC)或衍生自BHK-21(ATCC),更优选地所述宿主细胞为复制酶有限的真核生物宿主细胞,优选地所述宿主细胞为复制酶有限的动物宿主细胞,更优选地所述宿主细胞为复制酶有限的哺乳动物宿主细胞,更优选地所述宿主细胞选自复制酶有限的仓鼠成纤维细胞系或衍生自复制酶有限的仓鼠成纤维细胞系,更优选地所述宿主细胞选自复制酶有限的BHK-21(ATCC)或衍生自复制酶有限的BHK-21(ATCC)。In another embodiment, the host cell is a eukaryotic host cell, preferably the host cell is an animal host cell, more preferably the host cell is a mammalian host cell, more preferably the host cell is selected from a hamster fibroblast cell line or derived from a hamster fibroblast cell line, more preferably the host cell is selected from BHK-21 (ATCC) or derived from BHK-21 (ATCC), more preferably the host cell is a replicase-limited eukaryotic host cell, preferably the host cell is a replicase-limited animal host cell, more preferably the host cell is a replicase-limited mammalian host cell, more preferably the host cell is selected from a replicase-limited hamster fibroblast cell line or derived from a replicase-limited hamster fibroblast cell line, more preferably the host cell is selected from a replicase-limited BHK-21 (ATCC) or derived from a replicase-limited BHK-21 (ATCC).

在又一个实施方案中,所述诱变剂选自小分子诱变剂、核苷类似物中的任一种,任选地所述诱变剂选自favipiravir、molnupiravir中的任一种。In yet another embodiment, the mutagen is selected from any one of a small molecule mutagen, a nucleoside analog, and optionally the mutagen is selected from any one of favipiravir, molnupiravir.

在另一方面,本发明提供一种RNA复制酶辅助的连续体内进化(REPLACE)方法,其包括:提供自主复制RNA;提供宿主细胞;使自主复制RNA连续体内进化;任选地还提供诱导突变的诱变剂。In another aspect, the present invention provides an RNA replicase-assisted continuous in vivo evolution (REPLACE) method, comprising: providing autonomously replicating RNA; providing a host cell; allowing the autonomously replicating RNA to continuously evolve in vivo; and optionally providing a mutagen that induces mutation.

在一个实施方案中,所述自主复制RNA缺失RNA复制酶,并且所述宿主细胞组成型表达所述缺失的RNA复制酶。In one embodiment, the autonomously replicating RNA lacks RNA replicase, and the host cell constitutively expresses the lacked RNA replicase.

在另一个实施方案中,所述自主复制RNA衍生自正链RNA病毒,优选地衍生自甲病毒,更优选地衍生自Sindbis病毒,更优选地衍生自repRNA-v1,更优选地衍生自repRNA-v2。In another embodiment, the autonomously replicating RNA is derived from a positive-strand RNA virus, preferably from an alphavirus, more preferably from a Sindbis virus, more preferably from repRNA-v1, more preferably from repRNA-v2.

在又一个实施方案中,所述自主复制RNA选自repRNA-v3、repRNA-v4中的任一种或衍生自repRNA-v3、repRNA-v4中的任一种,优选地所述自主复制RNA为repRNA-v4或衍生自repRNA-v4,更优选地所述自主复制RNA为repRNA-v4。In another embodiment, the autonomously replicating RNA is selected from any one of repRNA-v3, repRNA-v4 or is derived from any one of repRNA-v3, repRNA-v4, preferably the autonomously replicating RNA is repRNA-v4 or is derived from repRNA-v4, more preferably the autonomously replicating RNA is repRNA-v4.

在另一个实施方案中,所述宿主细胞为真核生物宿主细胞,优选地所述宿主细胞为动物宿主细胞,更优选地所述宿主细胞为哺乳动物宿主细胞,更优选地所述宿主细胞选自仓鼠成纤维细胞系或衍生自仓鼠成纤维细胞系,更优选地所述宿主细胞选自BHK-21(ATCC)或衍生自BHK-21(ATCC),更优选地所述宿主细胞为复制酶有限的真核生物宿主细胞,优选地所述宿主细胞为复制酶有限的动物宿主细胞,更优选地所述宿主细胞为复制酶有限的哺乳动物宿主细胞,更优选地所述宿主细胞选自复制酶有限的仓鼠成纤维细胞系或衍生自复制酶有限的仓鼠成纤维细胞系,更优选地所述宿主细胞选自复制酶有限的BHK-21(ATCC)或衍生自复制酶有限的BHK-21(ATCC)中的任一种。In another embodiment, the host cell is a eukaryotic host cell, preferably the host cell is an animal host cell, more preferably the host cell is a mammalian host cell, more preferably the host cell is selected from a hamster fibroblast cell line or derived from a hamster fibroblast cell line, more preferably the host cell is selected from BHK-21 (ATCC) or derived from BHK-21 (ATCC), more preferably the host cell is a replicase-limited eukaryotic host cell, preferably the host cell is a replicase-limited animal host cell, more preferably the host cell is a replicase-limited mammalian host cell, more preferably the host cell is selected from a replicase-limited hamster fibroblast cell line or derived from a replicase-limited hamster fibroblast cell line, more preferably the host cell is selected from a replicase-limited BHK-21 (ATCC) or derived from any one of the replicase-limited BHK-21 (ATCC).

在又一个实施方案中,所述诱变剂选自小分子诱变剂、核苷类似物中的任一种,任选地所述诱变剂选自favipiravir、molnupiravir中的任一种。In yet another embodiment, the mutagen is selected from any one of a small molecule mutagen, a nucleoside analog, and optionally the mutagen is selected from any one of favipiravir, molnupiravir.

在又一方面,本发明提供本发明的REPLACE系统在制造生物大分子、疫苗、药物中的用途、在产生突变体库中的用途或者在实现达尔文式适应中的用途。In yet another aspect, the present invention provides the use of the REPLACE system of the present invention in the manufacture of biomacromolecules, vaccines, drugs, in the generation of mutant libraries, or in achieving Darwinian adaptation.

在另一方面,本发明提供一种载体,其用于表达本发明的自主复制RNA。In another aspect, the present invention provides a vector for expressing the autonomously replicating RNA of the present invention.

在又一方面,本发明提供一种宿主细胞,其包含表达载体,所述表达载体用于表达本发明的自主复制RNA。In yet another aspect, the present invention provides a host cell comprising an expression vector for expressing the autonomously replicating RNA of the present invention.

在另一方面,本发明提供一种疫苗组合物,其包含通过或者可通过本发明的REPLACE系统产生的RNA。In another aspect, the invention provides a vaccine composition comprising RNA produced or producible by the REPLACE system of the invention.

在又一方面,本发明提供一种药物组合物,其包含通过或者可通过本发明的REPLACE系统产生的RNA。In yet another aspect, the invention provides a pharmaceutical composition comprising RNA produced or producible by the REPLACE system of the invention.

在另一方面,本发明提供一种递送载体,其包含通过或者可通过本发明的REPLACE系统产生的RNA。In another aspect, the invention provides a delivery vector comprising RNA produced or producible by the REPLACE system of the invention.

本发明的有益效果包括本发明能确保正交性,构建相对大规模的突变体库,提供一个相对平衡和广泛的突变谱,允许控制诱变率,并以连续或非连续模式运行。Advantages of the present invention include that the present invention can ensure orthogonality, construct a relatively large-scale mutant library, provide a relatively balanced and broad mutation spectrum, allow control of the mutagenesis rate, and operate in a continuous or discontinuous mode.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

图1是哺乳动物细胞中自我复制RNA的示意图。这两种自我复制RNA来自于文献42,43,其中的候选RNA变体表现出降低的细胞病理特征(详见本申请的方法)。Figure 1 is a schematic diagram of self-replicating RNA in mammalian cells. These two self-replicating RNAs are from the literature 42,43 , and the candidate RNA variants therein show reduced cytopathic characteristics (see the method of this application for details).

图2是通过流式分析对电穿孔后24小时所表达的EGFP信号进行的量化,以及电穿孔后10天对指定的自主复制RNA的细胞数量进行量化(在10μg/ml嘌呤霉素的选择下)。P值来自双尾t检验,误差条表示标准差(n=3次生物学重复)。Figure 2 is a quantification of the EGFP signal expressed 24 hours after electroporation by flow cytometry, and the number of cells expressing the indicated autonomously replicating RNAs 10 days after electroporation (under selection with 10 μg/ml puromycin). P values are from two-tailed t-tests, and error bars represent standard deviations (n=3 biological replicates).

图3是哺乳动物细胞中自我复制的RNA的文库筛选实验。将加帽和尾部带有多聚腺苷酸的RNA文库(基于repRNA-v2)电穿孔到野生型BHK-21细胞中。nsP1和nsP2的倒数第二位的氨基酸由NNK密码子编码。EGFP(左)和明场(右)的代表图像展示电穿孔后7天的细胞。Figure 3 is a library screening experiment of self-replicating RNA in mammalian cells. The RNA library with caps and tails containing polyadenylic acid (based on repRNA-v2) was electroporated into wild-type BHK-21 cells. The penultimate amino acid of nsP1 and nsP2 is encoded by the NNK codon. Representative images of EGFP (left) and bright field (right) show cells 7 days after electroporation.

图4是对图3中筛选前后RNA文库的每个NNK密码子对应的氨基酸的相对比例进行了量化。误差条表示标准差(n=3次生物学重复)。Figure 4 quantifies the relative proportion of amino acids corresponding to each NNK codon in the RNA library before and after screening in Figure 3. Error bars represent standard deviations (n=3 biological replicates).

图5是在哺乳动物细胞中具有增强宿主兼容性的自主复制RNA的构建和表征。a,在哺乳动物细胞中设计病毒自主复制RNA的文库筛选实验。在表达nsP4的细胞中复制含有nsP1-3和一个由亚基因组(SG)启动子驱动的编码框的RNA。nsP1和nsP2的倒数第二位的两个氨基酸编码在NNK密码子中。b,代表性的EGFP(顶部)和明场图像(底部)展示文库电穿孔后7天的细胞。Figure 5 is a diagram of the construction and characterization of autonomously replicating RNAs with enhanced host compatibility in mammalian cells. a, Library screening experiments of designed viral autonomously replicating RNAs in mammalian cells. RNA containing nsP1-3 and a coding frame driven by a subgenomic (SG) promoter was replicated in cells expressing nsP4. The penultimate two amino acids of nsP1 and nsP2 are encoded in NNK codons. b, Representative EGFP (top) and bright field images (bottom) show cells 7 days after library electroporation.

图6是对两个RNA文库筛选后7天的细胞数进行量化(库1指图3-图4,库2指图5和图7)。P值来自双尾t检验,误差条表示标准差(n=3次生物学重复)。Figure 6 is a quantification of cell number 7 days after screening of two RNA libraries (library 1 refers to Figures 3-4, and library 2 refers to Figures 5 and 7). P values are from two-tailed t-tests, and error bars represent standard deviations (n=3 biological replicates).

图7是对筛选前后的突变体文库中每个NNK密码子的单个氨基酸的相对比例进行量化。误差条表示标准差(n=4个生物学重复)。Figure 7 quantifies the relative proportions of individual amino acids at each NNK codon in the mutant library before and after screening. Error bars represent standard deviation (n=4 biological replicates).

图8是三个版本的自主复制RNA及其相应的EGFP(顶部)和明场(底部)的代表性图片,分别在电穿孔进入宿主细胞后1天和7天拍摄。FIG8 shows representative images of three versions of self-replicating RNA and their corresponding EGFP (top) and bright field (bottom), taken 1 day and 7 days after electroporation into host cells.

图9是电穿孔7天后,不同的RNA版本量化得到的细胞数。P值来自双尾t检验,误差条表示标准差(n=3个生物学重复)。Figure 9 shows the cell numbers quantified by different RNA versions 7 days after electroporation. P values are from two-tailed t-tests, and error bars represent standard deviations (n=3 biological replicates).

图10是通过测量RNA在进入细胞的早期时间点表达的EGFP来量化和比较RNA的短期稳定性的试验设计。FIG. 10 is an experimental design for quantifying and comparing short-term stability of RNA by measuring EGFP expression at early time points after RNA entry into cells.

图11是在指定的时间点,用不同版本的RNA电穿孔的细胞的EGFP和iRFP荧光的流式细胞仪定量分析。注意只有repRNA-v4的宿主细胞表达iRFP信号(即表示复制酶水平)。Figure 11 is a flow cytometric quantitative analysis of EGFP and iRFP fluorescence in cells electroporated with different versions of RNA at the indicated time points. Note that only cells hosting repRNA-v4 expressed iRFP signals (i.e., indicating replicase levels).

图12是两种版本的自主复制RNA在3天内对EGFP信号的量化(包括EGFP阳性细胞的比例和EGFP阳性细胞的平均EGFP强度)(repRNA-v2;repRNA-v3)。n=2次生物学重复。Figure 12 is the quantification of EGFP signals (including the proportion of EGFP-positive cells and the average EGFP intensity of EGFP-positive cells) by two versions of autonomously replicating RNA over 3 days (repRNA-v2; repRNA-v3). n=2 biological replicates.

图13是一种版本的自主复制RNA在3天内对EGFP信号的量化(包括EGFP阳性细胞的比例和EGFP阳性细胞的平均EGFP强度)(repRNA-v4)。将具有不同iRFP信号水平的细胞分别进行了分选和量化,以比较其在不同复制酶水平下的行为。n=2次生物学重复。Figure 13 is a version of the self-replicating RNA quantification of EGFP signal over 3 days (including the proportion of EGFP-positive cells and the average EGFP intensity of EGFP-positive cells) (repRNA-v4). Cells with different iRFP signal levels were sorted and quantified to compare their behavior under different replicase levels. n = 2 biological replicates.

图14是与携带repRNA-v4的细胞相比,携带repRNA-v3的细胞中上调的基因的通路富集分析(详见方法)。FIG14 is a pathway enrichment analysis of genes upregulated in cells carrying repRNA-v3 compared to cells carrying repRNA-v4 (see Methods for details).

图15是短期RNA稳定性通过电穿孔后36小时和72小时EGFP阳性部分的相对变化进行量化。repRNA-v4在两个宿主中进行量化,其中任一宿主的细胞按复制酶水平(iRFP)进行分群,每个群体都分别计算EGFP阳性细胞的比例。Figure 15 shows short-term RNA stability quantified by relative changes in the EGFP-positive fraction at 36 and 72 hours after electroporation. repRNA-v4 was quantified in two hosts, where cells from either host were grouped by replicase level (iRFP) and the proportion of EGFP-positive cells was calculated for each population.

图16是携带repRNA-v4的低复制酶宿主示意图。与高复制酶宿主相比(图5a、图10和详见方法),低复制酶宿主通过在nsP4基因前面插入的一个终止密码子大大降低了其表达。Figure 16 is a schematic diagram of a low replicase host carrying repRNA-v4. Compared with the high replicase host (Figure 5a, Figure 10 and see Methods for details), the low replicase host greatly reduces its expression by inserting a stop codon in front of the nsP4 gene.

图17是3天内携带repRNA-v4的低复制酶宿主细胞的EGFP信号的量化(包括EGFP阳性细胞的比例和EGFP阳性细胞的平均EGFP强度)(如图10所示)。对具有不同iRFP信号水平的细胞分别进行分群和量化,以比较不同复制酶水平下的行为,数据也被用于图15。Figure 17 is a quantification of the EGFP signal of low replicase host cells carrying repRNA-v4 within 3 days (including the proportion of EGFP-positive cells and the average EGFP intensity of EGFP-positive cells) (as shown in Figure 10). Cells with different iRFP signal levels were grouped and quantified to compare the behavior under different replicase levels, and the data were also used in Figure 15.

图18是散点图显示图15中稳态EGFP水平和各个细胞群的相应复制酶水平。红线表示线性拟合。Figure 18 is a scatter plot showing the steady-state EGFP levels and the corresponding replicase levels of each cell population in Figure 15. The red line represents the linear fit.

图19是利用FACS对有无repRNA-v4的高复制酶宿主细胞的EGFP和iRFP信号进行量化的结果(电穿孔后3周)和其中数据的平均iRFP信号水平(即复制酶水平的代表),误差条表示标准差(n=3次生物学重复)。Figure 19 shows the results of quantification of EGFP and iRFP signals of high replicase host cells with and without repRNA-v4 using FACS (3 weeks after electroporation) and the average iRFP signal level of the data (i.e., a representative of the replicase level), with error bars representing standard deviations (n=3 biological replicates).

图20是与图19类似的低复制酶宿主的定量和其中数据的平均iRFP信号水平,误差条表示标准差(n=3次生物学重复)。FIG. 20 is a quantification of low replicase hosts similar to FIG. 19 and the mean iRFP signal levels of the data therein, with error bars representing standard deviation (n=3 biological replicates).

图21是使用培养一段时间后已经稳定的细胞群体(电穿孔后2周),在一个月的培养过程中,由EGFP阳性部分的变化来量化长期的RNA稳定性(n=3个生物学重复)。FIG. 21 quantifies long-term RNA stability by changes in the EGFP-positive fraction during one month of culture using a cell population that has stabilized after a period of culture (2 weeks after electroporation) (n=3 biological replicates).

图22是利用流式对携带repRNA-v4的高复制酶宿主细胞的EGFP和iRFP信号进行的一个月时间尺度上的量化。数据被量化并呈现在图21中。Figure 22 is a flow cytometry quantification of EGFP and iRFP signals in high replicase host cells carrying repRNA-v4 over a one month time scale. The data are quantified and presented in Figure 21.

图23是代表性的图像显示有无repRNA-v4的宿主细胞的dsRNA信号(红色,免疫染色的信号)与细胞核信号(蓝色,Hoechst)的叠加图像。FIG. 23 is a representative image showing the superposition of dsRNA signals (red, immunostaining signals) and nuclear signals (blue, Hoechst) of host cells with and without repRNA-v4.

图24是通过箱形图显示的图23(详见方法)中免疫染色实验的dsRNA信号的量化。FIG. 24 is a quantification of dsRNA signals from the immunostaining experiments in FIG. 23 (see Methods for details) shown by box plots.

图25是通过对有无repRNA-v4的宿主细胞进行RNA测序来对Gapdh和nsP1-nsP3的RPKM的量化结果(n=2个生物学重复)。FIG. 25 shows the quantification results of the RPKM of Gapdh and nsP1-nsP3 by RNA sequencing of host cells with and without repRNA-v4 (n=2 biological replicates).

图26是通过使用核苷类似物调节RNA复制酶的保真度来实现可控的RNA诱变。a,在没有扰动的情况下(即基础情况下),对复制RNA进行突变分析的试验设计。b,质粒DNA或自主复制RNA上的目标区域(nsP1-3)在电穿孔后指定时间点的总突变频率。误差条表示标准差(n=3个生物学重复)。Figure 26 is a controlled RNA mutagenesis achieved by regulating the fidelity of RNA replicase using nucleoside analogs. a, Experimental design for mutation analysis of replicating RNA in the absence of perturbations (i.e., basal conditions). b, Total mutation frequency of target regions (nsP1-3) on plasmid DNA or autonomously replicating RNA at indicated time points after electroporation. Error bars represent standard deviations (n = 3 biological replicates).

图27是散点图显示总突变频率与在宿主细胞中复制的天数的关系。数据来自图26的b。星号表示3次实验的平均值。红线表示对数据的线性拟合。Figure 27 is a scatter plot showing the relationship between total mutation frequency and the number of days replicated in host cells. Data are from b of Figure 26. Asterisks represent the average of 3 experiments. The red line represents the linear fit to the data.

图28是IVT RNAs和电穿孔后21天的细胞的自主复制RNA的突变谱。误差条表示标准差(n=3个生物学重复)。p值由双尾t检验计算(*p<0.05,**p<0.01,和***p<0.001)。Figure 28 is the mutation spectrum of IVT RNAs and autonomously replicating RNAs of cells 21 days after electroporation. Error bars represent standard deviation (n=3 biological replicates). p values were calculated by two-tailed t-test (*p<0.05, **p<0.01, and ***p<0.001).

图29是nsP4 C488G突变体突变能力的表征。左侧图,野生型nsP4和nsP4 C488G突变体电穿孔后10天的目标区域(nsP1-3)总突变频率的比较。右侧图,突变谱的比较。P值通过双尾t检验计算,误差条表示标准差(n=3次生物学重复)。Figure 29 is a characterization of the mutation capacity of the nsP4 C488G mutant. Left panel, comparison of the total mutation frequency of the target region (nsP1-3) 10 days after electroporation of wild-type nsP4 and nsP4 C488G mutant. Right panel, comparison of mutation spectra. P values were calculated by two-tailed t-test, and error bars represent standard deviations (n = 3 biological replicates).

图30是用指定浓度ribavirin和5-AzaC处理72小时后的EGFP荧光水平。误差条表示标准差(n=3次生物学重复)。Figure 30 shows the EGFP fluorescence level after treatment with the indicated concentrations of ribavirin and 5-AzaC for 72 hours. Error bars represent standard deviation (n=3 biological replicates).

图31是用核苷类似物molnupiravir和favipiravir在指定浓度下处理细胞72小时,FACS量化EGFP的荧光强度。右上图为代表性的图片展示molnupiravir处理后EGFP荧光的减少。误差条表示标准差(n=3个生物学重复)。Figure 31 is a FACS quantification of the fluorescence intensity of EGFP after treating cells with nucleoside analogs molnupiravir and favipiravir at the indicated concentrations for 72 hours. The upper right figure is a representative picture showing the reduction of EGFP fluorescence after molnupiravir treatment. The error bars represent standard deviations (n = 3 biological replicates).

图32是用molnupiravir或favipiravir在指定浓度下处理72小时后,复制RNA上的目标区域(nsP1-3)的总突变频率。FIG. 32 shows the total mutation frequency of the target region (nsP1-3) on the replicating RNA after treatment with molnupiravir or favipiravir at the indicated concentrations for 72 hours.

图33是携带repRNA-v4的细胞在指定浓度的molnupiravir或favipiravir处理72小时后的EGFP信号分布。FIG. 33 is the distribution of EGFP signals in cells carrying repRNA-v4 after treatment with the indicated concentrations of molnupiravir or favipiravir for 72 hours.

图34是携带repRNA-v4的细胞在72小时的时间过程中通过molnupiravir或favipiravir处理后的EGFP信号的分布和相应的条形图。误差条表示标准差(n=3次生物学重复),以及在72小时中,用molnupiravir或favipiravir处理的复制RNA上的目标区域的总突变频率。误差条表示标准差(n=3或6次生物学重复)。Figure 34 is the distribution and corresponding bar graph of the EGFP signal after the cells carrying repRNA-v4 were processed by molnupiravir or favipiravir in the time course of 72 hours. Error bars represent standard deviation (n=3 biological repeats), and in 72 hours, the total mutation frequency of the target region on the replication RNA processed with molnupiravir or favipiravir. Error bars represent standard deviation (n=3 or 6 biological repeats).

图35是携带repRNA-v4的宿主细胞的iRFP信号在molnupiravir或favipiravir的指定浓度下处理72小时后的分布情况;携带repRNA-v4的宿主细胞在72小时时间尺度上在用molnupiravir或favipiravir处理的iRFP信号的分布;以及在指定浓度molnupiravir或favipiravir处理72小时后,对照细胞系的组成型表达的EGFP信号的量化。误差条表示标准差(n=3次生物学重复)。Figure 35 is the distribution of iRFP signals of host cells carrying repRNA-v4 after treatment with the specified concentration of molnupiravir or favipiravir for 72 hours; distribution of iRFP signals of host cells carrying repRNA-v4 treated with molnupiravir or favipiravir on a 72-hour time scale; and quantification of constitutively expressed EGFP signals of control cell lines after treatment with the specified concentration of molnupiravir or favipiravir for 72 hours. Error bars represent standard deviations (n=3 biological replicates).

图36是molnupiravir或favipiravir诱导的自主复制RNA的突变谱。n=6(DMSOctrl)或n=3(其他条件)生物学重复。p值由双尾t检验计算(*p<0.05,**p<0.01,和***p<0.001)。Figure 36 is a mutation spectrum of autonomously replicating RNA induced by molnupiravir or favipiravir. n = 6 (DMSOctrl) or n = 3 (other conditions) biological replicates. P values were calculated by two-tailed t-test (*p < 0.05, **p < 0.01, and ***p < 0.001).

图37是携带EGFP的自主复制RNA和相应的宿主细胞的设计。FIG. 37 shows the design of the autonomously replicating RNA carrying EGFP and the corresponding host cells.

图38是蓝移EGFP的进化实验时间线和相应在指定时间点的流式分析的定量;以及2天molnupiravir处理后EGFP mRNA的突变谱。高基线突变频率(如DMSO条件)是由于细胞在实验前被长时间培养所致。误差条表示标准差(n=3次生物学重复)。P值通过双尾t检验计算(*p<0.05,**p<0.01,和***p<0.001)。Figure 38 is the evolution experiment timeline of blue-shifted EGFP and the quantification of the corresponding flow analysis at the specified time point; and the mutation spectrum of EGFP mRNA after 2 days of molnupiravir treatment. The high baseline mutation frequency (such as DMSO conditions) is due to the cells being cultured for a long time before the experiment. Error bars represent standard deviation (n=3 biological replicates). P values are calculated by two-tailed t-test (*p<0.05, **p<0.01, and ***p<0.001).

图39是分选的细胞群中EGFP mRNA的核苷酸水平突变分析。RNA水平和蛋白质水平的突变都被标记出来。分选的细节见图38的中部。Figure 39 is a nucleotide level mutation analysis of EGFP mRNA in the sorted cell population. Both RNA level and protein level mutations are marked. The details of the sorting are shown in the middle of Figure 38.

图40是不同细胞群中所述突变的百分比。误差条表示标准差(n=3次技术重复)。Figure 40 shows the percentage of the mutations in different cell populations. Error bars represent standard deviation (n=3 technical replicates).

图41是野生型EGFP和两个进化的突变体(Y66H、T203I)的流式定量分析,展示了三个生物学重复中的一个数据。FIG41 is a flow cytometric analysis of wild-type EGFP and two evolved mutants (Y66H, T203I), showing data from one of three biological replicates.

图42是使用REPLACE改造合成的响应配体的转录因子。a,设计携带TetR-VP48的自主复制RNA和含有nsP4和同源TRE3G报告基因的宿主细胞的设计。注意,这种自主复制RNA类似于repRNA-v4,nsP1和nsP2的倒数第二个氨基酸分都是缬氨酸。b,实验设计和TetR-VP48定向进化的时间线。详见方法。Figure 42 shows the transcription factors that respond to synthetic ligands using REPLACE. a, Design of self-replicating RNA carrying TetR-VP48 and host cells containing nsP4 and homologous TRE3G reporter genes. Note that this self-replicating RNA is similar to repRNA-v4, and the penultimate amino acid of nsP1 and nsP2 is valine. b, Experimental design and timeline of directed evolution of TetR-VP48. See Methods for details.

图43是分选的细胞群(即1%具有最高mCherry信号的细胞)中TetR mRNA的核苷酸水平突变分析。FIG. 43 is a nucleotide level mutation analysis of TetR mRNA in a sorted cell population (ie, 1% of cells with the highest mCherry signal).

图44是野生型TetR-VP48和进化突变体在指定的多西环素(Dox)浓度下激活的报告基因的荧光强度。蓝色表示有配体和无配体条件下的报告系统信号的倍数变化。误差条表示标准差(n=3个生物学重复)。Figure 44 shows the fluorescence intensity of the reporter gene activated by wild-type TetR-VP48 and evolved mutants at the indicated doxycycline (Dox) concentrations. Blue indicates the fold change of the reporter system signal under ligand and ligand-free conditions. Error bars indicate standard deviation (n=3 biological replicates).

图45是携带VP64-PadR的自主复制RNA的设计,以及含有nsP4和由含有PadR结合位点的启动子驱动的报告基因的宿主细胞的设计;以及实验设计和VP64-PadR定向进化的时间线。详见方法。Figure 45 shows the design of an autonomously replicating RNA carrying VP64-PadR, and the design of a host cell containing nsP4 and a reporter gene driven by a promoter containing a PadR binding site; and a timeline for the experimental design and directed evolution of VP64-PadR. See Methods for details.

图46是获得的细胞群中PadR mRNA的核苷酸水平突变分析(无分选)。FIG. 46 is a nucleotide level mutation analysis of PadR mRNA in the obtained cell population (without sorting).

图47是野生型VP64-PadR和进化的突变体在指定阿魏酸钠浓度下激活的报告基因的荧光强度。蓝色表示有配体和无配体条件下的报告系统荧光信号的倍数变化。误差条表示标准差(n=3-6个生物学重复)。Figure 47 is the fluorescence intensity of the reporter gene activated by wild-type VP64-PadR and evolved mutants at the specified sodium ferulate concentration. Blue represents the fold change of the reporter system fluorescence signal under ligand and ligand-free conditions. Error bars represent standard deviations (n=3-6 biological replicates).

图48是在指定的阿魏酸钠浓度下,由野生型VP64-PadR和进化突变体激活的报告基因的荧光强度。所选择的突变体是特别有意义的,因为它与野生型相比表现出相反的配体敏感性,这在图47中也得到了量化。误差条表示标准差(n=3次生物学重复)。Figure 48 is the fluorescence intensity of the reporter gene activated by wild-type VP64-PadR and the evolved mutants at the indicated sodium ferulate concentrations. The selected mutant is of particular interest because it exhibits an opposite ligand sensitivity compared to the wild type, which is also quantified in Figure 47. Error bars represent standard deviations (n=3 biological replicates).

图49是实验设计和VP64-PadR在没有配体的情况下定向进化的时间线(即无配体进化)。在这个实验中,使用了图45中的自主复制RNA和宿主细胞。详见方法。Figure 49 is a timeline of the experimental design and directed evolution of VP64-PadR in the absence of a ligand (i.e., ligand-free evolution). In this experiment, the self-replicating RNA and host cells of Figure 45 were used. See Methods for details.

图50是在图49中无配体进化实验中,流式分群所得PadR mRNA的核苷酸水平突变分析。FIG. 50 is a nucleotide level mutation analysis of PadR mRNA obtained by flow cytometry in the ligand-free evolution experiment in FIG. 49 .

图51是野生型VP64-PadR和进化的突变体(来自无配体进化实验)在所示阿魏酸钠浓度下激活的报告基因的荧光强度。每个变体的报告基因表达倍数变化用蓝色表示。误差条表示标准差(n=3次生物学重复)。P值通过双尾t检验计算(*p<0.05,**p<0.01,和***p<0.001)。Figure 51 is the fluorescence intensity of the reporter gene activated by wild-type VP64-PadR and evolved mutants (from ligand-free evolution experiments) at the indicated sodium ferulate concentrations. The reporter gene expression fold change for each variant is indicated in blue. Error bars represent standard deviations (n=3 biological replicates). P values were calculated by two-tailed t-test (*p<0.05, **p<0.01, and ***p<0.001).

图52是野生型VP64-PadR和VP64-PadR(D21G/D74V/M178T)突变体的配体剂量依赖性以及拟合所得的剂量反应曲线。每个材料的拟合所得的希尔系数和半抑制浓度(IC50)都被标明。误差条表示标准差(n=3次生物学重复)。Figure 52 shows the ligand dose dependence and fitted dose response curves of wild-type VP64-PadR and VP64-PadR (D21G/D74V/M178T) mutants. The Hill coefficient and half inhibitory concentration (IC50) obtained by fitting for each material are indicated. Error bars represent standard deviations (n=3 biological replicates).

图53是野生型dCasMINI-VPR和dCasMINI(S246R)-VPR激活的TRE3G报告基因的荧光强度。突变型和野生型的报告水平之间的倍数变化用蓝色表示。误差条表示标准差(n=5个生物学重复)。Figure 53 is the fluorescence intensity of the TRE3G reporter gene activated by wild-type dCasMINI-VPR and dCasMINI (S246R)-VPR. The fold change between the reporter levels of mutant and wild-type is indicated in blue. Error bars represent standard deviation (n = 5 biological replicates).

图54是CD2的定量与h类似(使用抗体染色)。Ctrl表示没有sgRNA对照。误差条表示标准差(n=4个生物学重复)。P值通过双尾t检验计算(*p<0.05,**p<0.01,和***p<0.001)。Figure 54 is the quantification of CD2 similar to h (using antibody staining). Ctrl indicates no sgRNA control. Error bars indicate standard deviation (n=4 biological replicates). P values were calculated by two-tailed t-test (*p<0.05, **p<0.01, and ***p<0.001).

图55是基于野生型dCasMINI或突变型dCasMINI(S246R)编辑后,HEK293T细胞中三个内源性位点的碱基编辑频率。误差条表示标准差(n=4次生物学重复)。P值通过双尾t检验计算(*p<0.05和**p<0.01)。以及CasMINI或突变体CasMINI(S246R)在HEK293T细胞中四个内源性位点的切割频率。误差条表示标准差(n=3次生物学重复)。其中的sgRNA采用的是文献20的sgRNA。Figure 55 is based on the base editing frequency of three endogenous sites in HEK293T cells after editing with wild-type dCasMINI or mutant dCasMINI (S246R). Error bars represent standard deviations (n = 4 biological replicates). P values were calculated by two-tailed t-test (*p<0.05 and **p<0.01). And the cutting frequency of four endogenous sites of CasMINI or mutant CasMINI (S246R) in HEK293T cells. Error bars represent standard deviations (n = 3 biological replicates). The sgRNA used is the sgRNA of literature 20 .

图56是测试携带MEK1的RNA是否能使细胞适应治疗性药物的挑战的实验设计。FIG. 56 is an experimental design for testing whether RNA carrying MEK1 can adapt cells to challenge with therapeutic drugs.

图57是在适应三种不同的MEK1抑制剂的细胞群中,MEK1 mRNA上已确定的突变的总结。每个抑制剂都有三个生物学重复。Figure 57 is a summary of the mutations identified in MEK1 mRNA in cell populations adapted to three different MEK1 inhibitors. Each inhibitor had three biological replicates.

图58是适应三种不同MEK1抑制剂的细胞群中MEK1 mRNA的突变分析。a,REPLACE实验前收集的细胞群中MEK1 mRNA的核苷酸水平的突变分析。b,适应考比替尼挑战的细胞群中MEK1 mRNA的核苷酸水平突变分析。c,适应曲美替尼挑战的细胞群中MEK1 mRNA的核苷酸水平突变分析。d,适应司美替尼挑战的细胞群中MEK1 mRNA的核苷酸水平突变分析。Figure 58 is a mutation analysis of MEK1 mRNA in cell populations adapted to three different MEK1 inhibitors. a, nucleotide level mutation analysis of MEK1 mRNA in cell populations collected before REPLACE experiment. b, nucleotide level mutation analysis of MEK1 mRNA in cell populations adapted to cobimetinib challenge. c, nucleotide level mutation analysis of MEK1 mRNA in cell populations adapted to trametinib challenge. d, nucleotide level mutation analysis of MEK1 mRNA in cell populations adapted to selumetinib challenge.

图59是沿着MEK1的结构域确认的突变总结。DD:对接域;NES:核输出信号;NRR:负调节区;KCD:激酶催化域;AL:激活环;DVD:多功能对接域。关于结构域的注释改编自文献60。Figure 59 is a summary of mutations identified along the domains of MEK1. DD: docking domain; NES: nuclear export signal; NRR: negative regulatory region; KCD: kinase catalytic domain; AL: activation loop; DVD: multifunctional docking domain. The annotations on the domains were adapted from reference 60.

图60是野生型MEK1和进化的突变体在指定条件下激活的SRE报告基因的荧光强度。误差条表示标准差(n=3个生物学重复)。Figure 60 is the fluorescence intensity of SRE reporter gene activated by wild-type MEK1 and evolved mutants under the indicated conditions. Error bars represent standard deviation (n=3 biological replicates).

图61是测试携带KRAS的RNA是否能使细胞适应细胞内在的挑战(即显性抑制的KRAS等位基因)的实验设计。Figure 61 is an experimental design to test whether RNA carrying KRAS can adapt cells to a cell-intrinsic challenge (i.e., a dominant suppressive KRAS allele).

图62是在适应显性抑制的细胞群中,KRAS(S17N)mRNA上确定的突变的总结。为便于比较,图表还包含了使用野生型KRAS的实验的相应突变信息。每个复制的RNA进行了三个生物学重复的实验。Figure 62 is a summary of mutations identified on KRAS(S17N) mRNA in a cell population adapted to dominant inhibition. For ease of comparison, the chart also includes the corresponding mutation information for experiments using wild-type KRAS. Three biological replicates were performed for each replicate RNA.

图63是KRAS适应性实验的其他特征。a,在REPLACE实验前收集的细胞群中KRAS(野生型)mRNA的核苷酸水平突变分析。b,第14天收集的细胞群中KRAS(野生型)mRNA的核苷酸水平突变分析。c,REPLACE实验前收集的细胞群中KRAS(S17N)mRNA的核苷酸水平突变分析。d,逃脱显性抑制的细胞群中KRAS(S17N)mRNA的核苷酸水平突变分析。Figure 63 is additional features of the KRAS adaptive experiment. a, Nucleotide level mutation analysis of KRAS (wild type) mRNA in the cell population collected before the REPLACE experiment. b, Nucleotide level mutation analysis of KRAS (wild type) mRNA in the cell population collected on day 14. c, Nucleotide level mutation analysis of KRAS (S17N) mRNA in the cell population collected before the REPLACE experiment. d, Nucleotide level mutation analysis of KRAS (S17N) mRNA in the cell population that escaped dominant inhibition.

图64是沿着KRAS的结构域确认的突变总结。P-loop:磷酸盐结合环;α3:α螺旋3;HVR:高变异区域。关于结构域的注释改编自文献69。Figure 64 is a summary of mutations identified along the structural domains of KRAS. P-loop: phosphate binding loop; α3: α helix 3; HVR: hypervariable region. The annotations on the structural domains were adapted from 69.

图65是由KRAS变体激活的SRE报告基因的荧光强度(顶部)和通过相对于基线的细胞数量表现的对细胞增殖的相应影响(底部)。误差条表示标准差(n=5或6个生物学重复)。对于(d,h),p值通过双尾t检验计算(*p<0.05,**p<0.01,和***p<0.001)。Figure 65 is the fluorescence intensity of the SRE reporter gene activated by KRAS variants (top) and the corresponding effect on cell proliferation expressed by the number of cells relative to the baseline (bottom). Error bars represent standard deviations (n=5 or 6 biological replicates). For (d, h), p values were calculated by two-tailed t-tests (*p<0.05, **p<0.01, and ***p<0.001).

图66是由KRAS变体激活的SRE报告基因的荧光强度(顶部)和相对于基线的细胞数的对细胞增殖的相应影响(底部)。误差条表示标准差(n=5-6次生物学重复)。P值通过双尾t检验计算(***p<0.001)。Figure 66 is the fluorescence intensity of the SRE reporter gene activated by KRAS variants (top) and the corresponding effect on cell proliferation relative to the number of cells at baseline (bottom). Error bars represent standard deviations (n=5-6 biological replicates). P values were calculated by two-tailed t-test (***p<0.001).

图67是携带dCasMINI-VPR的复制RNA和含有nsP4的宿主细胞的设计,该报告基因由TRE3G启动子驱动,而U6启动子驱动的sgRNA针对TRE3G启动子上的TetO位点。Figure 67 is the design of host cells carrying dCasMINI-VPR replicating RNA and nsP4, where the reporter gene is driven by the TRE3G promoter, and the U6 promoter-driven sgRNA targets the TetO site on the TRE3G promoter.

图68是实验设计和dCasMINI-VPR的定向进化时间线。详见方法。Figure 68 is the experimental design and directed evolution timeline of dCasMINI-VPR. See Methods for details.

图69是收获的细胞群中dCasMINI mRNA的核苷酸水平突变分析(无分选)。FIG. 69 is a nucleotide level mutation analysis of dCasMINI mRNA in harvested cell populations (without sorting).

图70是不同浓度的核苷类似物对细胞增殖的影响,以相对于DMSO处理的细胞数进行量化。误差条表示标准差(n=3次生物学重复)。Figure 70 is the effect of different concentrations of nucleoside analogs on cell proliferation, quantified relative to the number of cells treated with DMSO. Error bars represent standard deviation (n=3 biological replicates).

具体实施方式DETAILED DESCRIPTION

本发明可通过以下实施方案进行实施,但本发明并不限于此。The present invention can be implemented through the following embodiments, but the present invention is not limited thereto.

定义definition

如本文所用,术语“RNA复制酶”是指依赖于RNA的RNA聚合酶,它们以具有特定二级结构的RNA作为模板,对其进行复制。As used herein, the term "RNA replicase" refers to RNA-dependent RNA polymerases that use RNA with a specific secondary structure as a template and replicate it.

如本文所用,术语“自主复制RNA”或“自我复制RNA”是指在RNA复制酶的作用下,能够以自身的RNA序列为模板,进行自我复制的RNA。As used herein, the term "autonomous replicating RNA" or "self-replicating RNA" refers to RNA that can replicate itself using its own RNA sequence as a template under the action of RNA replicase.

如本文所用,术语“宿主细胞”是指能够承载自主复制RNA的底盘细胞。As used herein, the term "host cell" refers to a chassis cell capable of hosting autonomously replicating RNA.

如本文所用,术语“复制酶有限的宿主细胞”是指表达的RNA复制酶量浓度适中,可维持自我复制RNA长期稳定存在的宿主细胞。As used herein, the term "replicase-limited host cell" refers to a host cell that expresses an RNA replicase at a moderate concentration and can maintain the long-term stable existence of self-replicating RNA.

如本文所用,术语“诱变剂”是指能引起细胞的遗传物质发生改变,使其基因突变或染色体畸变达到自然水平以上的物质。As used herein, the term "mutagen" refers to a substance that can cause changes in the genetic material of a cell, causing gene mutations or chromosomal aberrations to exceed natural levels.

如本文所用,术语“小分子诱变剂”是指化学小分子类型的诱变剂。As used herein, the term "small molecule mutagen" refers to a mutagen of the chemical small molecule type.

如本文所用,术语“核苷类似物”是指结构上经过修饰的核苷(核苷酸的只含五碳糖和碱基的部分)。As used herein, the term "nucleoside analog" refers to a structurally modified nucleoside (the portion of a nucleotide containing only a five-carbon sugar and a base).

如本文所用,术语“正链RNA病毒”是指以正链RNA直接起mRNA作用,翻译早期蛋白质,然后在RNA复制酶作用下,以正链RNA为模板,复制形成互补链(负链)的病毒。As used herein, the term "positive-strand RNA virus" refers to a virus in which positive-strand RNA directly acts as mRNA, translates early proteins, and then, under the action of RNA replicase, uses the positive-strand RNA as a template to replicate to form a complementary strand (negative strand).

如本文所用,术语“甲病毒”是指病毒分类学上分类为Alphavirus的一类病毒。As used herein, the term "alphavirus" refers to a type of virus classified as Alphavirus in virus taxonomy.

如本文所用,术语“Sindbis病毒”是指甲病毒属下面的一种称为Sindbis virus的正链RNA病毒。As used herein, the term "Sindbis virus" refers to a positive-strand RNA virus called Sindbis virus under the genus Onychovirus.

如本文所用,术语“repRNA-v1”是指Sindbis病毒的nsP2的726位的脯氨酸替换为亮氨酸,结构蛋白被替换为EGFP-T2A-Puro后得到的复制性RNA。As used herein, the term "repRNA-v1" refers to the replicative RNA obtained after the proline at position 726 of nsP2 of the Sindbis virus is replaced with leucine and the structural protein is replaced with EGFP-T2A-Puro.

如本文所用,术语“repRNA-v2”是指Sindbis病毒的nsP1的539位甘氨酸替换为缬氨酸,nsP2的726位脯氨酸替换为亮氨酸,nsP2的806位甘氨酸替换为缬氨酸,结构蛋白被替换为EGFP-T2A-Puro后得到的复制性RNA。As used herein, the term "repRNA-v2" refers to the replicative RNA obtained after glycine at position 539 of nsP1 of Sindbis virus is replaced by valine, proline at position 726 of nsP2 is replaced by leucine, glycine at position 806 of nsP2 is replaced by valine, and the structural protein is replaced by EGFP-T2A-Puro.

如本文所用,术语“repRNA-v3”是指Sindbis病毒的nsP1的539位甘氨酸替换为丙氨酸,nsP2的726位脯氨酸替换为亮氨酸,nsP2的806位甘氨酸替换为异亮氨酸,结构蛋白被替换为EGFP-T2A-Puro后得到的复制性RNA。As used herein, the term "repRNA-v3" refers to the replicative RNA obtained after glycine at position 539 of nsP1 of Sindbis virus is replaced by alanine, proline at position 726 of nsP2 is replaced by leucine, glycine at position 806 of nsP2 is replaced by isoleucine, and the structural protein is replaced by EGFP-T2A-Puro.

如本文所用,术语“repRNA-v4”是指Sindbis病毒的nsP1的539位甘氨酸替换为丙氨酸,nsP2的726位脯氨酸替换为亮氨酸,nsP2的806位甘氨酸替换为异亮氨酸,nsP4从复制型RNA上删除,然后利用EF1α启动子驱动nsP4表达在宿主细胞中,结构蛋白被替换为EGFP-T2A-Puro后得到的复制性RNA。As used herein, the term "repRNA-v4" refers to the replicative RNA obtained by replacing the glycine at position 539 of nsP1 of the Sindbis virus with alanine, replacing the proline at position 726 of nsP2 with leucine, replacing the glycine at position 806 of nsP2 with isoleucine, deleting nsP4 from the replicative RNA, and then using the EF1α promoter to drive the expression of nsP4 in the host cell, and replacing the structural proteins with EGFP-T2A-Puro.

如本文所用,术语“进化”是指在细胞水平上,群体里的遗传性状在世代之间的变化。As used herein, the term "evolution" refers to changes in inherited traits in a population from generation to generation at the cellular level.

如本文所用,术语“定向进化”是指在实验室中人为制造大量突变,按照特定目的给予特定方向的选择压力,筛选出具有期望特征的蛋白质分子,实现分子水平的模拟进化的过程。As used herein, the term "directed evolution" refers to the process of artificially creating a large number of mutations in a laboratory, applying selection pressure in a specific direction according to a specific purpose, screening out protein molecules with desired characteristics, and realizing simulated evolution at the molecular level.

如本文所用,术语“体内进化”是指定向进化的制造突变的过程全部发生在细胞内。As used herein, the term "in vivo evolution" refers to the process of directed evolutionary mutation making occurring entirely within cells.

如本文所用,术语“连续体内进化”是指上一轮筛选得到的细胞不需要提取遗传物质用于体外诱变,等待细胞扩增后可直接用于下一轮诱变这一实验过程。As used herein, the term "continuous in vivo evolution" refers to an experimental process in which cells obtained from the previous round of screening do not need to extract genetic material for in vitro mutagenesis, but can be directly used for the next round of mutagenesis after waiting for cell expansion.

如本文所用,术语“载体”是指一段核酸物质,它可将遗传物质从一个细胞转移到另一个细胞,用于繁殖、表达或分离等需求。As used herein, the term "vector" refers to a nucleic acid material that can transfer genetic material from one cell to another for the purpose of propagation, expression or separation.

如本文所用,术语“原核载体”是指原核表达载体,能携带插入的外源核酸序列进入原核细胞中进行表达的载体。As used herein, the term "prokaryotic vector" refers to a prokaryotic expression vector, which is a vector that can carry an inserted foreign nucleic acid sequence into prokaryotic cells for expression.

如本文所用,术语“真核载体”是指真核表达载体,这里尤指能携带插入的外源核酸序列进入真核哺乳动物细胞中进行表达的载体。As used herein, the term "eukaryotic vector" refers to a eukaryotic expression vector, and in particular refers to a vector that can carry an inserted foreign nucleic acid sequence into eukaryotic mammalian cells for expression.

如本文所用,术语“病毒载体”是指基于病毒的可以携带插入的外源核酸序列进入哺乳动物细胞进行表达的载体。As used herein, the term "viral vector" refers to a virus-based vector that can carry inserted foreign nucleic acid sequences into mammalian cells for expression.

如本文所用,术语“自我复制型载体”是指可以在哺乳动物宿主细胞内自主进行复制的载体。As used herein, the term "self-replicating vector" refers to a vector that can replicate autonomously in a mammalian host cell.

如本文所用,术语“制造生物大分子”是指利用定向进化对蛋白质、核酸等生物大分子进行工程改造,使其符合特定的需求或使用目的的过程。As used herein, the term "manufacturing biomacromolecules" refers to the process of engineering biomacromolecules such as proteins and nucleic acids using directed evolution to make them meet specific needs or purposes.

如本文所用,术语“疫苗”是指用各类病原微生物或者他们的部分蛋白质或者核酸等生物大分子制作的用于预防接种的生物制品。As used herein, the term "vaccine" refers to a biological product used for vaccination made from various pathogenic microorganisms or their partial proteins or nucleic acids and other biological macromolecules.

如本文所用,术语“药物”是指能预防、治疗及诊断疾病的物质。As used herein, the term "drug" refers to a substance that can prevent, treat and diagnose a disease.

如本文所用,术语“突变体”是指某些碱基发生突变的核酸分子。As used herein, the term "mutant" refers to a nucleic acid molecule in which certain bases are mutated.

如本文所用,术语“突变体库”是指大量的(常常以亿万计)某些碱基发生突变的核酸分子所组成的群体。As used herein, the term "mutant library" refers to a population consisting of a large number (often in the billions) of nucleic acid molecules in which certain bases are mutated.

如本文所用,术语“达尔文式适应”是指类似自然选择过程,细胞在遭遇外部环境变化时,利用REPLACE系统提供突变,在环境提供的选择压力下,适应新的环境。As used herein, the term "Darwinian adaptation" refers to a process similar to natural selection, in which cells utilize the REPLACE system to provide mutations when encountering changes in the external environment, and adapt to the new environment under the selection pressure provided by the environment.

如本文所用,术语“正交的”是指宿主细胞基因组和REPLACE系统引入突变的过程互相独立,互不影响。As used herein, the term "orthogonal" means that the processes of introducing mutations into the host cell genome and the REPLACE system are independent of each other and do not affect each other.

如本文所用,术语“荧光蛋白”是指一类具有桶状结构的自身可以发射荧光的蛋白质。As used herein, the term "fluorescent protein" refers to a class of proteins having a barrel-shaped structure and capable of emitting fluorescence.

如本文所用,术语“转录因子”是指能与基因的顺式作用元件发生特异性相互作用,并对基因的转录有激活或抑制作用的DNA结合蛋白。As used herein, the term "transcription factor" refers to a DNA binding protein that can specifically interact with the cis-acting element of a gene and activate or inhibit the transcription of the gene.

如本文所用,术语“Cas蛋白”是指CRISPR(簇状规则间隔的短回文重复序列)相关蛋白的简称。As used herein, the term "Cas protein" refers to the abbreviation of CRISPR (clustered regularly interspaced short palindromic repeats) associated protein.

如本文所用,术语“微型Cas蛋白”是指蛋白质序列长度短于经典的Cas9,长度约为几百个氨基酸的Cas蛋白。As used herein, the term "mini-Cas protein" refers to a Cas protein with a shorter protein sequence length than the classic Cas9, which is about a few hundred amino acids in length.

如本文所用,术语“癌症相关的关键基因”是指本文中用到的MEK1和KRAS。As used herein, the term "cancer-related key genes" refers to MEK1 and KRAS as used herein.

如本文所用,术语“进化平台”是指可对生物大分子进行定向进化的技术。As used herein, the term "evolution platform" refers to a technology that can perform directed evolution of biological macromolecules.

如本文所用,术语“基于DNA的进化平台”是指在DNA层面引入突变并构建突变体库的定向进化技术。As used herein, the term "DNA-based evolution platform" refers to a directed evolution technology that introduces mutations at the DNA level and constructs a mutant library.

如本文所用,术语“基于RNA的进化平台”是指在RNA层面引入突变并构建突变体库的定向进化技术。As used herein, the term "RNA-based evolution platform" refers to a directed evolution technology that introduces mutations at the RNA level and constructs a mutant library.

如本文所用,术语“合成生物学”是指对生物体或者细胞进行有目标的设计、改造乃至重新合成,以使其具有特定生物学功能并可以在医学、农业、工业和环境等领域发挥价值的工程学科。As used herein, the term "synthetic biology" refers to an engineering discipline that specifically designs, modifies, or even resynthesizes organisms or cells to give them specific biological functions and to play a valuable role in fields such as medicine, agriculture, industry, and the environment.

如本文所用,术语“工程改造”是指对已有的生物大分子的序列进行改变,对其功能进行优化以使其满足人们的特定使用需求的过程。As used herein, the term "engineering" refers to the process of changing the sequence of an existing biomacromolecule and optimizing its function to make it meet people's specific usage requirements.

应理解,其他本文中出现的术语,应以本领域普通技术人员理解的本领域常规含义来理解相应的术语。在本文中所定义的术语与本领域其他文献中的相应定义相冲突的情况下,以本文中所定义的术语为准。It should be understood that other terms appearing in this article should be understood in accordance with the conventional meanings of the art as understood by those of ordinary skill in the art. In the event that a term defined in this article conflicts with a corresponding definition in other documents in the art, the term defined in this article shall prevail.

方法method

质粒的构建Plasmid construction

本研究中使用的质粒是通过使用Gibson组装或限制性酶切连接构建的。用于克隆或测序的引物从GENEWIZ订购。DNA或cDNA片段的扩增使用KOD OneTM PCR Master Mix(TOYOBO,#KMM-101)进行。质粒在大肠杆菌菌株5α(TSINGKE,TSC-C01)中维持和扩增,并用HiPure Plasmid DNA Mini Kit(Magen,#P100103)进行提取,最后通过Sanger测序(RUIBO)进行确认。Plasmids used in this study were constructed by using Gibson assembly or restriction ligation. Primers used for cloning or sequencing were ordered from GENEWIZ. Amplification of DNA or cDNA fragments was performed using KOD One PCR Master Mix (TOYOBO, #KMM-101). Plasmids were expressed in E. coli strains. 5α (TSINGKE, TSC-C01), extracted with HiPure Plasmid DNA Mini Kit (Magen, #P100103), and finally confirmed by Sanger sequencing (RUIBO).

相关质粒信息如下:The relevant plasmid information is as follows:

细胞培养Cell culture

本研究中使用了仓鼠成纤维细胞系BHK-21(ATCC)和人源细胞系HEK293T(ATCC)。BHK-21细胞在补充了10% FBS(Excell,#FSP500)、100单位/mL青霉素(penicillin)、100μg/mL链霉素(streptomycin)和1%MEM非必需氨基酸溶液(Gibco,#11140050)的MEM/EBSS培养基(HyClone,#SH3002401)中进行培养。HEK293T细胞在含有10% FBS(Excell,#FSP500)、100单位/mL青霉素和100μg/mL链霉素的DMEM培养基(Gibco,#C11995500BT)中培养。所有的细胞培养物都放置在37℃的、含有5%的CO2的加湿培养箱中,并日常进行支原体检测。Hamster fibroblast cell line BHK-21 (ATCC) and human cell line HEK293T (ATCC) were used in this study. BHK-21 cells were cultured in MEM/EBSS medium (HyClone, #SH3002401) supplemented with 10% FBS (Excell, #FSP500), 100 units/mL penicillin, 100 μg/mL streptomycin, and 1% MEM non-essential amino acid solution (Gibco, #11140050). HEK293T cells were cultured in DMEM medium (Gibco, #C11995500BT) containing 10% FBS (Excell, #FSP500), 100 units/mL penicillin, and 100 μg/mL streptomycin. All cell cultures were placed in a humidified incubator at 37°C with 5% CO2 , and mycoplasma testing was performed daily.

质粒转染和稳定细胞克隆的构建Plasmid transfection and construction of stable cell clones

质粒转染是通过使用LipofectamineTMLTX试剂与PLUSTM试剂(Thermofisher#15338100)或PEI试剂(聚乙二醇(polyethylenimine),1μg/μL)进行。PiggyBac(PB)转座子系统或慢病毒包装系统用于质粒稳定整合。为了构建复制酶有限复制的宿主细胞系(即在BHK-21基因组上稳定表达Sindbis病毒RNA依赖性RNA聚合酶(nsP4)的BHK-21细胞系),按照LipofectamineTMLTX试剂手册的说明,2.5μg的pEF1α-iRFP-NLS-P2A-ΔnsP3-nsP4质粒(或pEF1α-iRFP-NLS-TAG-ΔnsP3-nsP4用于nsP4表达减少的细胞系)和0.5μgPiggyBac转座酶质粒共同转染到含有BHK-21细胞的6孔板的一个孔中,细胞融汇度为~60%。转染后12小时更换培养基。培养一周后,使用BD FACSAriaTMIII细胞分选仪通过荧光激活细胞分选(FACS)富集iRFP阳性的BHK-21细胞。为了报告配体控制的合成转录因子或基于Cas的转录调控蛋白的活性,相应的报告质粒被包装到慢病毒中,所得的病毒颗粒被用来转染复制酶有限的BHK-21细胞系。具体来说,对于TetR-VP48,使用质粒pTRE3G-puroR-P2A-NLS-mCherry-BGHployA-pEF1α/HTLV-mTagBFP2-NLS-WPRE;对于PadR-VP64,质粒(OPadR)6-phCMVmin-puroR-NLS-mCherry-hPEST-BGHployA-pEF1α/HTLV-mTagBFP2-NLS-WPRE;而对于dCasMINI-VPR,使用质粒pTRE3G-puroR-P2A-NLS-mCherry-BGHployA-phU6-sgRNA_for_TetO-pEF1α/HTLV-tagBFP2-NLS-WPRE。对于病毒包装,HEK293T细胞在6孔板的一个孔中培养到~70%细胞汇合度,并按照PEI试剂手册的说明,用3μg报告质粒、2.66μg pCMVDR8.91质粒和0.34μgPMD2.G质粒进行转染。转染后8小时用2mL新鲜培养基更换培养基。48小时后,将含有病毒颗粒的上清液以500g离心3分钟,并通过0.45μm的过滤器过滤。将500μL的病毒上清液滴加到6孔板的一个孔中,该孔中的BHK-21细胞汇合度为~30%。病毒转导后12小时更换培养基。4天后,通过流式分选富集BFP阳性的BHK-21细胞。这些多克隆细胞被培养并用于后续实验。Plasmid transfection was performed by using Lipofectamine LTX reagent with PLUS reagent (Thermofisher #15338100) or PEI reagent (polyethylenimine, 1 μg/μL). PiggyBac (PB) transposon system or lentiviral packaging system was used for stable integration of plasmids. To construct a replicase-limited host cell line (i.e., a BHK-21 cell line stably expressing Sindbis viral RNA-dependent RNA polymerase (nsP4) on the BHK-21 genome), 2.5 μg of pEF1α-iRFP - NLS-P2A-ΔnsP3-nsP4 plasmid (or pEF1α-iRFP-NLS-TAG-ΔnsP3-nsP4 for cell lines with reduced nsP4 expression) and 0.5 μg of PiggyBac transposase plasmid were co-transfected into one well of a 6-well plate containing BHK-21 cells according to the instructions of the Lipofectamine ™ LTX reagent manual, with a cell confluency of ∼60%. The medium was changed 12 hours after transfection. After one week of culture, iRFP-positive BHK-21 cells were enriched by fluorescence-activated cell sorting (FACS) using a BD FACSAria™ III cell sorter. To report the activity of ligand-controlled synthetic transcription factors or Cas-based transcriptional regulatory proteins, the corresponding reporter plasmids were packaged into lentiviruses, and the resulting viral particles were used to transfect the replicase-limited BHK-21 cell line. Specifically, for TetR-VP48, the plasmid pTRE3G-puroR-P2A-NLS-mCherry-BGHployA-pEF1α/HTLV-mTagBFP2-NLS-WPRE was used; for PadR-VP64, the plasmid (O PadR ) 6 -phCMVmin-puroR-NLS-mCherry-hPEST-BGHployA-pEF1α/HTLV-mTagBFP2-NLS-WPRE was used; and for dCasMINI-VPR, the plasmid pTRE3G-puroR-P2A-NLS-mCherry-BGHployA-phU6-sgRNA_for_TetO-pEF1α/HTLV-tagBFP2-NLS-WPRE was used. For viral packaging, HEK293T cells were cultured to ∼70% cell confluence in one well of a 6-well plate and transfected with 3 μg of reporter plasmid, 2.66 μg of pCMVDR8.91 plasmid, and 0.34 μg of PMD2.G plasmid according to the instructions of the PEI reagent manual. The medium was replaced with 2 mL of fresh medium 8 hours after transfection. After 48 hours, the supernatant containing viral particles was centrifuged at 500 g for 3 minutes and filtered through a 0.45 μm filter. 500 μL of viral supernatant was added dropwise to one well of a 6-well plate in which the confluence of BHK-21 cells was ∼30%. The medium was replaced 12 hours after viral transduction. Four days later, BFP-positive BHK-21 cells were enriched by flow sorting. These polyclonal cells were cultured and used for subsequent experiments.

自主复制RNA的体外转录和电转In vitro transcription and electroporation of autonomously replicating RNA

RNA首先在体外转录,然后电穿孔转染到BHK-21细胞。为了准备体外转录的模板DNA,将6μg质粒DNA与4μL XbaI(NEB,#R0145)在80μL反应中在37℃下线性化12小时以上。为了终止酶切反应,向反应中加入4μL 0.5M EDTA、8μL 3M醋酸钠(sodium acetate)和184μL乙醇(ethanol)。混合物充分混合,并在-80℃下冷冻至少20分钟,然后在4℃下以13000×g离心15分钟,以沉淀线性化的DNA。去除上清液后,用500μL 75%冰冷的乙醇,仔细清洗通常看不见的沉淀,然后在4℃下以13,000×g离心3分钟。洗涤过程重复两次。完全除去乙醇后,将线性化的DNA重悬于10μL 10%(v/v)RNAse抑制剂溶液(RNasin,Promega,#N2111)中,立即作为模板用于体外转录。按照mMESSAGE SP6转录试剂盒(Thermo,#AM1340)的用户指南,使用制备的模板DNA进行加帽转录反应。对于一个10μL的反应,使用5μL的2X NTP/CAP,1μL的10X反应缓冲液,1μL的GTP,300ng的线性模板DNA,1μL的混合酶,以及~1μL的无RNA的H2O。在37℃下孵育2小时后,立即使用或在-80℃下保存混合物。必要时,加入1μL TURBO DNase去除DNA,然后在37℃孵育15分钟,再用氯化锂(lithium choloride)沉淀法进行纯化。对于电穿孔步骤,本发明人修改了以前文献中的操作流程81。简而言之,在100毫米培养皿中培养的BHK-21细胞,在90%汇合度时,通过胰蛋白酶消化收获,然后用10mL冰冷的DPBS清洗两次。然后将4×106细胞悬浮在390μL冰冷的DPBS中,与10μL体外转录的RNA混合。将混合物转移到一个冷冻的2毫米间隙的无菌电穿孔比色皿,用Bio-rad GenePulser II间隔3秒的三个指数衰减脉冲进行电穿孔实验。该设备的设置如下:场强2250V/cm,电容25μF,电阻无限大。电穿孔后,让细胞在0℃下恢复10分钟,在10mL BHK-21细胞培养基中稀释,然后转移到100毫米的组织培养皿中进行正常培养。RNA was first transcribed in vitro and then electroporated into BHK-21 cells. To prepare the template DNA for in vitro transcription, 6 μg of plasmid DNA was linearized with 4 μL XbaI (NEB, #R0145) in an 80 μL reaction at 37°C for more than 12 hours. To terminate the enzyme digestion reaction, 4 μL 0.5M EDTA, 8 μL 3M sodium acetate, and 184 μL ethanol were added to the reaction. The mixture was thoroughly mixed and frozen at -80°C for at least 20 minutes, then centrifuged at 13,000 × g for 15 minutes at 4°C to precipitate the linearized DNA. After removing the supernatant, the usually invisible precipitate was carefully washed with 500 μL 75% ice-cold ethanol, and then centrifuged at 13,000 × g for 3 minutes at 4°C. The washing process was repeated twice. After completely removing the ethanol, the linearized DNA was resuspended in 10 μL of 10% (v/v) RNAse inhibitor solution (RNasin, Promega, #N2111) and immediately used as a template for in vitro transcription. The capping transcription reaction was performed using the prepared template DNA according to the user guide of the SP6 transcription kit (Thermo, #AM1340). For a 10 μL reaction, 5 μL of 2X NTP/CAP, 1 μL of 10X reaction buffer, 1 μL of GTP, 300 ng of linear template DNA, 1 μL of enzyme mix, and ~1 μL of RNA-free H 2 O were used. After incubation at 37°C for 2 hours, the mixture was used immediately or stored at -80°C. If necessary, 1 μL of TURBO DNase was added to remove DNA, followed by incubation at 37°C for 15 minutes and purification by lithium choloride precipitation. For the electroporation step, the inventors modified the protocol from a previous literature 81 . Briefly, BHK-21 cells cultured in 100 mm dishes were harvested by trypsinization at 90% confluence and then washed twice with 10 mL of ice-cold DPBS. Then 4×10 6 cells were suspended in 390 μL of ice-cold DPBS and mixed with 10 μL of in vitro transcribed RNA. The mixture was transferred to a frozen 2 mm gap sterile electroporation cuvette and electroporated using a Bio-rad GenePulser II with three exponential decay pulses at 3 s intervals. The settings of the device were as follows: field strength 2250 V/cm, capacitance 25 μF, and infinite resistance. After electroporation, the cells were allowed to recover at 0°C for 10 minutes, diluted in 10 mL of BHK-21 cell culture medium, and then transferred to a 100 mm tissue culture dish for normal culture.

nsP1和nsP2的倒数第二个氨基酸的突变库筛选Screening of mutation library of the penultimate amino acid of nsP1 and nsP2

为了确定nsP1和nsP2的倒数第二个氨基酸的对RNA的宿主兼容性最有利的组合,本发明人构建了一个基于'NNK'的两点定点饱和突变库(nsP1:G539X,nsP2:G806X;X代表20个编码蛋白的氨基酸之一)。突变库的构建包括两个连续的步骤:首先,构建基于Sindbis复制子质粒的中间载体(ML702|pSP6-nsP1-nsP3-nsP4-pSG-GFP-P2A-puroR),缺失nsP2,并加入两个限制酶位点(NheI和EcoRI)。其次,用简并引物对nsP2进行PCR扩增,并通过Gibson组装将这些片段与NheI和EcoRI切割过的ML702相连。然后将突变体文库在体外转录并电转导入野生型BHK-21细胞。转染后,细胞恢复24小时,然后加入10μg/mL的嘌呤霉素。每2天用新鲜的含嘌呤霉素的培养基替换旧的培养基。两周后,使用TRIzolTM试剂(Thermo Fisher,#15596018)收集细胞以提取RNA。使用HiScript III第一链cDNA合成试剂盒(Vazyme,#R312-02)对约3μg的总RNA进行逆转录,所用逆转录引物为复制子特异性引物。使用KOD OneTM PCRMaster Mix和目标特异引物对cDNA进行PCR扩增。使用快速NGS服务(TsingkeBiotechnology)对PCR产物进行测序,以量化nsP1和nsP2倒数第二个氨基酸的每种氨基酸类型的比例。在筛选前对突变库进行了类似的量化。每个氨基酸的富集倍数被量化为筛选后和筛选前的比率。对复制酶有限的BHK-21稳定细胞系进行了类似的文库筛选实验并进行了量化,其中nsP4被NeoR取代。To determine the most favorable combination of the penultimate amino acids of nsP1 and nsP2 for RNA host compatibility, the inventors constructed a two-site saturation mutation library based on 'NNK' (nsP1: G539X, nsP2: G806X; X represents one of the 20 protein-encoding amino acids). The construction of the mutation library involved two consecutive steps: First, an intermediate vector based on the Sindbis replicon plasmid (ML702|pSP6-nsP1-nsP3-nsP4-pSG-GFP-P2A-puroR) was constructed, nsP2 was deleted, and two restriction enzyme sites (NheI and EcoRI) were added. Second, nsP2 was PCR amplified with degenerate primers, and these fragments were connected to ML702 cut with NheI and EcoRI by Gibson assembly. The mutant library was then transcribed in vitro and electroporated into wild-type BHK-21 cells. After transfection, the cells recovered for 24 hours, and then 10 μg/mL of puromycin was added. The old medium was replaced with fresh medium containing puromycin every 2 days. After two weeks, cells were collected using TRIzol TM reagent (Thermo Fisher, #15596018) to extract RNA. About 3 μg of total RNA was reverse transcribed using HiScript III First Strand cDNA Synthesis Kit (Vazyme, #R312-02), and the reverse transcription primers used were replicon-specific primers. cDNA was PCR amplified using KOD One TM PCR Master Mix and target-specific primers. PCR products were sequenced using a fast NGS service (Tsingke Biotechnology) to quantify the ratio of each amino acid type of the penultimate amino acid of nsP1 and nsP2. Similar quantification was performed on the mutant library before screening. The enrichment multiple of each amino acid was quantified as the ratio after screening and before screening. Similar library screening experiments and quantification were performed on the replicase-limited BHK-21 stable cell line, in which nsP4 was replaced by NeoR.

活细胞成像Live cell imaging

在用不同版本的Sindbis病毒RNA转染的BHK-21细胞的培养过程中,在相关实验的指定时间点将培养有细胞的100毫米组织培养皿从培养箱中取出,进行活细胞成像。在FL自动成像系统(Thermo Fisher Scientific)上用LPlanFL PH2 4x/0.13或LPlanFL PH2 10x/0.30物镜拍摄细胞的明场和GFP荧光。所得图像使用ImageJ2(https://imagej.net/software/imagej2/)进行调整和剪裁。During the culture of BHK-21 cells transfected with different versions of Sindbis viral RNA, 100 mm tissue culture dishes containing cells were removed from the incubator at designated time points in the relevant experiments for live cell imaging. Bright field and GFP fluorescence of cells were captured using LPlanFL PH2 4x/0.13 or LPlanFL PH2 10x/0.30 objective lens on FL automatic imaging system (Thermo Fisher Scientific). The obtained images were adjusted and cropped using ImageJ2 (https://imagej.net/software/imagej2/).

流式分析Streaming analysis

为了分析荧光蛋白的表达(即mTagBFP2、EGFP、mCherry和iRFP),制备了单细胞悬浮液并在BD LSRFortessaTM细胞分析仪上进行流式分析。数据由MATLAB脚本(https://antebilab.github.io/easyflow/)进行分析。除非另有说明,阳性细胞是根据非转染的对照细胞来确定的,平均荧光强度(MFI)被用来代表蛋白表达水平。为了分析细胞进入的早期时间点的RNA的复制,复制酶水平(即nsP4的表达水平)由iRFP水平近似。具有类似复制酶水平的细胞被分类,每群细胞的EGFP(从自主复制RNA表达而来)的MFI被计算作为RNA水平的代表。对于使用流式细胞仪分析CD2的表达的实验,本发明人采用了以前文献中的方法20。简而言之,对于每个重复,使用PEI试剂将2μg相应的质粒(pEF1α-NLSdCasMINI-NLS-VPR-phU6-sgRNA_to_CD2-pEF1α/HTLV-mTagBFP2-NLS)转染到HEK293T细胞。三天后,制备单细胞悬液,用FITC-CD2抗体(Biolegend公司,#309206)在PBS中加5%FBS在4℃下染色1小时,并通过流式细胞仪进行分析。得到的数据按所述方式进行处理。To analyze the expression of fluorescent proteins (i.e., mTagBFP2, EGFP, mCherry, and iRFP), single cell suspensions were prepared and flow cytometry was performed on a BD LSRFortessa TM cell analyzer. Data were analyzed by a MATLAB script (https://antebilab.github.io/easyflow/). Unless otherwise stated, positive cells were determined based on non-transfected control cells, and mean fluorescence intensity (MFI) was used to represent protein expression levels. To analyze RNA replication at early time points of cell entry, replicase levels (i.e., nsP4 expression levels) were approximated by iRFP levels. Cells with similar replicase levels were sorted, and the MFI of EGFP (expressed from autonomously replicating RNA) for each group of cells was calculated as a representative of RNA levels. For experiments analyzing the expression of CD2 using flow cytometry, the inventors used methods from previous literature20 . In brief, for each replicate, 2 μg of the corresponding plasmid (pEF1α-NLSdCasMINI-NLS-VPR-phU6-sgRNA_to_CD2-pEF1α/HTLV-mTagBFP2-NLS) was transfected into HEK293T cells using PEI reagent. Three days later, single cell suspensions were prepared, stained with FITC-CD2 antibody (Biolegend, #309206) in PBS plus 5% FBS at 4°C for 1 hour, and analyzed by flow cytometry. The data obtained were processed as described.

dsRNA的免疫荧光分析Immunofluorescence analysis of dsRNA

为了进行病毒的双链RNA(dsRNA)的免疫染色,提前24小时将BHK-21细胞培养在24孔玻璃板上。细胞在室温下用4%多聚甲醛(paraformaldehyde)固定20分钟,用PBS洗涤两次,并在-20℃下用100%冰冷的甲醇(methanol)通透30分钟。用PBS洗两次后,用1%不含IgG的牛血清白蛋白(BSA)封闭细胞30分钟。然后用0.5μg/mL含有1%BSA的J2抗体(Scicons#10010200)孵育细胞1小时。由于稀释的J2抗体可能含有少量变性的蛋白质聚合物,可能导致免疫荧光检测的高背景,稀释的J2抗体在使用前需要在13000×g离心3分钟。用1% BSA洗涤两次后,用1/1000稀释的山羊抗小鼠IgG H&L(Alexa568)二抗(Abcam)在黑暗中孵育细胞45分钟,然后在黑暗中用PBS洗涤两次。然后加入5μg/mLHoechst 33342,在室温下对细胞核进行染色,在黑暗中持续10分钟,然后用PBS在黑暗中清洗两次。然后在Dragon Fly200高速共聚焦平台(ANDOR)上用Plan-Apo 63x/1.4油镜(Leica)对细胞进行成像。使用ImageJ和内部的MATLAB脚本处理图像。在图像处理方面,首先对荧光Z轴切片图像根据最大值进行合并、阈值处理和分割。对于dsRNA免疫荧光图像,使用灰度值200作为阈值,ImageJ中的颗粒分析插件被应用于所得到的二进制图像,以量化单个颗粒。对于细胞核图像,使用Huang方法82计算阈值,在连续的扩张和侵蚀操作后,通过分水岭算法生成单个细胞的核掩模。颗粒分析被应用于核掩模图像,以量化细胞核的大小。粒子分析插件的输出在MATLAB中进一步处理,以估计每个视场中每个细胞的dsRNA数量。通过比较对照组和实验组的图像,真正的dsRNA信号被经验性地定义为至少有6个连接像素的颗粒。单个dsRNA分子的信号强度是通过使用最小尺寸的减去背景的dsRNA信号的平均强度来估计的。然后将每个视野中的减去背景的真实dsRNA信号相加,再除以单个dsRNA的估计强度和细胞核的数量,得到目标视野中每个细胞的估计的dsRNA数量。For immunostaining of viral double-stranded RNA (dsRNA), BHK-21 cells were cultured on 24-well glass plates 24 hours in advance. The cells were fixed with 4% paraformaldehyde for 20 minutes at room temperature, washed twice with PBS, and permeabilized with 100% ice-cold methanol for 30 minutes at -20°C. After washing twice with PBS, the cells were blocked with 1% IgG-free bovine serum albumin (BSA) for 30 minutes. The cells were then incubated with 0.5 μg/mL J2 antibody (Scicons #10010200) containing 1% BSA for 1 hour. Since the diluted J2 antibody may contain a small amount of denatured protein aggregates, which may cause high background in immunofluorescence detection, the diluted J2 antibody needs to be centrifuged at 13,000 × g for 3 minutes before use. After washing twice with 1% BSA, the cells were washed with 1/1000 diluted goat anti-mouse IgG H&L (Alexa Fluor ELISA Kit). 568) secondary antibody (Abcam) was incubated in the dark for 45 min and then washed twice with PBS in the dark. Then 5 μg/mL Hoechst 33342 was added to stain the nuclei at room temperature for 10 min in the dark and then washed twice with PBS in the dark. The cells were then imaged with a Plan-Apo 63x/1.4 oil objective (Leica) on a Dragon Fly200 high-speed confocal platform (ANDOR). Images were processed using ImageJ and in-house MATLAB scripts. In terms of image processing, the fluorescence Z-axis slice images were first merged, thresholded, and segmented according to the maximum value. For dsRNA immunofluorescence images, a gray value of 200 was used as the threshold, and the particle analysis plug-in in ImageJ was applied to the resulting binary images to quantify individual particles. For nucleus images, the threshold was calculated using the Huang method82 , and nuclear masks of individual cells were generated by the watershed algorithm after consecutive dilation and erosion operations. Particle analysis was applied to the nuclear mask images to quantify the size of the nucleus. The output of the particle analysis plugin was further processed in MATLAB to estimate the number of dsRNA per cell in each field of view. By comparing the images of the control and experimental groups, the true dsRNA signal was empirically defined as particles with at least 6 connected pixels. The signal intensity of a single dsRNA molecule was estimated by using the average intensity of the background-subtracted dsRNA signal of the smallest size. The background-subtracted true dsRNA signal in each field of view was then summed and divided by the estimated intensity of a single dsRNA and the number of cell nuclei to obtain the estimated number of dsRNA per cell in the field of view of interest.

RNA测序实验和数据分析RNA sequencing experiments and data analysis

为了比较携带不同自主复制RNA的细胞之间的转录组,使用BD FACSAriaTMIII细胞分选仪分选出携带repRNA-v3细胞或repRNA-v4细胞(电穿孔后培养约3周)的30万个BHK-21细胞。使用TRIzolTM试剂(Thermo Fisher Scientific,#15596018)提取总RNA,使用VAHTSUniversal V6 RNA-seq Library Prep Kit for MGI(Vazyme,#NRM604-2)进行文库制备。通过安诺优达基因技术公司在DNBSEQ-T7平台上对所得文库进行下一代测序。使用STAR2.7.8a将去掉低质量序列的双端150bp序列与添加了自主复制RNA基因组(nsP1、nsP2、nsP3和GFP)的Mesocricetus auratus基因组(GCF_017639785.1_BCM_Maur_2.0_genomic.fna)进行比对。基因组注释(GCF_017639785.1_BCM_Maur_2.0_genomic.gtf)是从NCBI下载并进行编辑,以包括自主复制RNA基因组上的基因信息。转录物丰度由featureCounts计算,并使用DESeq2进行归一化和统计学比较。然后计算RPKM值,并在Reactome网站(https://reactome.org/)上进行生物途径富集分析。In order to compare the transcriptomes between cells carrying different autonomously replicating RNAs, 300,000 BHK-21 cells carrying repRNA-v3 cells or repRNA-v4 cells (cultured for about 3 weeks after electroporation) were sorted using a BD FACSAria TM III cell sorter. Total RNA was extracted using TRIzol TM reagent (Thermo Fisher Scientific, #15596018), and the library was prepared using VAHTSUniversal V6 RNA-seq Library Prep Kit for MGI (Vazyme, #NRM604-2). The resulting library was sequenced by next generation sequencing on the DNBSEQ-T7 platform by Annouda Gene Technology. The paired-end 150bp sequences with low-quality sequences removed were aligned to the Mesocricetus auratus genome (GCF_017639785.1_BCM_Maur_2.0_genomic.fna) with the addition of the autonomously replicating RNA genome (nsP1, nsP2, nsP3, and GFP) using STAR2.7.8a. The genome annotation (GCF_017639785.1_BCM_Maur_2.0_genomic.gtf) was downloaded from NCBI and edited to include gene information on the autonomously replicating RNA genome. Transcript abundance was calculated by featureCounts and normalized and statistically compared using DESeq2. RPKM values were then calculated and biological pathway enrichment analysis was performed on the Reactome website (https://reactome.org/).

自主复制RNA的突变分析Mutation analysis of autonomously replicating RNA

为了分析在不同条件下,如有无复制酶诱变剂,以及进化前或后,自主复制RNA(repRNA-v4,除非另有说明)的突变率和突变谱,相应的细胞被收集,并进行总RNA提取和逆转录。使用KOD OneTM PCR Master Mix和目标特异性引物从cDNA样品中扩增感兴趣的序列(例如,nsP1-nsP3、EGFP、TetR-VP64、VP64-PadR、dCasMINI、MEK1、KRAS)。突变率和突变谱通过使用NGS测序(擎科生物的快速NGS服务)进行量化,单个cDNA变体通过克隆到质粒中的cDNA的Sanger测序(RUIBIO)得到。对于NGS数据的处理,使用BWA 0.7.17-r1188将清理过的读数比对到相应的参考序列。使用内部的Bash和Python脚本处理得到的Bam文件,以提取所需的突变信息。为了对突变谱进行分析,每种突变类型(如u到c)的突变频率被计算为相应突变的数量除以测序核苷酸的总数。为了分析进化的RNA的突变,用每个核苷酸的突变的核苷酸所占的百分比表示。To analyze the mutation rate and mutation spectrum of autonomously replicating RNA (repRNA-v4, unless otherwise stated) under different conditions, such as with or without replicase mutagens, and before or after evolution, the corresponding cells were collected and subjected to total RNA extraction and reverse transcription. The sequences of interest (e.g., nsP1-nsP3, EGFP, TetR-VP64, VP64-PadR, dCasMINI, MEK1, KRAS) were amplified from cDNA samples using KOD One TM PCR Master Mix and target-specific primers. The mutation rate and mutation spectrum were quantified by using NGS sequencing (Qingke Bio's Rapid NGS Service), and individual cDNA variants were obtained by Sanger sequencing of cDNA cloned into plasmids (RUIBIO). For the processing of NGS data, the cleaned reads were aligned to the corresponding reference sequences using BWA 0.7.17-r1188. The resulting Bam files were processed using in-house Bash and Python scripts to extract the required mutation information. For analysis of mutational spectra, the frequency of each mutation type (e.g., U to C) was calculated as the number of corresponding mutations divided by the total number of sequenced nucleotides. For analysis of mutations in evolved RNA, the percentage of nucleotides with mutations per nucleotide was expressed.

EGFP进化实验EGFP evolution experiment

该系统的设计和该实验的时间线展示在图37和图38的顶部中。本实验中使用的细胞在电穿孔后培养了约2周,并最初在图26、28、31、32、36中用于突变相关实验。进化实验是在100毫米的组织培养皿中进行的。用10μM的Molnupiravir(MCE,#HY-135853)处理~20%汇合度的细胞2天,并每天更换培养基。然后将细胞转移到5个100毫米的培养皿里,让其生长3天。对大约3000万个细胞进行流式分选以获得蓝移的EGFP突变体,其中0.2%的细胞被分选为阳性。分选的细胞在10μg/mL嘌呤霉素的作用下培养约7天,直到获得足够的细胞进行第二轮进化,其过程与第一轮相同。实验期间从细胞中提取的自主复制RNA(中间细胞群或最终细胞群)被逆转录,并通过NGS测序来量化突变。将cDNA库(包含EGFP片段)克隆到一个载体(pEF1α-EGFP mutant-P2A-Puro-BGH polyA-pEF1α/HTLV-iRFP-NLS)中,将其转化到大肠杆菌中,通过Sanger测序鉴定各个突变体。为了量化EGFP变体的发射光光谱,使用PEI试剂将600ng的EGFP野生型或突变体质粒转染到24孔板的HEK293T细胞中。转染12小时后,用新鲜培养基替换,一天后用流式细胞仪测量EGFP和BFP通道的荧光强度。The design of the system and the timeline of the experiment are shown in the top of Figures 37 and 38. The cells used in this experiment were cultured for about 2 weeks after electroporation and were initially used for mutation-related experiments in Figures 26, 28, 31, 32, and 36. The evolution experiment was performed in a 100 mm tissue culture dish. Cells at ~20% confluence were treated with 10 μM Molnupiravir (MCE, #HY-135853) for 2 days, and the culture medium was changed every day. The cells were then transferred to 5 100 mm culture dishes and allowed to grow for 3 days. Approximately 30 million cells were flow sorted to obtain blue-shifted EGFP mutants, of which 0.2% of the cells were sorted as positive. The sorted cells were cultured for about 7 days under the action of 10 μg/mL puromycin until enough cells were obtained for a second round of evolution, which was the same as the first round. Autonomously replicating RNA extracted from cells during the experiment (intermediate cell population or final cell population) was reverse transcribed and mutations were quantified by NGS sequencing. The cDNA library (containing the EGFP fragment) was cloned into a vector (pEF1α-EGFP mutant-P2A-Puro-BGH polyA-pEF1α/HTLV-iRFP-NLS), which was transformed into E. coli, and each mutant was identified by Sanger sequencing. To quantify the emission spectrum of EGFP variants, 600 ng of EGFP wild-type or mutant plasmids were transfected into HEK293T cells in 24-well plates using PEI reagent. After 12 hours of transfection, the medium was replaced with fresh medium, and the fluorescence intensity of the EGFP and BFP channels was measured by flow cytometry one day later.

TetR-VP48(tTA)进化实验TetR-VP48 (tTA) evolution experiments

该系统的设计和该实验的时间线展示在图42中。TetR-VP48被克隆到类似于repRNA-v4的质粒中(其3个关键氨基酸是'VLV'而不是repRNA-v4中的'ALI'),并将得到的自主复制RNA电穿孔到表达nsP4的BHK-21细胞(含有ML700质粒的稳定细胞系)。电穿孔后24小时用含有5μg/mL嘌呤霉素和100μg/mL新霉素(neomycin)的新鲜培养基替换。经过3天的选择,获得了稳定携带自主复制RNA的细胞群(约100万个细胞),并作为定向进化实验的起始材料,在两个100毫米的培养皿里进行。诱变和选择的步骤见图42的b。两个培养皿中的细胞合并起来进行FACS。分选的细胞在80μg/mL嘌呤霉素的作用下培养3天,收集后进行RNA提取、测序和突变分析。将cDNA库(包含TetR基因)克隆到一个载体(pEF1α-TetR_mutant-VP48-BGH polyA-pEF1α/HTLV-iRFP-NLS)中,将其转化到大肠杆菌中,通过Sanger测序确定单个突变体。为了量化TetR变体的配体敏感性,将400ng TetR(野生型或突变体)-VP48质粒和200ng报告质粒(pTRE3G-Citline-NLS)在24孔板的含有1ng/μL多西环素(Dox)的培养基的孔中共同转染到HEK293T细胞内。转染12小时后,用含有Dox的新鲜培养基替换培养基,一天后用流式细胞仪测量荧光强度。The design of the system and the timeline of the experiment are shown in Figure 42. TetR-VP48 was cloned into a plasmid similar to repRNA-v4 (its three key amino acids are 'VLV' instead of 'ALI' in repRNA-v4), and the resulting self-replicating RNA was electroporated into BHK-21 cells expressing nsP4 (a stable cell line containing the ML700 plasmid). 24 hours after electroporation, the culture medium was replaced with fresh medium containing 5μg/mL puromycin and 100μg/mL neomycin. After 3 days of selection, a cell population (about 1 million cells) that stably carries the self-replicating RNA was obtained and used as the starting material for the directed evolution experiment in two 100 mm culture dishes. The steps of mutagenesis and selection are shown in Figure 42b. The cells in the two culture dishes were combined for FACS. The sorted cells were cultured for 3 days under the action of 80μg/mL puromycin and collected for RNA extraction, sequencing and mutation analysis. The cDNA library (containing the TetR gene) was cloned into a vector (pEF1α-TetR_mutant-VP48-BGH polyA-pEF1α/HTLV-iRFP-NLS), which was transformed into E. coli, and individual mutants were determined by Sanger sequencing. To quantify the ligand sensitivity of TetR variants, 400ng of TetR (wild type or mutant)-VP48 plasmid and 200ng of reporter plasmid (pTRE3G-Citline-NLS) were co-transfected into HEK293T cells in a well of a 24-well plate containing 1ng/μL doxycycline (Dox). After 12 hours of transfection, the medium was replaced with fresh medium containing Dox, and the fluorescence intensity was measured by flow cytometry one day later.

VP64-PadR进化实验VP64-PadR evolution experiments

该系统的设计和该实验的时间线展示在图45的顶部中。VP64-PadR56(ENCU,由叶海峰教授赠予)被克隆到repRNA-v4质粒中,并将得到的自主复制RNA电穿孔到表达nsP4(含有ML700质粒的稳定细胞系)和携带稳定整合的报告质粒((OPadR)6-phCMVmini-PuroR-NLS-mCherry-hPEST-BGHpolyA-pEF1α/HTLV-tag BFP2-NLS)的BHK-21细胞。在电穿孔后24小时,用含有5μg/mL嘌呤霉素和10μg/mL杀稻瘟菌素(blasticidin)的新鲜培养基替换。经过3天的选择,获得了稳定携带自主复制RNA的细胞群(约500万个细胞),并作为在100毫米的培养皿中进行定向进化实验的起始材料。该实验包括两个独立的定向进化活动,即一个是进化出抗配体的突变体,另一个是在没有配体的情况下进化出靶基因的激活增强。相应的突变和选择过程总结在了图45的底部、图49中。在第21天,收集第一个实验的所有细胞和第二个实验通过mCherry筛选的细胞,进行RNA提取、测序和突变分析。将cDNA库(包含PadR基因)连接到一个载体(pEF1α-VP64-PadR_mutant-BGHpolyA-pEF1α/HTLViRFP-NLS),再将其转化到大肠杆菌中,通过Sanger测序确定单个突变体。为了量化PadR变体的配体敏感性,将400ngVP64-PadR(野生型或突变体)质粒和200ng报告质粒((OPadR)6-phCMVmini-Puro-NLS-mCherry-hPEST-BGHpolyA-pEF1α/HTLV-mTagBFP2-NLS)共同转染在24孔板的一个含有阿魏酸钠(SF)的培养基孔中的HEK293T细胞。转染后12小时,用新鲜的含阿魏酸钠的培养基替换培养基,一天后用流式细胞仪测量荧光强度。The design of the system and the timeline of the experiment are shown at the top of Figure 45. VP64-PadR 56 (ENCU, gifted by Professor Ye Haifeng) was cloned into the repRNA-v4 plasmid, and the resulting self-replicating RNA was electroporated into BHK-21 cells expressing nsP4 (stable cell line containing ML700 plasmid) and carrying a stably integrated reporter plasmid ((O PadR ) 6 -phCMVmini-PuroR-NLS-mCherry-hPEST-BGHpolyA-pEF1α/HTLV-tag BFP2-NLS). 24 hours after electroporation, fresh culture medium containing 5 μg/mL puromycin and 10 μg/mL blasticidin was replaced. After 3 days of selection, a cell population (about 5 million cells) that stably carried the self-replicating RNA was obtained and used as the starting material for directed evolution experiments in 100 mm culture dishes. The experiment included two independent directed evolution activities, one to evolve ligand-resistant mutants and the other to evolve enhanced activation of target genes in the absence of ligands. The corresponding mutation and selection processes are summarized at the bottom of Figure 45 and Figure 49. On day 21, all cells in the first experiment and cells screened by mCherry in the second experiment were collected for RNA extraction, sequencing and mutation analysis. The cDNA library (containing the PadR gene) was connected to a vector (pEF1α-VP64-PadR_mutant-BGHpolyA-pEF1α/HTLViRFP-NLS), which was then transformed into Escherichia coli, and single mutants were determined by Sanger sequencing. To quantify the ligand sensitivity of PadR variants, 400 ng of VP64-PadR (wild type or mutant) plasmid and 200 ng of reporter plasmid ((O PadR ) 6 -phCMVmini-Puro-NLS-mCherry-hPEST-BGHpolyA-pEF1α/HTLV-mTagBFP2-NLS) were co-transfected into HEK293T cells in one well of a 24-well plate in a medium containing sodium ferulate (SF). 12 hours after transfection, the medium was replaced with fresh medium containing sodium ferulate, and the fluorescence intensity was measured by flow cytometry one day later.

CasMINI进化实验CasMINI evolution experiment

该系统的设计和该实验的时间线展示在图67和68中。NLS-dCasMINI(v4)-NLS-VPR20被克隆到repRNA-v4质粒中,并将得到的自主复制RNA电穿孔到表达nsP4(含有ML700质粒的稳定细胞系)和携带稳定整合的报告质粒(pTRE3G-PuroR-P2A-NLS-mCherry-BGHpolyA-phU6-sgRNA_TetO-pEF1α/HTLV-tagBFP2-NLS)的BHK-21细胞。在电穿孔后24小时,用含有5μg/mL嘌呤霉素和10μg/mL杀稻瘟菌素的新鲜培养基替换旧的培养基。经过5天的选择,获得了稳定携带自主复制RNA的细胞群(约200万个细胞),并被用作定向进化实验的起始材料,在100毫米的培养皿里进行。在第22天,收集细胞进行RNA提取、测序和突变分析。将cDNA库(含dCasMIN基因)连接到一个载体(pEF1α-NLS-dCasMINI_mutant-NLS-VPR-BGH polyA-pEF1α/HTLV-iRFP-NLS),转化到大肠杆菌中,通过Sanger测序进行突变体鉴定。本发明人在两次重复中只得到一个富集的突变(S246R)。本发明人表征了该突变体的三种功能,包括转录激活功能、碱基编辑功能和核酸酶功能。sgRNA的信息和相应的目标基因信息来自于以前的研究20。作为转录激活因子,本发明人对野生型(dCasMINI(V4))和突变体(S246R)激活进化过程中使用的报告系统和内源基因CD2的能力进行了表征。作为碱基编辑器或核酸酶,本发明人对两个内源基因IFNγ和VEGFA进行了测试。这些试验中使用的质粒信息详见Supplementary Table 1。在所有的测试中,将2μg质粒转染到12孔板的一个孔中的HEK293T细胞。转染后12小时,用新鲜培养基替换,60小时后,收集细胞进行流式分析或测序。The design of the system and the timeline of the experiment are shown in Figures 67 and 68. NLS-dCasMINI (v4) -NLS-VPR 20 was cloned into the repRNA-v4 plasmid, and the resulting self-replicating RNA was electroporated into BHK-21 cells expressing nsP4 (stable cell line containing ML700 plasmid) and carrying a stably integrated reporter plasmid (pTRE3G-PuroR-P2A-NLS-mCherry-BGHpolyA-phU6-sgRNA_TetO-pEF1α/HTLV-tagBFP2-NLS). 24 hours after electroporation, the old culture medium was replaced with fresh culture medium containing 5 μg/mL puromycin and 10 μg/mL blasticidin. After 5 days of selection, a cell population (about 2 million cells) that stably carried self-replicating RNA was obtained and used as the starting material for directed evolution experiments in 100 mm culture dishes. On the 22nd day, cells were collected for RNA extraction, sequencing, and mutation analysis. The cDNA library (containing the dCasMIN gene) was connected to a vector (pEF1α-NLS-dCasMINI_mutant-NLS-VPR-BGH polyA-pEF1α/HTLV-iRFP-NLS), transformed into Escherichia coli, and mutants were identified by Sanger sequencing. The inventors only obtained one enriched mutation (S246R) in two replicates. The inventors characterized three functions of the mutant, including transcriptional activation function, base editing function and nuclease function. The information of sgRNA and the corresponding target gene information came from previous studies20 . As a transcriptional activator, the inventors characterized the ability of the wild type (dCasMINI (V4)) and mutant (S246R) to activate the reporter system used in the evolution process and the endogenous gene CD2. As a base editor or nuclease, the inventors tested two endogenous genes IFNγ and VEGFA. The plasmid information used in these experiments is detailed in Supplementary Table 1. In all tests, 2 μg of plasmid was transfected into HEK293T cells in one well of a 12-well plate. 12 hours after transfection, the medium was replaced with fresh medium, and 60 hours later, the cells were collected for flow cytometry analysis or sequencing.

MEK1进化实验MEK1 evolution experiments

该系统的设计和该实验的时间线如图56所示。将人源的MEK1 cDNA克隆到repRNA-v4质粒中,并将得到的自主复制RNA电穿孔到表达nsP4的BHK-21细胞(含有ML700质粒的稳定细胞系)。电穿孔后24小时(即第1天),用含有5μg/mL嘌呤霉素的新鲜培养基替换培养基。选择4天后(即第5天),在培养基中加入2μM的Molnupiravir。第7天,向培养基中加入治疗药物(即2μM考比替尼、5μM曲美替尼(trametinib)或50μM司美替尼(selumetinib))。一周后(即第14天),收集细胞进行总RNA提取、测序和突变分析。将cDNA库(包含MEK1基因)克隆到一个载体(pEF1α-MEK1_mutant-BGHpolyA-pEF1α/HTLV-iRFP-NLS)中,将其转化到大肠杆菌中,通过Sanger测序鉴定单个突变体。为了量化MEK1变体的药物敏感性,将400ng含MEK1(野生型或突变体)的质粒和200ng SRE报告质粒(7×SREphCMVmini-NLS-mCherry-BGHpolyA-pEF1α/HTLV-mTagBF P2-NLS)共同转染到24孔板上的HEK293T细胞中,培养基含有2μM考比替尼。转染12小时后,用含有考比替尼的新鲜培养基替换培养基,一天后用流式细胞仪测量荧光强度。The design of the system and the timeline of the experiment are shown in Figure 56. Human MEK1 cDNA was cloned into the repRNA-v4 plasmid, and the resulting self-replicating RNA was electroporated into BHK-21 cells expressing nsP4 (stable cell line containing the ML700 plasmid). 24 hours after electroporation (i.e., day 1), the culture medium was replaced with fresh culture medium containing 5μg/mL puromycin. After 4 days of selection (i.e., day 5), 2μM Molnupiravir was added to the culture medium. On day 7, therapeutic drugs (i.e., 2μM cobimetinib, 5μM trametinib, or 50μM selumetinib) were added to the culture medium. One week later (i.e., day 14), cells were collected for total RNA extraction, sequencing, and mutation analysis. The cDNA library (containing the MEK1 gene) was cloned into a vector (pEF1α-MEK1_mutant-BGHpolyA-pEF1α/HTLV-iRFP-NLS), which was transformed into E. coli, and individual mutants were identified by Sanger sequencing. To quantify the drug sensitivity of MEK1 variants, 400 ng of plasmid containing MEK1 (wild type or mutant) and 200 ng of SRE reporter plasmid (7×SREphCMVmini-NLS-mCherry-BGHpolyA-pEF1α/HTLV-mTagBF P2-NLS) were co-transfected into HEK293T cells in 24-well plates, and the culture medium contained 2 μM cobimetinib. After 12 hours of transfection, the culture medium was replaced with fresh culture medium containing cobimetinib, and the fluorescence intensity was measured by flow cytometry one day later.

KRAS(S17N)进化实验KRAS(S17N) evolution experiment

该系统的设计和该实验的时间线如图61所示。人源的野生型KRAS或KRAS(S17N)的cDNA被克隆到repRNA-v4质粒中,并将得到的自主复制RNA电穿孔到表达nsP4的BHK-21细胞(含有ML700质粒的稳定细胞系)。电穿孔后24小时(即第1天),用含有5μg/mL嘌呤霉素的新鲜培养基替换培养基。选择4天后(即第5天),在培养基中加入2μM的molnupiravir。培养10天后,收集细胞进行总RNA提取、测序和突变分析。将cDNA库(包含KRAS基因)克隆到一个载体(pEF1α-KRAS_mutant-BGHpolyA-pEF1α/HTLViRFP-NLS)中,将其转化到大肠杆菌中,通过Sanger测序进行单个突变体鉴定。为了量化KRAS变体的信号活性,将400ng含KRAS(野生型或突变体)的质粒和200ng SRE报告质粒(7×SRE-phCMVmini-NLS-mCherry-BGHpolyA-pEF1α/HTLV-mTagBF P2-NLS)共同转染到24孔板的HEK293T细胞。转染12小时后,用新鲜培养基替换培养基,一天后用流式细胞仪测量细胞数量和荧光强度。The design of the system and the timeline of the experiment are shown in Figure 61. The cDNA of human wild-type KRAS or KRAS (S17N) was cloned into the repRNA-v4 plasmid, and the resulting self-replicating RNA was electroporated into BHK-21 cells expressing nsP4 (stable cell line containing the ML700 plasmid). 24 hours after electroporation (i.e., day 1), the culture medium was replaced with fresh culture medium containing 5μg/mL puromycin. After 4 days of selection (i.e., day 5), 2μM molnupiravir was added to the culture medium. After 10 days of culture, the cells were collected for total RNA extraction, sequencing, and mutation analysis. The cDNA library (containing the KRAS gene) was cloned into a vector (pEF1α-KRAS_mutant-BGHpolyA-pEF1α/HTLViRFP-NLS), which was transformed into Escherichia coli, and single mutants were identified by Sanger sequencing. To quantify the signaling activity of KRAS variants, 400 ng of plasmid containing KRAS (wild type or mutant) and 200 ng of SRE reporter plasmid (7×SRE-phCMVmini-NLS-mCherry-BGHpolyA-pEF1α/HTLV-mTagBF P2-NLS) were co-transfected into HEK293T cells in 24-well plates. After 12 hours of transfection, the medium was replaced with fresh medium, and the cell number and fluorescence intensity were measured by flow cytometry one day later.

实施例Example

实施例1-工程改造适用于体内进化的自主复制RNAExample 1 - Engineering autonomously replicating RNA suitable for in vivo evolution

来自甲病毒(正链RNA病毒)的RNA复制子不仅被广泛用于表达异源蛋白,如动物细胞和组织中的疫苗抗原37–39,也被用于哺乳动物的合成生物学9,40。然而,它们会诱发强烈的细胞病变效应35,41,扰乱细胞的生理功能,并阻止复制子的长期繁殖。因此,必须通过对甲病毒自主复制RNA的工程改造来提高宿主细胞的兼容性,从而利用这种RNA在体内进行定向进化。RNA replicons from alphaviruses (positive-strand RNA viruses) have been widely used not only to express heterologous proteins such as vaccine antigens in animal cells and tissues37–39 but also in synthetic biology in mammals9,40 . However, they induce strong cytopathic effects35,41 , disrupting cellular physiological functions and preventing long-term propagation of replicons. Therefore, it is necessary to improve host cell compatibility by engineering alphavirus self-replicating RNA so that directed evolution can be performed in vivo using this RNA.

在Sindbis病毒(甲病毒的一种)的衍生复制子的工作基础上,本发明人从两个已证实具有低细胞病变性的复制子版本开始42,43。第一个版本(repRNA-v1)含有非结构蛋白2(nsP2)的突变,而第二个版本(repRNA-v2)含有这个突变以及分别在nsP1-nsP2和nsP2-nsP3的接头肽段(即nsP1和nsP2的倒数第二个残基;图1)上的两个突变。这些复制子在体外转录并电转进入到BHK-21(小仓鼠肾脏成纤维细胞)细胞(详见方法部分的内容)。本发明人发现,与第一个版本相比,第二个版本电转后细胞存活数明显更高(图2),这表明对多聚蛋白(即P123或P1234)处理的扰动大大影响了细胞病变情况42,43。因此,本发明人对这两个连接残基进行了筛选(图3,参见方法部分的内容)。本发明人发现丙氨酸在nsP1的倒数第二个残基上高度富集,而没有氨基酸在nsP2上明显富集(图3和图4),这表明repRNA-v2的单一连接体突变可以改善细胞的兼容性。此外,由于nsP1的倒数第二个残基的完全相同的突变被证明可以干扰病毒的复制42,因此可以合理地推论:宿主细胞的兼容性可能可以通过干扰RNA的复制来改善。Based on work with replicons derived from Sindbis virus (an alphavirus), the inventors started with two versions of the replicon that had been shown to have low cytopathicity. 42, 43 The first version (repRNA-v1) contained a mutation in nonstructural protein 2 (nsP2), while the second version (repRNA-v2) contained this mutation and two mutations in the linker peptides of nsP1-nsP2 and nsP2-nsP3, respectively (i.e., the penultimate residues of nsP1 and nsP2; Figure 1). These replicons were transcribed in vitro and electroporated into BHK-21 (baby hamster kidney fibroblast) cells (see Methods for details). The inventors found that the number of cells surviving electroporation was significantly higher in the second version compared to the first version (Figure 2), indicating that perturbations in the processing of the polyprotein (i.e., P123 or P1234) significantly affected the cytopathicity. 42, 43 Therefore, the inventors screened these two linker residues (Figure 3, see Methods for details). The present inventors found that alanine was highly enriched at the penultimate residue of nsP1, while no amino acid was significantly enriched at nsP2 (Figures 3 and 4), suggesting that a single linker mutation of repRNA-v2 could improve cell compatibility. In addition, since the exact same mutation of the penultimate residue of nsP1 was shown to interfere with viral replication42 , it is reasonable to infer that host cell compatibility may be improved by interfering with RNA replication.

在上述发现的基础上,本发明人试图通过在宿主基因组中表达RNA复制酶nsP4(而不是从RNA本身)来进一步提高宿主细胞的兼容性。这将制约RNA的复制,因为复制反应将受制于有限的复制酶和非自主催化。本发明人于是将一个组成型表达的nsP4整合到基因组中(详见方法),并使用没有nsP4的RNA在相同的残基位置上重新进行建库筛选(图5的a和b)。有趣的是,这次筛选中的细胞比使用repRNA-v2的筛选生长得更快(图6)。此外,丙氨酸在nsP1-nsP2连接体中再次富集,像前一次筛选那样,同样没有氨基酸在另一个连接位置中明显富集(图7)。Based on the above findings, the inventors attempted to further improve the compatibility of host cells by expressing the RNA replicase nsP4 in the host genome (rather than from the RNA itself). This will restrict the replication of RNA because the replication reaction will be subject to limited replicase and non-autonomous catalysis. The inventors then integrated a constitutively expressed nsP4 into the genome (see Methods for details) and re-constructed the library screen at the same residue position using RNA without nsP4 (a and b in Figure 5). Interestingly, the cells in this screen grew faster than the screen using repRNA-v2 (Figure 6). In addition, alanine was enriched again in the nsP1-nsP2 linker, and like the previous screen, no amino acid was significantly enriched in the other linker position (Figure 7).

基于这些筛选,本发明人构建了两个新版本的自主复制RNA:repRNA-v3,它以repRNA-v2为基础,分别含有丙氨酸和异亮氨酸作为nsP1和nsP2的倒数第二个残基;repRNA-v4则含有与repRNA-v3相同的突变,但没有nsP4(因此需要在表达nsP4的细胞中复制)(当构建两个自主复制RNA版本(repRNA-v3和repRNA-v4)时,本发明人试图将相同的自主复制RNA与来自不同位置表达的复制酶进行比较,无论是来自RNA本身(repRNA-v3)还是来自宿主基因组(repRNA-v4)。因此,希望两种版本的RNA包括相同的nsP1和nsP2倒数第二个残基。由于丙氨酸在nsP1-2接头残基的两个筛选中都发生富集,因此本发明人选择丙氨酸作为两个版本的nsP1的倒数第二个残基。相比之下,没有氨基酸在来自任一筛选的nsP2-3接头中发生显著富集。因此,本发明人决定选择这样一种氨基酸(用于nsP2的倒数第二个残基),当使用具有宿主表达的复制酶的版本时,该氨基酸可能会提高宿主相容性,因为该版本是体内进化实验的首选。因此,本发明人选择了异亮氨酸,因为它在其中一个重复筛选中富集)。本发明人将这两个新版本与repRNA-v2进行比较。本发明人发现接头肽段突变确实提高了细胞的兼容性(即repRNA-v3相对于repRNA-v2),并且通过从宿主基因组中表达来限制复制酶的水平大大增强了宿主细胞的兼容性(即repRNA-v4相对于repRNA-v3)(图8和图9)。这些结果表明,当复制酶水平受到限制时,病毒的RNA复制似乎可以被宿主细胞所接受,实现在一周内产生~108个携带自主复制RNA的宿主细胞(图9)。然而,上述测量是在电穿孔后一周进行的,所以RNA被宿主细胞在早期和晚期(即短期和长期稳定性)的接受程度仍然未知。Based on these screens, the inventors constructed two new versions of self-replicating RNA: repRNA-v3, which is based on repRNA-v2 and contains alanine and isoleucine as the penultimate residues of nsP1 and nsP2, respectively; and repRNA-v4, which contains the same mutations as repRNA-v3 but lacks nsP4 (and therefore requires replication in cells expressing nsP4). When constructing two versions of self-replicating RNA (repRNA-v3 and repRNA-v4), the inventors sought to compare the same self-replicating RNA with replicases expressed from different locations, either from the RNA itself (repRNA-v3) or from the host genome (repRNA -v4). Therefore, it was desired that both versions of RNA include the same penultimate residues of nsP1 and nsP2. Since alanine was enriched in both screens of nsP1-2 linker residues, the inventors selected alanine as the penultimate residue of both versions of nsP1. In contrast, no amino acid was significantly enriched in the nsP2-3 linker from either screen. Therefore, the inventors decided to select an amino acid (for the penultimate residue of nsP2) that might improve host compatibility when using a version with host-expressed replicase, as this version is the first choice for in vivo evolution experiments. Therefore, the inventors selected isoleucine because it was enriched in one of the repeated screens). The inventors compared the two new versions with repRNA-v2. The inventors found that the linker peptide mutation did improve cell compatibility (i.e., repRNA-v3 relative to repRNA-v2), and that limiting the level of replicase by expression from the host genome greatly enhanced host cell compatibility (i.e., repRNA-v4 relative to repRNA-v3) (Figures 8 and 9). These results suggest that when replicase levels are limiting, viral RNA replication appears to be accepted by host cells, resulting in ~10 8 host cells carrying autonomously replicating RNA within a week (Figure 9). However, the above measurements were performed one week after electroporation, so the extent to which RNA is accepted by host cells at early and late stages (i.e., short-term and long-term stability) remains unknown.

实施例2-改造后的RNA同时具有短期和长期的稳定性Example 2 - Modified RNA has both short-term and long-term stability

本发明人首先通过测量RNA在进入细胞的早期时间点所表达的EGFP来量化三个自主复制RNA版本(repRNA-v2至v4)的短期稳定性(图10和图11)。对于没有限制复制酶的版本,本发明人发现接头肽段的优化(repRNA-v3)比未优化的版本(repRNA-v2)提高了宿主细胞的兼容性,导致了更高的EGFP阳性细胞的最大占比以及72小时时更高比例的EGFP阳性细胞剩余(图12)。然而,对于这两种RNA,EGFP阳性细胞的比例在电穿孔后3天内下降到10%以下,表明宿主兼容性总体较低。相反,对于复制酶有限的版本(repRNA-v4),EGFP阳性细胞比例在第3天可以保持高达约50%,并显示出对复制酶水平依赖的行为模式(图13)。因此,有限复制酶的复制模式可能实现较低的细胞病理性,以更好地维持连续的RNA复制。从机制层面讲,宿主转录组分析表明,相比于复制酶限制的复制,无复制酶限制的复制(即repRNA-v3)显著上调了干扰素信号通路相关的基因(图14),这是一种已知的病毒诱导的细胞致病性的反映44The inventors first quantified the short-term stability of three autonomously replicating RNA versions (repRNA-v2 to v4) by measuring the EGFP expressed by the RNA at the early time point of entering the cell (Figures 10 and 11). For the version without limiting the replicase, the inventors found that the optimization of the linker peptide segment (repRNA-v3) improved the compatibility of the host cell compared with the unoptimized version (repRNA-v2), resulting in a higher maximum proportion of EGFP-positive cells and a higher proportion of EGFP-positive cells remaining at 72 hours (Figure 12). However, for these two RNAs, the proportion of EGFP-positive cells dropped to less than 10% within 3 days after electroporation, indicating that the host compatibility is generally low. In contrast, for the version with limited replicase (repRNA-v4), the proportion of EGFP-positive cells can be maintained as high as about 50% on the 3rd day, and a behavior pattern dependent on the replicase level is shown (Figure 13). Therefore, the replication mode of the limited replicase may achieve lower cell pathology to better maintain continuous RNA replication. Mechanistically, host transcriptome analysis showed that, compared with replicase-restricted replication, replication without replicase restriction (i.e., repRNA-v3) significantly upregulated genes involved in the interferon signaling pathway (Figure 14), which is a reflection of known virus-induced cellular pathogenicity44 .

随着复制酶水平的降低,RNA的短期稳定性显示出增加的趋势,因为EGFP阳性比例在从EGFP水平达到稳定状态(36小时)到结束(72小时)表现出更稳定的维持状态(图13,图15的左侧)。为了测试复制酶水平的降低与短期稳定性的提高是否具有因果关系,本发明人通过在nsP4前面插入一个终止密码子来创建了一个新的宿主细胞系(指"低复制酶宿主")(图16)。与之前的宿主细胞系(指"高复制酶宿主")相比,进一步降低了复制酶水平。在这个新的宿主细胞中,从RNA(repRNA-v4)中表达的EGFP水平如同预计中比以前的宿主细胞低(比较图17和图13)。最重要的是,在新宿主细胞中,自主复制RNA的短期稳定性确实得到了增强(图15)。这些数据共同表明,复制酶水平的降低导致了RNA复制率的降低(即EGFP水平的降低),从而导致自主复制RNA的短期稳定性增强。As the replicase level decreases, the short-term stability of RNA shows an increasing trend, because the EGFP-positive ratio shows a more stable maintenance state from the time when the EGFP level reaches a steady state (36 hours) to the end (72 hours) (Figure 13, left side of Figure 15). In order to test whether the reduction in replicase levels is causally related to the improvement in short-term stability, the inventors created a new host cell line (referred to as the "low replicase host") by inserting a stop codon in front of nsP4 (Figure 16). Compared with the previous host cell line (referred to as the "high replicase host"), the replicase level was further reduced. In this new host cell, the level of EGFP expressed from RNA (repRNA-v4) was lower than that of the previous host cell as expected (compare Figure 17 and Figure 13). Most importantly, in the new host cell, the short-term stability of the autonomously replicating RNA is indeed enhanced (Figure 15). These data collectively indicate that the reduction in replicase levels leads to a reduction in the RNA replication rate (i.e., a reduction in the EGFP level), which in turn leads to an enhanced short-term stability of the autonomously replicating RNA.

基于这些数据,本发明人构建了一个简单的RNA复制动力学模型,从而提出了稳态RNA水平与复制酶水平成正比的假设。通过量化两个宿主细胞系不同复制酶水平下的稳态RNA(EGFP)水平,本发明人欣慰地发现,对于低复制酶水平,稳态RNA水平与复制酶水平呈线性比例(图18)。然而,对于高复制酶水平(与高复制酶宿主),似乎稳态RNA水平在经过一个线性阶段后达到一个平稳时期(图18),这可能反映了由于RNA诱导的细胞病理学效应决定着宿主细胞对RNA的承载程度。与其相一致的是,当培养时间较长时,只有高复制酶宿主中低复制酶水平的细胞存活(图19),这与低复制酶宿主(图20)明显不同。Based on these data, the inventors have constructed a simple RNA replication kinetic model, thereby proposing the hypothesis that the steady-state RNA level is proportional to the replicase level. By quantifying the steady-state RNA (EGFP) level under different replicase levels of two host cell lines, the inventors are pleased to find that for low replicase levels, the steady-state RNA level is linearly proportional to the replicase level (Figure 18). However, for high replicase levels (with high replicase hosts), it seems that the steady-state RNA level reaches a stable period after a linear stage (Figure 18), which may reflect that the cytopathological effects induced by RNA determine the carrying degree of host cells to RNA. Consistent with this, when the culture time is long, only cells with low replicase levels in high replicase hosts survive (Figure 19), which is significantly different from low replicase hosts (Figure 20).

总之,这些定量数据有助于描述短期内RNA复制维持的过程,并说明了RNA复制的有限复制酶模式的动力学和功能。从动力学上讲,有限复制酶模式在饱和的动力学状态下运行,其稳态水平受到调节,有助于提高宿主的兼容性。在功能上,通过增强宿主的兼容性,这种模式实现在一周内产生~108个细胞(从4×106细胞开始)(图9)。重要的是,RNA(repRNA-v4)可以稳定地繁殖超过一个月(图21,图22),从中RNA在单个细胞中均匀地复制,这是通过对复制中间物--双链RNA(dsRNA)的免疫染色所确认的(图23,图24,详见方法)。通过使用RNA测序进一步量化病毒基因组的水平(详见方法),本发明人估计每个细胞可以承载大约100个RNA分子(图25,本发明人使用bulk poly-A RNA-seq来量化自主复制RNA和内源性基因的表达水平。本发明人发现repRNA-v4(在高复制酶宿主中)的表达水平约为Gapdh基因的60%,这是通过读数映射到nsP1-3的读数来测量的。由于哺乳动物细胞中的GapdhmRNA通常为每个细胞1000个分子3,因此本发明人推断工程改造的自主复制RNA即repRNA-v4在每个细胞中至少具有100个分子的数量级)。从而,多达超过100亿的RNA分子(108个细胞×100个拷贝/细胞)可以随后在活的哺乳动物细胞内被多样化和选择。In summary, these quantitative data help describe the process of RNA replication maintenance in the short term and illustrate the dynamics and function of the limited replicase mode of RNA replication. Kinetically, the limited replicase mode operates in a saturated kinetic state, and its steady-state level is regulated, which helps to improve host compatibility. Functionally, by enhancing host compatibility, this mode achieves the production of ~10 8 cells (starting from 4×10 6 cells) in one week (Figure 9). Importantly, RNA (repRNA-v4) can be stably propagated for more than a month (Figure 21, Figure 22), from which RNA is replicated uniformly in single cells, which is confirmed by immunostaining of replication intermediates-double-stranded RNA (dsRNA) (Figure 23, Figure 24, see Methods for details). By further quantifying the levels of viral genomes using RNA sequencing (see Methods for details), the inventors estimated that each cell could carry approximately 100 RNA molecules (Figure 25, the inventors used bulk poly-A RNA-seq to quantify the expression levels of autonomously replicating RNAs and endogenous genes. The inventors found that the expression level of repRNA-v4 (in high replicase hosts) was approximately 60% of the Gapdh gene, as measured by reads mapped to reads of nsP1-3. Since GapdhmRNA in mammalian cells is typically 1000 molecules per cell3, the inventors inferred that the engineered autonomously replicating RNA, namely repRNA-v4, has at least 100 molecules per cell). Thus, up to more than 10 billion RNA molecules ( 108 cells × 100 copies/cell) can then be diversified and selected within living mammalian cells.

实施例3-通过干预RNA复制精确度实现可控的RNA多样化Example 3 - Controllable RNA diversification by interfering with RNA replication accuracy

本发明人接下来探讨了这个改造过的系统的诱变能力,这可以决定自主复制RNA的进化和适应效率。本发明人首先量化了RNA的基础诱变率,然后试图通过干预复制酶的精确度来调控这一比率。The inventors then explored the mutagenic capacity of this modified system, which can determine the evolution and adaptation efficiency of self-replicating RNA. The inventors first quantified the basic mutagenic rate of RNA and then tried to regulate this rate by intervening in the precision of the replicase.

为了量化复制酶的保真度,本发明人用二代测序来测量RNA在细胞中维持1、3、7、14和21天后的突变率(图26的a和b)。为了建立基线,本发明人对质粒DNA以及体外转录的RNA进行了测序,并发现测序和体外转录可以共同引入总计约0.4个碱基/kb的突变率。一旦进入细胞,RNA上的突变以每天约0.01个碱基/kb的速度积累,21天后达到约0.22个碱基/kb(图26的b,图27),其中包含了四个主要的突变类型(即A到U、A到G、G到A和C到U)(图28)。因此,在长期的RNA复制过程中,复制酶并没有引入很多错误,这与RNA被缓慢复制的情况一致。关键是,如此低的基础突变率为可控的诱变率提供了机会。In order to quantify the fidelity of replicase, the inventors used second generation sequencing to measure the mutation rate of RNA after maintaining 1, 3, 7, 14 and 21 days in cells (a and b of Figure 26). In order to establish a baseline, the inventors sequenced plasmid DNA and in vitro transcribed RNA, and found that sequencing and in vitro transcription can introduce a mutation rate of about 0.4 bases/kb in total together. Once entering the cell, the mutation on RNA accumulates at a speed of about 0.01 bases/kb per day, reaching about 0.22 bases/kb after 21 days (b of Figure 26, Figure 27), which contains four main mutation types (i.e. A to U, A to G, G to A and C to U) (Figure 28). Therefore, in the long-term RNA replication process, replicase does not introduce many errors, which is consistent with the situation that RNA is slowly replicated. The key is that such a low basic mutation rate provides an opportunity for a controllable mutagenesis rate.

为了改变复制酶的保真度,本发明人测量了携带已知会降低精确度的突变体nsP4的宿主细胞中RNA的突变率45。本发明人观察到四种突变类型的突变率只有轻微但有统计学意义上显著的增加,然而这仅仅非常有限地提高了RNA的多样化能力(图29)。本发明人接下来研究了小分子介导的RNA复制酶干预,因为一些治疗性核苷类似物通过降低RNA复制酶的保真度来对抗RNA病毒(例如,用于治疗COVID-19的molnupiravir)46–50。本发明人测试了四种不同的核苷类似物,比较它们消除RNA表达的EGFP信号的能力,当中拥有更强的诱导突变能力的类似物会导致非荧光EGFP突变体更快地积累(图30,图31)。本发明人发现,在这四种药物中,favipiravir和molnupiravir不影响细胞生长(图70),并且在3天内以剂量依赖的方式加入到含有自主复制RNA的宿主细胞的培养基中时,都能剂量依赖性地(图32)有效地减少EGFP信号(图31,图33)。通过量化药物处理整个时间过程中的RNA突变率,本发明人发现这两种药物可以将RNA复制酶的诱变能力提高两个数量级,实现每天高达1个碱基/kb的突变率(图34),这应该能实现基于RNA的合成装置进行快速多样化。重要的是,正交RNA复制系统增强的诱变率很可能不会影响宿主基因组表达的基因,这一点从iRFP信号(与nsP4一起表达)的稳定性和对照细胞系中的EGFP信号(图35)可以看出。To alter the fidelity of the replicase, the inventors measured the mutation rate of RNA in host cells carrying a mutant nsP4 known to reduce accuracy 45 . The inventors observed only a slight but statistically significant increase in the mutation rate of the four mutation types, however this only very limitedly improved the diversification capacity of the RNA ( FIG. 29 ). The inventors next studied small molecule-mediated intervention of the RNA replicase, as some therapeutic nucleoside analogs fight RNA viruses by reducing the fidelity of the RNA replicase (e.g., molnupiravir used to treat COVID-19) 46–50 . The inventors tested four different nucleoside analogs and compared their ability to eliminate the EGFP signal expressed by RNA, among which analogs with stronger ability to induce mutations resulted in faster accumulation of non-fluorescent EGFP mutants ( FIG. 30 , FIG. 31 ). The inventors found that among these four drugs, favipiravir and molnupiravir did not affect cell growth (Figure 70), and when added to the culture medium of the host cell containing the autonomously replicating RNA in a dose-dependent manner within 3 days, the EGFP signal (Figure 31, Figure 33) can be effectively reduced in a dose-dependent manner (Figure 32). By quantifying the RNA mutation rate during the entire time course of drug treatment, the inventors found that these two drugs can improve the mutagenic ability of RNA replicase by two orders of magnitude, achieving a mutation rate of up to 1 base/kb per day (Figure 34), which should be able to achieve rapid diversification based on RNA synthesis devices. Importantly, the enhanced mutagenic rate of the orthogonal RNA replication system is likely not to affect the genes expressed by the host genome, as can be seen from the stability of the iRFP signal (expressed together with nsP4) and the EGFP signal (Figure 35) in the control cell line.

本发明人进一步比较了带有核苷类似物或载体对照的RNA的突变谱。本发明人发现,molnupiravir,但不是favipiravir,通过平均地提高所有四种主要转换类型的突变频率,诱导出一个相对平衡的突变谱(图36)。因此,本发明人选择使用molnupiravir,它可以诱导出相对广泛和平衡的突变谱,并让本发明人实现以可控的方式使RNA多样化。The inventor further compared the mutation spectrum of RNA with nucleoside analogs or carrier control.The inventor found that molnupiravir, but not favipiravir, induces a relatively balanced mutation spectrum (Figure 36) by improving the mutation frequency of all four major conversion types on average.Therefore, the inventor chooses to use molnupiravir, which can induce a relatively extensive and balanced mutation spectrum, and allows the inventor to realize RNA diversification in a controlled manner.

实施例4-基于REPLACE系统的EGFP信号蓝移Example 4 - Blue shift of EGFP signal based on REPLACE system

通过将自主复制RNA的产物与表型联系起来,原则上这些RNA可以发生突变从而产生可被定向选择的多样化表型。因此,本发明人通过REPLACE来探索正交RNA复制系统的效用。通过REPLACE,含有目标蛋白质基因序列的基因可以通过RNA上的亚基因组启动子表达,这些RNA可以被复制、诱变和定向选择。By linking the products of autonomously replicating RNA to phenotypes, in principle, these RNAs can mutate to produce diverse phenotypes that can be directed selected. Therefore, the inventors explored the effectiveness of orthogonal RNA replication systems through REPLACE. Through REPLACE, genes containing target protein gene sequences can be expressed through subgenomic promoters on RNAs, and these RNAs can be replicated, mutated, and directed selected.

本发明人试图通过EGFP信号的颜色改变来测试REPLACE,因为EGFP本身就是一种验证定向进化工具的材料,因为只需一个氨基酸的替换GFP就会变成为BFP51,52,19。具体来说,本发明人将EGFP基因置于自主复制RNA的亚基因组启动子的调控之下(图37),然后培养出含有这种RNA的细胞群(详见方法)。随后在培养基中加入molnupiravir来加速诱变,这一过程持续两天(图38)。此后,对GFP和BFP信号的流式分析显示,有一小部分细胞偏离了对角线,这表明荧光发生了蓝移(图38的中部)。通过细胞分选,本发明人定向选择了显示不同蓝移信号的两个细胞群,对其进行第二轮诱变和筛选。经测序,本发明人观察到垂直群体有一个高度富集的错义突变(r.196U>C,即mRNA的第196个核苷酸从U突变为C),而另一个富集的错义突变(r.608C>U)存在于非对角线群体中(图39,图40)。这两个富集的突变分别与从GFP到BFP或到Sapphire53,54信号的转换有关51(图41)。值得注意的是,尽管两种出现蓝移信号的突变体各只需要突变一个残基就会出现,但导致两个突变体的两个残基相距甚远,而且这两个突变体在第一次分选后很容易被检测到,这证明了REPLACE系统在进化需要多个突变的合成装置方面的可行性。The inventors attempted to test REPLACE by the color change of the EGFP signal, because EGFP itself is a material for verifying the directed evolution tool, because only one amino acid replacement is needed for GFP to become BFP 51,52,19 . Specifically, the inventors placed the EGFP gene under the regulation of the subgenomic promoter of the autonomously replicating RNA (Figure 37), and then cultivated a cell group containing this RNA (see method for details). Subsequently, molnupiravir was added to the culture medium to accelerate mutagenesis, and this process lasted for two days (Figure 38). Thereafter, the flow analysis of GFP and BFP signals showed that a small part of cells deviated from the diagonal, which indicated that blue shift occurred in fluorescence (middle part of Figure 38). By cell sorting, the inventors directed and selected two cell groups showing different blue shift signals, and subjected them to a second round of mutagenesis and screening. After sequencing, the inventors observed that the vertical population had a highly enriched missense mutation (r.196U>C, i.e., the 196th nucleotide of the mRNA mutated from U to C), while another enriched missense mutation (r.608C>U) was present in the non-diagonal population (Figure 39, Figure 40). These two enriched mutations were associated with the conversion of signals from GFP to BFP or to Sapphire 53,54 , respectively 51 (Figure 41). It is worth noting that although the two mutants that appeared blue-shifted signals only needed to mutate one residue each, the two residues that led to the two mutants were far apart, and the two mutants were easily detected after the first sorting, which proved the feasibility of the REPLACE system in evolving synthetic devices that require multiple mutations.

实施例5-利用REPLACE系统改造转录调控蛋白Example 5 - Using the REPLACE system to modify transcriptional regulatory proteins

本发明人接下来试图利用REPLACE来进化合成的转录调控蛋白,包括配体控制的转录因子(TF)和基于Cas(CRISPR相关)蛋白的转录调控蛋白——它们已经在哺乳动物合成电路中被广泛应用7The inventors next sought to use REPLACE to evolve synthetic transcriptional regulators, including ligand-controlled transcription factors (TFs) and Cas (CRISPR-associated) protein-based transcriptional regulators, which have been widely used in mammalian synthetic circuits7 .

本发明人首先构建了可以减少或增强配体敏感性的合成TF。作为REPLACE系统的测试,本发明人的理想方法是选择重现其他定向进化方法已经达成的研究结果16,18:进化TetR以降低其对多西环素(doxycycline)的敏感性。简而言之,携带TetR-VP48的自主复制RNA(即tTA)在相对低剂量的molnupiravir(2μM)环境下连续进行多样化7天(图42的a和b,详见方法)。同时,在2周的时间里,培养基中多西环素和嘌呤霉素(puromycin)的浓度逐渐增加,在此期间,能够与DNA结合(通过抵抗多西环素浓度增加)并驱动嘌呤霉素抗性基因表达的TetR突变体将被富集。本发明人之后分选出表达高水平mCherry信号的细胞,并提取其RNA进行逆转录和测序。令人欣慰的是,本发明人发现了三个富集的错义突变(>5%),其中富集度最高的突变T103P直接位于配体结合口袋,其他两个突变(R80C和E157G)则位于口袋附近55(图43)。使用HEK293T细胞的报告系统,本发明人发现TetR变体(从Sanger测序中确定)随着突变残基的数量从一个增加到三个,对多西环素的抗性(即敏感性降低)不断增加(图44)。总之,这些结果表明,REPLACE能够通过哺乳动物细胞内的自主复制RNA多样化和定向选择来构建新的合成TF。The inventors first constructed synthetic TFs that could reduce or enhance ligand sensitivity. As a test of the REPLACE system, the inventors' ideal approach was to choose to reproduce the research results that other directed evolution methods have achieved16,18 : evolving TetR to reduce its sensitivity to doxycycline. In short, the autonomously replicating RNA carrying TetR-VP48 (i.e., tTA) was continuously diversified for 7 days in the environment of relatively low doses of molnupiravir (2μM) (Figure 42a and b, see methods for details). At the same time, over a period of 2 weeks, the concentrations of doxycycline and puromycin in the culture medium were gradually increased, during which time TetR mutants that were able to bind to DNA (by resisting the increase in doxycycline concentration) and drive the expression of the puromycin resistance gene would be enriched. The inventors then sorted out cells expressing high levels of mCherry signals and extracted their RNA for reverse transcription and sequencing. Reassuringly, the inventors found three enriched missense mutations (>5%), of which the most enriched mutation, T103P, is directly located in the ligand binding pocket, and the other two mutations (R80C and E157G) are located near the pocket 55 (Figure 43). Using a reporter system of HEK293T cells, the inventors found that TetR variants (determined from Sanger sequencing) showed increasing resistance to doxycycline (i.e., decreased sensitivity) as the number of mutated residues increased from one to three (Figure 44). In summary, these results indicate that REPLACE is able to construct new synthetic TFs through autonomously replicating RNA diversification and directed selection in mammalian cells.

本发明人接下来试图改造一个新开发的配体控制的哺乳动物合成TF,VP64-PadR56,其DNA结合能力可以被美国FDA批准的药物(阿魏酸钠,sodium ferulate)关闭。改造目的是降低其配体敏感性或提高其响应的动态范围。为了降低配体的敏感性,本发明人通过RNA多样化然后在配体(阿魏酸钠)存在的情况下用不断增加的嘌呤霉素浓度进行选择,从而定向进化含有VP64-PadR突变体的RNA(图45,详见方法)。通过逆转录和测序从存活细胞群体中提取的RNA,本发明人确定了8个富集的错义突变(图46),其中4个(E28K、E92G、G153S、K166E)位于配体结合口袋附近57。然后,本发明人通过报告系统来量化一代测序得到的突变体的配体敏感性,并发现在结合口袋附近有单个(E92G或G153S)或双(E28K/G153S)突变的突变体表现出敏感性降低的特征(图47)。出乎意料的是,结合袋外的突变本身或与结合口袋附近的突变协同,可以降低(图47)甚至逆转(图48)对阿魏酸钠的敏感性(T55I/Q78P、L72P/K166E、E28K/T55I/Q78P),这意味着配体结合有着潜在变构调节以及两类残基之间的合作互动。The inventors next attempted to modify a newly developed ligand-controlled mammalian synthetic TF, VP64-PadR 56 , whose DNA binding ability can be shut down by a drug approved by the US FDA (sodium ferulate). The purpose of the modification was to reduce its ligand sensitivity or to increase the dynamic range of its response. In order to reduce the sensitivity of the ligand, the inventors directed evolution of RNA containing VP64-PadR mutants by RNA diversification followed by selection with increasing concentrations of puromycin in the presence of the ligand (sodium ferulate) ( FIG. 45 , see methods for details). By reverse transcription and sequencing of RNA extracted from a population of surviving cells, the inventors identified eight enriched missense mutations ( FIG. 46 ), four of which (E28K, E92G, G153S, K166E) were located near the ligand binding pocket 57 . Then, the inventors quantified the ligand sensitivity of the mutants obtained by first-generation sequencing through a reporter system, and found that mutants with single (E92G or G153S) or double (E28K/G153S) mutations near the binding pocket showed reduced sensitivity characteristics (Figure 47). Unexpectedly, mutations outside the binding pocket, by themselves or in concert with mutations near the binding pocket, can reduce (Figure 47) or even reverse (Figure 48) the sensitivity to sodium ferulate (T55I/Q78P, L72P/K166E, E28K/T55I/Q78P), which means that ligand binding has potential allosteric regulation and cooperative interaction between the two types of residues.

接下来,本发明人通过在没有配体的情况下构建具有增强激活的突变体来尝试扩大VP64-PadR的响应动态范围。本发明人首先进行RNA多样化,然后在不添加阿魏酸钠的情况下选择具有增强目标基因激活的RNA(图49,详见方法)。最终本发明人确认了6个富集的错义突变,其中三个(Y25C、E92K、D130G)位于结合口袋附近(图50)。值得注意的是,含有一个或多个结合口袋附近残基突变的PadR变体(Y25C、Y25C/S174N、D130G/S174N、Y25C/E92K、D74V/R86K/D130G/S174N)会在没有配体的情况下亦能表现出增强的目标激活。然而,与野生型相比,反应的动态范围(通过有无配体时目标反应的倍数变化来量化)却降低了(图51)。相反,对于携带远离结合口袋的突变体(D74V、D21G/D74V/M178T)来说,反应的动态范围至少与野生型相当。重要的是,其中一个变体(D21G/D74V/M178T)与野生型(38倍)相比,表现出急剧增强的动态范围(85倍)(图51),而配体亲和力只是略有变化(图52)。Next, the inventors attempted to expand the dynamic range of the response of VP64-PadR by constructing mutants with enhanced activation in the absence of a ligand. The inventors first performed RNA diversification and then selected RNA with enhanced target gene activation without the addition of sodium ferulate (Figure 49, see Methods for details). Finally, the inventors confirmed 6 enriched missense mutations, three of which (Y25C, E92K, D130G) were located near the binding pocket (Figure 50). It is worth noting that PadR variants containing one or more mutations in residues near the binding pocket (Y25C, Y25C/S174N, D130G/S174N, Y25C/E92K, D74V/R86K/D130G/S174N) can also show enhanced target activation in the absence of a ligand. However, compared with the wild type, the dynamic range of the reaction (quantified by the fold change of the target response with or without a ligand) is reduced (Figure 51). In contrast, for mutants carrying residues far from the binding pocket (D74V, D21G/D74V/M178T), the dynamic range of the response was at least comparable to that of the wild type. Importantly, one of the variants (D21G/D74V/M178T) exhibited a dramatically enhanced dynamic range (85-fold) compared to the wild type (38-fold) (Figure 51), with only a slight change in ligand affinity (Figure 52).

在确立了REPLACE具有改造配体控制的哺乳动物合成TF的能力后,本发明人进一步寻求进化基于Cas的转录调控蛋白。小型化的Cas12f蛋白,CasMINI20,长度约为500个残基,可以被便捷地递送进入细胞,以前是根据理性设计的原则20改造的。本发明人希望通过REPLACE来进一步提高此蛋白的转录调控能力。本发明人对携带内切酶失活的CasMINI-v420(dCasMINI-VPR)的自主复制RNA进行了多样化和筛选(图67和68)。令人意外的是,在两次独立的实验中,本发明人只确认了一个富集的突变(S246R)(图69)。通过报告系统对转录激活能力进行定量,本发明人发现相比于亲本,S246R变体对于含有TetO的启动子(进化实验中用到的)的激活能力提高了约30%(图53),并且其利用一个sgRNA激活一个内源基因(CD2)的能力提高了约20%(图54)。本发明人进一步发现,进化的突变体并没有导致相关的碱基编辑能力和核酸酶的活性明显增强(图55),这可能是因为转录调控活性可能与碱基编辑或DNA切割的活性不相关20After establishing that REPLACE has the ability to transform ligand-controlled mammalian synthetic TF, the inventors further sought to evolve Cas-based transcriptional regulatory proteins. Miniaturized Cas12f protein, CasMINI 20 , is about 500 residues in length and can be easily delivered into cells. It was previously transformed according to the principle of rational design 20. The inventors hope to further improve the transcriptional regulatory ability of this protein through REPLACE. The inventors have diversified and screened the autonomously replicating RNA of CasMINI-v4 20 (dCasMINI-VPR) carrying endonuclease inactivation (Figures 67 and 68). Surprisingly, in two independent experiments, the inventors only confirmed one enriched mutation (S246R) (Figure 69). By quantifying the transcriptional activation ability through a reporter system, the inventors found that compared with the parent, the S246R variant had an activation ability of about 30% for a promoter containing TetO (used in the evolution experiment) (Figure 53), and its ability to activate an endogenous gene (CD2) using an sgRNA was increased by about 20% (Figure 54). The inventors further found that the evolved mutants did not lead to a significant increase in the related base editing ability and nuclease activity (Figure 55), which may be because the transcriptional regulatory activity may not be related to the activity of base editing or DNA cutting 20 .

这些结果共同证明了REPLACE在构建配体控制的或基于Cas的哺乳动物合成转录调控蛋白方面的能力和通用性,说明此系统具有改善哺乳动物合成生物学应用中其他TF的功能。Together, these results demonstrate the power and versatility of REPLACE for constructing ligand-controlled or Cas-based synthetic transcriptional regulatory proteins in mammals, suggesting that this system has the potential to improve the functionality of other TFs in mammalian synthetic biology applications.

实施例6-携带MEK1的自主复制RNA实现达尔文式适应 Example 6 - Self-replicating RNA carrying MEK1 achieves Darwinian adaptation

通过RNA复制系统的正交性,REPLACE系统实现了一种连续的运行模式——RNA可以自主地跨代复制,为能够以达尔文式适应的方式来构建细胞开辟了途径(即通过适应性选择)。因此,本发明人接下来测试了哺乳动物细胞是否可以利用REPLACE来进化具有重要生理学意义的基因,从而自主地适应新环境。Through the orthogonality of the RNA replication system, the REPLACE system achieves a continuous mode of operation - RNA can replicate autonomously across generations, opening up a way to build cells in a Darwinian adaptive manner (i.e., through adaptive selection). Therefore, the inventors next tested whether mammalian cells can use REPLACE to evolve genes with important physiological significance, thereby autonomously adapting to new environments.

本发明人首先探寻携带人源MEK1基因的自主复制RNA的细胞是否能适应含有MEK1变构抑制剂的环境。由于已经确认MEK1的体内或体外诱变可以产生MEK1的抗抑制剂突变体58,19,所以原则上,细胞可以利用自主复制RNA的可演化性在抑制剂存在的环境下生存下来。于是本发明人用含有亚基因组启动子表达的野生型MEK1基因的repRNA-v4来电转细胞(第0天)(图56,详见方法)。从第5天开始向培养物中加入molnupiravir,以使RNA多样化。然后从第7天开始,用三种独立的MEK1变构抑制剂向细胞施加生存压力,并在第14天收集细胞进行测序。在7天的时间里,RNA的多样化和选择在抑制剂环境下同时进行,实现了达尔文式的适应(图56)。The inventors first explored whether cells carrying self-replicating RNA of the human MEK1 gene could adapt to an environment containing MEK1 allosteric inhibitors. Since it has been confirmed that in vivo or in vitro mutagenesis of MEK1 can produce inhibitor-resistant mutants of MEK158,19 , in principle, cells can use the evolvability of self-replicating RNA to survive in the presence of inhibitors. So the inventors used repRNA-v4 containing the wild-type MEK1 gene expressed by a subgenomic promoter to electrotransfect cells (day 0) (Figure 56, see methods for details). From the 5th day, molnupiravir was added to the culture to diversify the RNA. Then, starting from the 7th day, three independent MEK1 allosteric inhibitors were used to apply survival pressure to the cells, and the cells were collected for sequencing on the 14th day. Over a period of 7 days, RNA diversification and selection were carried out simultaneously in the inhibitor environment, achieving Darwinian adaptation (Figure 56).

有趣的是,本发明人发现存活的细胞含有高度富集MEK1突变的RNA(即在一个或多个生物学重复中>5%),并且有几个突变在三种抑制剂中均有富集(图57,图58),这表明对MEK1的变构抑制剂的达尔文适应可以通过保守的机制发生。从结构上看,突变位于不同功能域内59,58,60(图59):抑制剂结合口袋(L115P、V211D、L215P),负调节域(R47Q、Q56P、K57N),可能干扰抑制剂功能的残基(H119Y)以及核输出信号(E39A)61。其中许多是以前在MEK1的诱变筛选58中发现过的。Sanger测序使本发明人能够鉴定出每个MEK1突变体,它们平均带有1到3个突变。Interestingly, the inventors found that surviving cells contained RNA highly enriched for MEK1 mutations (i.e., >5% in one or more biological replicates), and several mutations were enriched for all three inhibitors (Figures 57, 58), suggesting that Darwinian adaptation to allosteric inhibitors of MEK1 can occur through conserved mechanisms. Structurally, the mutations were located in different functional domains59,58,60 (Figure 59): the inhibitor binding pocket (L115P, V211D, L215P), the negative regulatory domain (R47Q, Q56P, K57N), residues that may interfere with inhibitor function (H119Y), and the nuclear export signal (E39A) 61 . Many of these were previously discovered in mutagenesis screens for MEK158 . Sanger sequencing allowed the inventors to identify each MEK1 mutant, which carried an average of 1 to 3 mutations.

本发明人接下来使用荧光报告系统来测量MEK1活性(即反映MAPK/ERK信号传导活性的SRE报告系统19,62)从而确认不同突变体对考比替尼(Cobimetinib)的抗性。本发明人发现,在抑制剂存在的情况下,除了H119Y突变外,所有的突变体都显示出比野生型更强的报告活性(即增强的抑制剂抗性)(图60)。令人困惑的是,与单个突变体(L115P和V211D)相比,在抑制剂结合口袋中含有两个突变体时(L115P/V211D)表现出的基础信号活性大大降低,这表明变构口袋中的残基之间有着相互作用(图60)。耐人寻味的是,抑制剂结合口袋和负调控域的联合突变(Q56P/V211D和R47Q/K57N/V211D)与只有结合口袋突变的突变体相比,不论在有无抑制剂的情况下,MEK1的活性都急剧增强(图60)。对于其中一个突变体(Q56P/V211D),抑制剂甚至增加了其活性(而不是抑制)(图60)。这些结果强调了REPLACE在细胞适应MEK1的抑制过程中穿越MEK1的复杂序列功能图谱的能力。The inventors then used a fluorescent reporter system to measure MEK1 activity (i.e., an SRE reporter system that reflects MAPK/ERK signaling activity 19,62 ) to confirm the resistance of different mutants to cobimetinib. The inventors found that in the presence of an inhibitor, all mutants except the H119Y mutation showed stronger reporter activity than the wild type (i.e., enhanced inhibitor resistance) ( FIG. 60 ). Puzzlingly, the basal signaling activity exhibited when two mutants were contained in the inhibitor binding pocket (L115P/V211D) was greatly reduced compared to single mutants (L115P and V211D), indicating that there is an interaction between the residues in the allosteric pocket ( FIG. 60 ). Interestingly, the combined mutations of the inhibitor binding pocket and the negative regulatory domain (Q56P/V211D and R47Q/K57N/V211D) dramatically enhanced the activity of MEK1 in the presence or absence of an inhibitor compared to mutants with only the binding pocket mutation ( FIG. 60 ). For one of the mutants (Q56P/V211D), the inhibitor even increased its activity (rather than inhibiting it) (Figure 60). These results highlight the ability of REPLACE to traverse the complex sequence-function landscape of MEK1 during cellular adaptation to MEK1 inhibition.

实施例7-利用REPLACE系统逃脱显性失活的KRAS基因 Example 7 - Escape from the dominant negative KRAS gene using the REPLACE system

在确定了REPLACE能够使细胞适应外在的挑战(即小分子抑制剂)后,本发明人接下来开始研究配备了自主复制RNA的细胞是否以及如何适应细胞内在的挑战。本发明人选择了一个原癌基因,即KRAS,它是癌症中最频繁的突变基因之一63。除了基于小分子的癌症KRAS靶向治疗外,研究者们还提出并测试了显性失活基因疗法64,65。通过引入一个显性失活的KRAS等位基因(S17N)来抑制致癌的KRAS活性(和细胞增殖)66。原则上,细胞可以通过逆转单一突变(S17N)而对这种治疗产生抗性,然而还有其他潜在的途径来赋予细胞同样的抗性。Having established that REPLACE enables cells to adapt to external challenges (i.e., small molecule inhibitors), the inventors next set out to investigate whether and how cells equipped with autonomously replicating RNA could adapt to cell-intrinsic challenges. The inventors chose a proto-oncogene, KRAS, which is one of the most frequently mutated genes in cancer63 . In addition to small molecule-based cancer KRAS-targeted therapies, researchers have proposed and tested dominant-negative gene therapies64,65 . Oncogenic KRAS activity (and cell proliferation) is inhibited by introducing a dominant-negative KRAS allele (S17N) 66 . In principle, cells could become resistant to this treatment by reversing a single mutation (S17N), but there are other potential pathways to confer similar resistance.

因此,本发明人试图研究携带KRAS(S17N)等位基因的自主复制RNA的细胞是否以及如何通过达尔文适应来克服其对KRAS活性的(细胞内在的)抑制(图61)。作为对照,本发明人还进化了携带野生型KRAS的自主复制RNA的细胞(图61)。RNA首先在含有molnupiravir的细胞培养基中连续多样化9天(图61,详见方法),在此期间,KRAS活性增加和增殖增强的细胞应该比KRAS活性受到抑制的细胞生存优势更大。通过对存活群体的RNA进行测序,本发明人在KRAS(S17N)上发现了六个野生型KRAS上没有的富集的错义突变(图62,图63)。有趣的是,本发明人发现了一个富集的突变将天冬酰胺(asparagine)N17改变为一个羟基氨基酸即苏氨酸(threonine,N17T),而不是野生型中变为的丝氨酸(serine,N17S)(图62)。由于丝氨酸或苏氨酸都对对鸟嘌呤核苷酸的结合至关重要——它们可以通过羟基与Mg2+离子协调67,因此N17T突变可能足以逆转KRAS(S17N)的显性失活表型的影响68。而其他几个突变位于重要的功能区域69(图64),包括负责效应器参与的开关I区(Y32C)70和介导关键变构相互作用的疏水枢纽(L79F)71。然而,仍然难以定论哪一个突变在功能上与摆脱显性失活的KRAS有关。Therefore, the inventors attempted to study whether and how cells carrying autonomously replicating RNA of the KRAS (S17N) allele overcome their (cell-intrinsic) inhibition of KRAS activity through Darwinian adaptation (Figure 61). As a control, the inventors also evolved cells carrying autonomously replicating RNA of wild-type KRAS (Figure 61). RNA was first continuously diversified for 9 days in a cell culture medium containing molnupiravir (Figure 61, see method for details), during which cells with increased KRAS activity and enhanced proliferation should have a greater survival advantage than cells with suppressed KRAS activity. By sequencing the RNA of the surviving population, the inventors found six enriched missense mutations on KRAS (S17N) that were not found on wild-type KRAS (Figure 62, Figure 63). Interestingly, the inventors found an enriched mutation that changed asparagine (asparagine) N17 to a hydroxy amino acid, threonine (N17T), rather than serine (serine, N17S) (Figure 62) that became in the wild type. Since either serine or threonine is essential for binding to guanine nucleotides—they can coordinate Mg2 + ions through their hydroxyl groups67 —the N17T mutation may be sufficient to reverse the effects of the dominant-negative phenotype of KRAS(S17N) 68 . Several other mutations are located in important functional regions69 (Figure 64), including the switch I region (Y32C) responsible for effector engagement70 and a hydrophobic hub (L79F) mediating key allosteric interactions71 . However, it remains difficult to determine which mutation is functionally responsible for escaping the dominant-negative KRAS.

因此,本发明人使用SRE报告系统来量化每个KRAS突变体(最多包含四个突变)的活性,同时表征它们对细胞增殖的影响。作为对照,本发明人发现与显性失活突变体(S17N)相比,野生型KRAS的信号活性明显增强,而众所周知的致癌性KRAS突变,G12C,也导致了更高的活性(图65的顶部)。与活性读数一致的是,野生型KRAS和KRAS(G12C)与KRAS(S17N)相比都能诱导更快的细胞增殖,而这两个变体本身在细胞增殖方面是没有区别的(图65的底部)。然后本发明人测试了由KRAS(S17N)演变而来的已确认的突变体,发现N17T突变体逆转了显性失活效应,因为它导致了明显高于野生型的活性水平,以及与野生型相当的细胞增殖率(图65)。另一个突变,L79F,进一步增强了信号活性,但没有增强细胞增殖(图65)。这些结果表明,与野生型KRAS的S17相比,T17能更好地与Mg2+离子协调,而更高的信号活性可能不会导致细胞增殖的增强。对于其他没有N17T突变的突变体,虽然它们都使KRAS信号活性在统计学意义上增强,但没有一个引起可观的增殖优势(图66),这意味着这些突变体在摆脱显性失活抑制期间可能赋予了细胞其他类型的生存优势。Therefore, the inventors used the SRE reporter system to quantify the activity of each KRAS mutant (up to four mutations) and characterize their effects on cell proliferation. As a control, the inventors found that the signaling activity of wild-type KRAS was significantly enhanced compared to the dominant negative mutant (S17N), and the well-known oncogenic KRAS mutation, G12C, also resulted in higher activity (top of Figure 65). Consistent with the activity readings, both wild-type KRAS and KRAS (G12C) induced faster cell proliferation compared to KRAS (S17N), while the two variants themselves were indistinguishable in terms of cell proliferation (bottom of Figure 65). The inventors then tested confirmed mutants evolved from KRAS (S17N) and found that the N17T mutant reversed the dominant negative effect because it resulted in significantly higher activity levels than the wild type, as well as cell proliferation rates comparable to the wild type (Figure 65). Another mutation, L79F, further enhanced signaling activity, but did not enhance cell proliferation (Figure 65). These results suggest that T17 can better coordinate with Mg 2+ ions than S17 of wild-type KRAS, and the higher signaling activity may not lead to enhanced cell proliferation. For other mutants without N17T mutation, although they all enhanced KRAS signaling activity statistically, none of them caused a significant proliferation advantage (Figure 66), which means that these mutants may confer other types of survival advantages to cells during the period of escape from dominant negative inhibition.

总之,这些结果表明,细胞能够通过REPLACE介导的达尔文适应来摆脱显性失活KRAS的影响。Together, these results suggest that cells are able to escape the effects of dominant-negative KRAS through REPLACE-mediated Darwinian adaptation.

实施例8-讨论Example 8 - Discussion

合成生物学经常被与电气工程等学科72相比,因为它们都使用工程原理来构建按设计运行的通路和系统。然而,生物系统本质上是复杂的且可自然进化的,这就要求合成生物学的理论和工具超越传统学科的范围,才能实现构建的生物系统的可进化性25。为了实现这一目标,本发明人开发了REPLACE:RNA在哺乳动物细胞内正交地复制、持续进化和稳定繁殖。REPLACE不仅可以直接在哺乳动物系统中实现与哺乳动物兼容的生物大分子工程改造,而且还可以为哺乳动物细胞提供合成的可进化性,从而使工程细胞可以利用自主复制RNA的可进化性来适应新环境。Synthetic biology is often compared to disciplines such as electrical engineering72 because both use engineering principles to construct pathways and systems that operate as designed. However, biological systems are inherently complex and naturally evolvable, which requires the theory and tools of synthetic biology to go beyond the scope of traditional disciplines to achieve the evolvability of constructed biological systems25 . To achieve this goal, the inventors developed REPLACE: RNA orthogonally replicated, continuously evolved, and stably propagated in mammalian cells. REPLACE not only enables the engineering of mammalian-compatible biomacromolecules directly in mammalian systems, but also provides synthetic evolvability to mammalian cells, allowing engineered cells to adapt to new environments using the evolvability of self-replicating RNA.

与现有的定向进化方法相比,REPLACE的一个关键的概念和技术进步在于调整和设计RNA病毒复制系统以适应哺乳动物细胞中的体内定向进化。虽然病毒以前曾辅助过生物大分子的定向进化73,74,16,18,但REPLACE从根本上说是不同的,因为只有部分病毒基因组完全地在细胞内繁殖,而且不产生活的病毒。与会在复制过程中积累普遍突变75,76的RNA病毒(如SARS-CoV-2)相比,REPLACE系统中的Sindbis病毒衍生的RNA是在复制酶有限的模式和饱和动力学下复制,以确保低水平的复制和基础错误积累,为从化学上控制RNA复制酶的精确度(以及由此产生的RNA诱变)奠定了基础。与天然的RNA病毒相比,本发明人设计的自主复制RNA的突变主要来自对RNA复制酶的扰动,而不是来自稳态(无扰动)复制。这一发现也表明了广泛使用诱变性核苷类似物来治疗COVID-19的潜在不利影响49,77——这是一把双刃剑,可能会通过诱导突变和促进病毒的快速进化而大大加快新病毒变种的出现。Compared to existing directed evolution methods, a key conceptual and technical advance of REPLACE lies in adapting and engineering the RNA virus replication system to in vivo directed evolution in mammalian cells. While viruses have previously assisted in the directed evolution of biomacromolecules73,74,16,18 , REPLACE is fundamentally different because only a portion of the viral genome is fully propagated intracellularly and no live virus is produced. In contrast to RNA viruses (such as SARS-CoV-2) that accumulate widespread mutations75,76 during replication, the Sindbis virus-derived RNA in the REPLACE system replicates in a replicase-limited mode and with saturating kinetics to ensure low levels of replication and base error accumulation, laying the foundation for chemically controlling the precision of the RNA replicase (and the resulting RNA mutagenesis). In contrast to natural RNA viruses, mutations in the autonomously replicating RNAs designed by the inventors arise primarily from perturbations to the RNA replicase rather than from steady-state (undisturbed) replication. This finding also suggests a potential adverse effect of widespread use of mutagenic nucleoside analogs to treat COVID- 1949,77 – a double-edged sword that could significantly accelerate the emergence of new viral variants by inducing mutations and promoting rapid viral evolution.

REPLACE系统的多功能性使本发明人能够利用定向选择来改造功能性生物分子,包括合成的转录调节蛋白,并使细胞在面临挑战时能够进行达尔文适应。这些成功的应用突出了REPLACE的三个显著特点。第一,本发明人的基于RNA的可进化系统在很大程度上与宿主细胞正交,并独特地实现了宿主细胞内RNA装置的体内进化(与体外进化不同78,79)。每个细胞可容纳约100个拷贝,形成一个庞大的突变体库,以确保各种各样的进化实验。第二,本发明人的系统为多样化和选择步骤提供了灵活性——多样化不仅可以赋予相对广泛的突变谱,并且是完全可控的;选择可以是定向的或是基于适应性的;这两个步骤甚至可以同时进行,以实现连续进化。第三,基于自主复制RNA的装置提供了一种简单而强大的手段来使哺乳动物细胞能够完成适应,传统的合成生物学工具是很难实现的、复杂的和适应性的任务。The versatility of the REPLACE system enabled the inventors to use directed selection to engineer functional biomolecules, including synthetic transcriptional regulators, and to enable Darwinian adaptation of cells in response to challenges. These successful applications highlight three notable features of REPLACE. First, our RNA-based evolvable system is largely orthogonal to host cells and uniquely enables in vivo evolution of RNA machinery within host cells (as distinct from in vitro evolution78,79 ). Each cell can accommodate approximately 100 copies, forming a large mutant library to ensure a wide variety of evolutionary experiments. Second, our system provides flexibility for both diversification and selection steps—diversification can not only confer a relatively broad mutation spectrum, but is also fully controllable; selection can be directed or fitness-based; and the two steps can even be performed simultaneously to achieve continuous evolution. Third, self-replicating RNA-based machinery provides a simple and powerful means to enable mammalian cells to accomplish adaptation, a task that is difficult, complex, and adaptive for traditional synthetic biology tools.

本发明人考虑了REPLACE有着广泛的应用,包括哺乳动物生物大分子工程改造、合成生物学和发育生物学(例如,用于追踪细胞系)。为了进一步扩大其应用范围,它可以在几个方面进行改进,包括筛选方法(如纳入负选择)、目标模式(如进化出功能性RNA)和细胞兼容性(如进一步减少细胞病理学和扩展到其他细胞类型)。The inventors consider that REPLACE has a wide range of applications, including mammalian biomacromolecule engineering, synthetic biology, and developmental biology (e.g., for tracking cell lines). To further expand its scope of application, it can be improved in several aspects, including screening methods (e.g., incorporating negative selection), target modes (e.g., evolving functional RNA), and cell compatibility (e.g., further reducing cell pathology and expanding to other cell types).

广泛而言,本发明人开发的适应性RNA是可进化的,这将促进哺乳动物合成RNA装置的改造和应用形成新模式。目前基于RNA的治疗的热潮32,33,80可能会促进未来可进化和适应性的基于RNA的治疗装置的发展,这些装置可以直接输送到活体动物组织的细胞中,以应对不可预见的挑战。Broadly speaking, the adaptive RNAs developed by the inventors are evolvable, which will promote new paradigms for the engineering and application of synthetic RNA devices in mammals. The current boom in RNA-based therapeutics32,33,80 may promote the development of future evolvable and adaptable RNA-based therapeutic devices that can be delivered directly into cells in living animal tissues to address unforeseen challenges.

参考文献References

1.Hendel,S.J.&Shoulders,M.D.Directed evolution inmammaliancells.Nat.Methods 18,346–357(2021).1. Hendel, S. J. & Shoulders, M. D. Directed evolution of inmammalian cells. Nat. Methods 18, 346–357 (2021).

2.Majors,B.S.,Chiang,G.G.&Betenbaugh,M.J.Protein andGenome Evolutionin Mammalian Cells for Biotechnology Applications.Mol.Biotechnol.42,216–223(2009).2. Majors, B.S., Chiang, G.G. & Betenbaugh, M.J. Protein and Genome Evolution in Mammalian Cells for Biotechnology Applications. Mol. Biotechnol. 42, 216–223 (2009).

3.Morrison,M.S.,Podracky,C.J.&Liu,D.R.The developingtoolkit ofcontinuous directed evolution.Nat.Chem.Biol.16,610–619(2020).3. Morrison, M.S., Podracky, C.J. & Liu, D.R. The developing toolkit of continuous directed evolution. Nat. Chem. Biol. 16, 610–619 (2020).

4.Wang,Y.et al.Directed Evolution:Methodologies andApplications.Chem.Rev.121,12384–12444(2021).4. Wang, Y. et al. Directed Evolution: Methodologies and Applications. Chem. Rev. 121, 12384–12444 (2021).

5.Molina,R.S.et al.In vivo hypermutation and continuousevolution.Nat.Rev.Methods Primer 2,1–22(2022).5. Molina, R.S. et al. In vivo hypermutation and continuousevolution. Nat. Rev. Methods Primer 2, 1–22 (2022).

6.Purnick,P.E.M.&Weiss,R.The second wave of synthetic biology:frommodules to systems.Nat.Rev.Mol.Cell Biol.10,410–422(2009).6. Purnick, P.E.M. & Weiss, R. The second wave of synthetic biology: from modules to systems. Nat. Rev. Mol. Cell Biol. 10, 410–422 (2009).

7.Lienert,F.,Lohmueller,J.J.,Garg,A.&Silver,P.A.Syntheticbiology inmammalian cells:next generation research tools andtherapeutics.Nat.Rev.Mol.Cell Biol.15,95–107(2014).7. Lienert, F., Lohmueller, J.J., Garg, A. & Silver, P.A. Syntheticbiology inmammalian cells: next generation research tools and therapeutics. Nat. Rev. Mol. Cell Biol. 15, 95–107 (2014).

8.Church,G.M.,Elowitz,M.B.,Smolke,C.D.,Voigt,C.A.&Weiss,R.Realizingthe potential of synthetic biology.Nat.Rev.Mol.Cell Biol.15,289–294(2014).8. Church, G.M., Elowitz, M.B., Smolke, C.D., Voigt, C.A. & Weiss, R. Realizing the potential of synthetic biology. Nat. Rev. Mol. Cell Biol. 15, 289–294 (2014).

9.Tan,X.,Letendre,J.H.,Collins,J.J.&Wong,W.W.Syntheticbiology in theclinic:engineering vaccines,diagnostics,and therapeutics.Cell 184,881–898(2021).9. Tan,

10.Gallup,O.,Ming,H.&Ellis,T.Ten future challenges forsyntheticbiology.Eng.Biol.5,51–59(2021).10. Gallup, O., Ming, H. & Ellis, T. Ten future challenges for synthetic biology. Eng. Biol. 5, 51–59 (2021).

11.Azam,M.,Latek,R.R.&Daley,G.Q.Mechanisms ofAutoinhibition and STI-571/Imatinib Resistance Revealed by Mutagenesisof BCR-ABL.Cell 112,831–843(2003).11. Azam, M., Latek, R.R. & Daley, G.Q. Mechanisms of Autoinhibition and STI-571/Imatinib Resistance Revealed by Mutagenesis of BCR-ABL. Cell 112, 831–843 (2003).

12.Wang,L.,Jackson,W.C.,Steinbach,P.A.&Tsien,R.Y.Evolutionof newnonantibody proteins via iterative somatic hypermutation.Proc.Natl.Acad.Sci.101,16745–16749(2004).12. Wang, L., Jackson, W.C., Steinbach, P.A. & Tsien, R.Y. Evolution of new nonantibody proteins via iterative somatic hypermutation. Proc. Natl. Acad. Sci. 101, 16745–16749 (2004).

13.Peck,S.H.,Chen,I.&Liu,D.R.Directed Evolution of aSmall-Molecule-Triggered Intein with Improved Splicing Properties inMammalianCells.Chem.Biol.18,619–630(2011).13. Peck, S. H., Chen, I. & Liu, D. R. Directed Evolution of a Small-Molecule-Triggered Intein with Improved Splicing Properties in Mammalian Cells. Chem. Biol. 18, 619–630 (2011).

14.Hess,G.T.et al.Directed evolution using dCas9-targetedsomatichypermutation in mammalian cells.Nat.Methods 13,1036–1042(2016).14.Hess, G.T. et al. Directed evolution using dCas9-targetedsomatichypermutation in mammalian cells. Nat. Methods 13, 1036–1042 (2016).

15.Ma,Y.et al.Targeted AID-mediated mutagenesis(TAM)enablesefficientgenomic diversification in mammalian ce 846 lls.Nat.Methods 13,1029–1035(2016).15.Ma,Y.et al.Targeted AID-mediated mutagenesis(TAM)enables efficientgenomic diversification in mammalian ce 846 lls.Nat.Methods 13,1029–1035(2016).

16.Berman,C.M.et al.An Adaptable Platform for DirectedEvolution inHuman Cells.J.Am.Chem.Soc.140,18093–18103(2018).16. Berman, C.M. et al. An Adaptable Platform for Directed Evolution in Human Cells. J. Am. Chem. Soc. 140, 18093–18103 (2018).

17.Piatkevich,K.D.et al.A robotic multidimensional directedevolutionapproach applied to fluorescent voltage reporters.Nat.Chem.Biol.14,352–360(2018).17. Piatkevich, K.D. et al. A robotic multidimensional directedevolution approach applied to fluorescent voltage reporters. Nat. Chem. Biol. 14, 352–360 (2018).

18.English,J.G.et al.VEGAS as a Platform for Facile DirectedEvolutionin Mammalian Cells.Cell 178,748-761.e17(2019).18.English,J.G.et al.VEGAS as a Platform for Facile DirectedEvolutionin Mammalian Cells.Cell 178,748-761.e17(2019).

19.Chen,H.et al.Efficient,continuous mutagenesis in human cellsusinga pseudo-random DNA editor.Nat.Biotechnol.38,165–168(2020).19. Chen, H. et al. Efficient, continuous mutagenesis in human cells using a pseudo-random DNA editor. Nat. Biotechnol. 38, 165–168 (2020).

20.Xu,X.et al.Engineered miniature CRISPR-Cas system formammaliangenome regulation and editing.Mol.Cell 81,4333-4345.e4(2021).20. Xu, X. et al. Engineered miniature CRISPR-Cas system formammaliangenome regulation and editing. Mol. Cell 81, 4333-4345.e4 (2021).

21.Weber,W.&Fussenegger,M.Emerging biomedical applicationsofsynthetic biology.Nat.Rev.Genet.13,21–35(2012).21. Weber, W. & Fussenegger, M. Emerging biomedical applications of synthetic biology. Nat. Rev. Genet. 13, 21–35 (2012).

22.Lim,W.A.&June,C.H.The Principles of Engineering ImmuneCells toTreat Cancer.Cell 168,724–740(2017).22. Lim, W. A. & June, C. H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 168, 724–740 (2017).

23.Sedlmayer,F.,Aubel,D.&Fussenegger,M.Synthetic gene circuitsfor thedetection,elimination and prevention of disease.Nat.Biomed.Eng.2,399–415(2018).23. Sedlmayer, F., Aubel, D. & Fussenegger, M. Synthetic gene circuits for the detection, elimination and prevention of disease. Nat. Biomed. Eng. 2, 399–415 (2018).

24.Ruder,W.C.,Lu,T.&Collins,J.J.Synthetic Biology Moving intotheClinic.Science 333,1248–1252(2011).24. Ruder, W.C., Lu, T. & Collins, J.J. Synthetic Biology Moving into the Clinic. Science 333, 1248–1252 (2011).

25.Castle,S.D.,Grierson,C.S.&Gorochowski,T.E.Towards anengineeringtheory of evolution.Nat.Commun.12,3326(2021).25. Castle, S. D., Grierson, C. S. & Gorochowski, T. E. Towards an engineering theory of evolution. Nat. Commun. 12, 3326 (2021).

26.Kheir Gouda,M.,Manhart,M.&Balázsi,G.Evolutionary regainof lostgene circuit function.Proc.Natl.Acad.Sci.116,25162–25171(2019).26. Kheir Gouda, M., Manhart, M. & Balázsi, G. Evolutionary regain of lostgene circuit function. Proc. Natl. Acad. Sci. 116, 25162–25171 (2019).

27.Lezia,A.,Csicsery,N.&Hasty,J.Design,mutate,screen:Multiplexedcreation and arrayed screening of synchronized genetic clocks.Cell Syst.13,365-375.e5(2022).27. Lezia, A., Csicsery, N. & Hasty, J. Design, mutate, screen: Multiplexed creation and arrayed screening of synchronized genetic clocks. Cell Syst. 13, 365-375.e5 (2022).

28.Cobb,R.E.,Sun,N.&Zhao,H.Directed Evolution as a PowerfulSyntheticBiology Tool.Methods San Diego Calif 60,81–90(2013).28. Cobb, R.E., Sun, N. & Zhao, H. Directed Evolution as a Powerful Synthetic Biology Tool. Methods San Diego Calif 60, 81–90 (2013).

29.Haseltine,E.L.&Arnold,F.H.Synthetic gene circuits:designwithdirected evolution.Annu.Rev.Biophys.Biomol.Struct.36,1–19(2007).29. Haseltine, E.L. & Arnold, F.H. Synthetic gene circuits: design with directed evolution. Annu. Rev. Biophys. Biomol. Struct. 36, 1–19 (2007).

30.Yokobayashi,Y.,Weiss,R.&Arnold,F.H.Directed evolution of ageneticcircuit.Proc.Natl.Acad.Sci.99,16587–16591(2002).30. Yokobayashi, Y., Weiss, R. & Arnold, F.H. Directed evolution of agenetic circuit. Proc. Natl. Acad. Sci. 99, 16587–16591 (2002).

31.Ravikumar,A.,Arzumanyan,G.A.,Obadi,M.K.A.,Javanpour,A.A.&Liu,C.C.Scalable,Continuous Evolution of Genes at Mutation Ratesabove GenomicError Thresholds.Cell 175,1946-1957.e13(2018).31. Ravikumar, A., Arzumanyan, G.A., Obadi, M.K.A., Javanpour, A.A. & Liu, C.C. Scalable, Continuous Evolution of Genes at Mutation Ratesabove GenomicError Thresholds. Cell 175, 1946-1957.e13 (2018).

32.Sahin,U.,Karikó,K.&Türeci,mRNA-based therapeutics—developing anew class of drugs.Nat.Rev.Drug Discov.13,759–780(2014).32. Sahin, U., Karikó, K. & Türeci, mRNA-based therapeutics—developing a new class of drugs. Nat. Rev. Drug Discov. 13, 759–780 (2014).

33.Paunovska,K.,Loughrey,D.&Dahlman,J.E.Drug deliverysystems for RNAtherapeutics.Nat.Rev.Genet.23,265–280(2022).33. Paunovska, K., Loughrey, D. & Dahlman, J.E. Drug delivery systems for RNAtherapeutics. Nat. Rev. Genet. 23, 265–280 (2022).

34.Khan,K.H.Gene Expression in Mammalian Cells and itsApplications.Adv.Pharm.Bull.3,257–263(2013).34. Khan, K. H. Gene Expression in Mammalian Cells and its Applications. Adv. Pharm. Bull. 3, 257–263 (2013).

35.Strauss,J.H.&Strauss,E.G.The alphaviruses:gene expression,replication,and evolution.Microbiol.Rev.58,491–562(1994).35. Strauss, J.H. & Strauss, E.G. The alphaviruses: gene expression, replication, and evolution. Microbiol. Rev. 58, 491–562 (1994).

36.Frolov,I.et al.Alphavirus-based expression vectors:strategies andapplications.Proc.Natl.Acad.Sci.93,11371–11377(1996).36. Frolov, I. et al. Alphavirus-based expression vectors: strategies and applications. Proc. Natl. Acad. Sci. 93, 11371–11377 (1996).

37.Xiong,C.et al.Sindbis Virus:An Efficient,Broad Host 890RangeVector for Gene Expression in Animal Cells.Science 243,1188–1191(1989).37. Xiong, C. et al. Sindbis Virus: An Efficient, Broad Host 890 RangeVector for Gene Expression in Animal Cells. Science 243, 1188–1191 (1989).

38.Wolff,J.A.et al.Direct Gene Transfer into Mouse Muscle inVivo.Science 247,1465–1468(1990).38. Wolff, J.A. et al. Direct Gene Transfer into Mouse Muscle inVivo. Science 247, 1465–1468 (1990).

39.Zhou,X.et al.Self-replicating Semliki Forest virus RNAasrecombinant vaccine.Vaccine 12,1510–1514(1994).39. Zhou,

40.Wroblewska,L.et al.Mammalian synthetic circuits with RNAbindingproteins for RNA-only delivery.Nat.Biotechnol.33,839–841(2015).40. Wroblewska, L. et al. Mammalian synthetic circuits with RNAbindingproteins for RNA-only delivery. Nat. Biotechnol. 33, 839–841 (2015).

41.Jose,J.,Taylor,A.B.&Kuhn,R.J.Spatial and Temporal AnalysisofAlphavirus Replication and Assembly in Mammalian and MosquitoCells.mBio 8,e02294-16(2017).41. Jose, J., Taylor, A. B. & Kuhn, R. J. Spatial and Temporal Analysis of Alphavirus Replication and Assembly in Mammalian and Mosquito Cells. mBio 8, e02294-16 (2017).

42.Shirako,Y.&Strauss,J.H.Cleavage between nsP1 and nsP2initiates theprocessing pathway of Sindbis virus nonstructural polyproteinP123.Virology177,54–64(1990).42.Shirako,Y.&Strauss,J.H.Cleavage between nsP1 and nsP2initiates theprocessing pathway of Sindbis virus nonstructural polyproteinP123.Virology177,54–64(1990).

43.Frolov,I.et al.Selection of RNA Replicons Capable ofPersistentNoncytopathic Replication in Mammalian Cells.J.Virol.(1999)doi:10.1128/JVI.73.5.3854-3865.1999.43.Frolov,I.et al.Selection of RNA Replicons Capable ofPersistentNoncytopathic Replication in Mammalian Cells.J.Virol.(1999)doi:10.1128/JVI.73.5.3854-3865.1999.

44.Guo,J.-T.,Zhu,Q.&Seeger,C.Cytopathic and NoncytopathicInterferonResponses in Cells Expressing Hepatitis C VirusSubgenomicReplicons.J.Virol.77,10769–10779(2003).44. Guo, J.-T., Zhu, Q. & Seeger, C. Cytopathic and Noncytopathic InterferonResponses in Cells Expressing Hepatitis C Virus Subgenomic Replicons. J. Virol. 77, 10769–10779 (2003).

45.Rozen-Gagnon,K.et al.Alphavirus Mutator Variants PresentHost-Specific Defects and Attenuation in Mammalian and Insect Models.PLOSPathog.10,e1003877(2014).45. Rozen-Gagnon, K. et al. Alphavirus Mutator Variants PresentHost-Specific Defects and Attenuation in Mammalian and Insect Models. PLOS Pathog. 10, e1003877 (2014).

46.Urakova,N.et al.β-d-N4-Hydroxycytidine Is a PotentAnti-alphavirusCompound That Induces a High Level of Mutations in theViralGenome.J.Virol.92,e01965-17(2018).46. Urakova, N. et al. β-d-N4-Hydroxycytidine Is a PotentAnti-alphavirus Compound That Induces a High Level of Mutations in the ViralGenome. J. Virol. 92, e01965-17 (2018).

47.Delang,L.,Abdelnabi,R.&Neyts,J.Favipiravir as apotentialcountermeasure against neglected and emerging RNA viruses.AntiviralRes.153,85–94(2018).47. Delang, L., Abdelnabi, R. & Neyts, J. Favipiravir as a potential countermeasure against neglected and emerging RNA viruses. Antiviral Res. 153, 85–94 (2018).

48.Sheahan,T.P.et al.An orally bioavailable broad-spectrumantiviralinhibits SARS-CoV-2 in human airway epithelial cell cultures andmultiplecoronaviruses in mice.Sci.Transl.Med.12,eabb5883(2020).48. Sheahan, T.P. et al. An orally bioavailable broad-spectrum antiviralinhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci. Transl. Med. 12, eabb5883 (2020).

49.Tian,L.et al.RNA-dependent RNA polymerase(RdRp)inhibitors:Thecurrent landscape and repurposing for the COVID-19pandemic.Eur.J.Med.Chem.213,113201(2021).49. Tian, L. et al. RNA-dependent RNA polymerase (RdRp) inhibitors: The current landscape and repurposing for the COVID-19 pandemic. Eur. J. Med. Chem. 213, 113201 (2021).

50.Kabinger,F.et al.Mechanism of molnupiravir-inducedSARS-CoV-2 mutagenesis.Nat.Struct.Mol.Biol.28,740–746(2021).50. Kabinger, F. et al. Mechanism of molnupiravir-induced SARS-CoV-2 mutagenesis. Nat. Struct. Mol. Biol. 28, 740–746 (2021).

51.Heim,R.,Prasher,D.C.&Tsien,R.Y.Wavelength mutationsandposttranslational autoxidation of green fluorescentprotein.Proc.Natl.Acad.Sci.91,12501–12504(1994).51. Heim, R., Prasher, D.C. & Tsien, R.Y. Wavelength mutations and posttranslational autoxidation of green fluorescent protein. Proc. Natl. Acad. Sci. 91, 12501–12504 (1994).

52.Sawano,A.&Miyawaki,A.Directed evolution of greenfluorescentprotein by a new versatile PCR strategy for site-directed andsemi-randommutagenesis.Nucleic Acids Res.28,e78(2000).52. Sawano, A. & Miyawaki, A. Directed evolution of green fluorescent protein by a new versatile PCR strategy for site-directed and semi-randommutagenesis. Nucleic Acids Res. 28, e78 (2000).

53.Zapata-Hommer,O.&Griesbeck,O.Efficiently folding andcircularlypermuted variants of the Sapphire mutant of GFP.BMCBiotechnol.3,5(2003).53. Zapata-Hommer, O. & Griesbeck, O. Efficiently folding and circularlypermuted variants of the Sapphire mutant of GFP. BMCBiotechnol. 3, 5 (2003).

54.Stoltzfus,C.R.et al.Two-photon directed evolution ofgreenfluorescent proteins.Sci.Rep.5,11968(2015).54. Stoltzfus, C.R. et al. Two-photon directed evolution of green fluorescent proteins. Sci. Rep. 5, 11968 (2015).

55.Ramos,J.L.et al.The TetR Family of Transcriptional Repressors.Microbiol.Mol.Biol.Rev.69,326–356(2005).55. Ramos, J.L. et al. The TetR Family of Transcriptional Repressors. Microbiol. Mol. Biol. Rev. 69, 326–356 (2005).

56.Wang,Y.et al.A versatile genetic control system in mammaliancellsand mice responsive to clinically licensed sodium ferulate.Sci.Adv.6,eabb9484(2020).56. Wang, Y. et al. A versatile genetic control system in mammalian cells and mice responsive to clinically licensed sodium ferulate. Sci. Adv. 6, eabb9484 (2020).

57.Park,S.C.,Kwak,Y.M.,Song,W.S.,Hong,M.&Yoon,S.Structural basis ofeffector and operator recognition by the phenolicacid-responsivetranscriptional regulator PadR.Nucleic Acids Res.45,13080–13093(2017).57. Park, S.C., Kwak, Y.M., Song, W.S., Hong, M. & Yoon, S. Structural basis of effector and operator recognition by the phenolicacid-responsivetranscriptional regulator Pad R. Nucleic Acids Res. 45, 13080–13093 (2017).

58.Emery,C.M.et al.MEK1 mutations confer resistance to MEKand B-RAFinhibition.Proc.Natl.Acad.Sci.106,20411–20416(2009).58. Emery, C.M. et al. MEK1 mutations confer resistance to MEKand B-RAFinhibition. Proc. Natl. Acad. Sci. 106, 20411–20416 (2009).

59.Roskoski,R.Classification of small molecule proteinkinaseinhibitors based upon the structures of their drug-enzymecomplexes.Pharmacol.Res.103,26–48(2016).59. Roskoski, R. Classification of small molecule proteinkinaseinhibitors based upon the structures of their drug-enzymecomplexes. Pharmacol. Res. 103, 26–48 (2016).

60.Caunt,C.J.,Sale,M.J.,Smith,P.D.&Cook,S.J.MEK1 andMEK2 inhibitorsand cancer therapy:the long and winding road.Nat.Rev.Cancer 15,577–592(2015).60. Caunt, C. J., Sale, M. J., Smith, P. D. & Cook, S. J. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat. Rev. Cancer 15, 577–592 (2015).

61.Delaney,A.M.,Printen,J.A.,Chen,H.,Fauman,E.B.&Dudley,D.T.Identification of a Novel Mitogen-Activated Protein KinaseKinaseActivation Domain Recognized by the Inhibitor PD184352.Mol.Cell.Biol.22,7593–7602(2002).61. Delaney, A.M., Printen, J.A., Chen, H., Fauman, E.B. & Dudley, D.T. Identification of a Novel Mitogen-Activated Protein KinaseKinaseActivation Domain Recognized by the Inhibitor PD184352. Mol. Cell. Biol. 22, 7593–7602 (2002 ).

62.Treisman,R.The serum response element.Trends Biochem.Sci.17,423–426(1992).62. Treisman, R. The serum response element. Trends Biochem. Sci. 17, 423–426 (1992).

63.Waters,A.M.&Der,C.J.KRAS:The Critical Driver andTherapeutic Targetfor Pancreatic Cancer.Cold Spring Harb.Perspect.Med.8,a031435(2018).63. Waters, A.M. & Der, C.J. KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb. Perspect. Med. 8, a031435 (2018).

64.Adjei,A.A.Blocking Oncogenic Ras Signaling for Cancer Therapy.JNCIJ.Natl.Cancer Inst.93,1062–1074(2001).64. Adjei, A.A. Blocking Oncogenic Ras Signaling for Cancer Therapy. JNCIJ. Natl. Cancer Inst. 93, 1062–1074 (2001).

65.Weng,T.-Y.et al.DNA vaccine elicits an efficient antitumorresponseby targeting the mutant Kras in a transgenic mouse lung cancermodel.GeneTher.21,888–896(2014).65. Weng, T.-Y. et al. DNA vaccine elicits an efficient antitumor response by targeting the mutant Kras in a transgenic mouse lung cancer model. GeneTher. 21, 888–896 (2014).

66.Stewart,S.&Guan,K.-L.The Dominant Negative Ras Mutant,N17Ras,CanInhibit Signaling Independently of Blocking Ras Activation*.J.Biol.Chem.275,8854–8862(2000).66.Stewart,S.&Guan,K.-L.The Dominant Negative Ras Mutant,N17Ras,CanInhibit Signaling Independently of Blocking Ras Activation*.J.Biol.Chem.275,8854–8862(2000).

67.Zhang,B.,Zhang,Y.,Wang,Z.&Zheng,Y.The Role of Mg2+Cofactor in theGuanine Nucleotide Exchange and GTP HydrolysisReactions of Rho Family GTP-binding Proteins*.J.Biol.Chem.275,25299–25307(2000).67.Zhang,B.,Zhang,Y.,Wang,Z.&Zheng,Y.The Role of Mg2+Cofactor in theGuanine Nucleotide Exchange and GTP HydrolysisReactions of Rho Family GTP-binding Proteins*.J.Biol.Chem.275, 25299–25307(2000).

68.Feig,L.A.Dominant Inhibitory Ras Mutants:Tools forElucidating RasFunction.in GTPases in Biology I(eds.Dickey,B.F.&Birnbaumer,L.)289–300(Springer,1993).doi:10.1007/978-3-642-78267-1_19.68. Feature -1_19.

69.Kwan,A.K.,Piazza,G.A.,Keeton,A.B.&Leite,C.A.The pathto the clinic:a comprehensive review on direct KRASG12C inhibitors.J.Exp.Clin.CancerRes.41,27(2022).69. Kwan, A.K., Piazza, G.A., Keeton, A.B. & Leite, C.A. The path to the clinic: a comprehensive review on direct KRASG12C inhibitors. J. Exp. Clin. Cancer Res. 41, 27 (2022).

70.Spoerner,M.et al.Conformational States of Human Rat Sarcoma(Ras)Protein Complexed with Its Natural Ligand GTP and Their Role forEffectorInteraction and GTP Hydrolysis*.J.Biol.Chem.285,39768–39778(2010).70.Spoerner, M.et al.Conformational States of Human Rat Sarcoma(Ras)Protein Complexed with Its Natural Ligand GTP and Their Role for EffectorInteraction and GTP Hydrolysis*.J.Biol.Chem.285,39768–39778(2010).

71.Pantsar,T.et al.Assessment of mutation probabilities of KRASG12missense mutants and their long-timescale dynamics by atomisticmolecularsimulations and Markov state modeling.PLOS Comput.Biol.14,e1006458(2018).71.Pantsar,T.et al.Assessment of mutation probabilities of KRASG12missense mutants and their long-timescale dynamics by atomisticmolecularsimulations and Markov state modeling.PLOS Comput.Biol.14,e1006458(2018).

72.Cheng,A.A.&Lu,T.K.Synthetic Biology:An EmergingEngineering Discipline.Annu.Rev.Biomed.Eng.14,155–178(2012).72. Cheng, A. A. & Lu, T. K. Synthetic Biology: An EmergingEngineering Discipline. Annu. Rev. Biomed. Eng. 14, 155–178 (2012).

73.Esvelt,K.M.,Carlson,J.C.&Liu,D.R.A System for theContinuousDirected Evolution of Biomolecules.Nature 472,499–503(2011).73. Esvelt, K. M., Carlson, J. C. & Liu, D. R. A System for the Continuous Directed Evolution of Biomolecules. Nature 472, 499–503 (2011).

74.Miller,S.M.,Wang,T.&Liu,D.R.Phage-assisted continuous andnon-continuous evolution.Nat.Protoc.15,4101–4127(2020).74. Miller, S. M., Wang, T. & Liu, D. R. Phage-assisted continuous and non-continuous evolution. Nat. Protoc. 15, 4101–4127 (2020).

75.Lauring,A.S.&Andino,R.Quasispecies Theory and the Behaviorof RNAViruses.PLoS Pathog.6,e1001005(2010).75. Lauring, A.S. & Andino, R. Quasispecies Theory and the Behavior of RNAViruses. PLoS Pathog. 6, e1001005 (2010).

76.Harvey,W.T.et al.SARS-CoV-2 variants,spike mutations andimmuneescape.Nat.Rev.Microbiol.19,409–424(2021).76. Harvey, W.T. et al. SARS-CoV-2 variants, spike mutations and immunescape. Nat. Rev. Microbiol. 19, 409–424 (2021).

77.Robson,F.et al.Coronavirus RNA Proofreading:978 MolecularBasis andTherapeutic Targeting.Mol.Cell 79,710–727(2020).77. Robson, F. et al. Coronavirus RNA Proofreading:978 MolecularBasis andTherapeutic Targeting. Mol. Cell 79,710–727(2020).

78.Mills,D.R.,Peterson,R.L.&Spiegelman,S.An extracellularDarwinianexperiment with a self-duplicating nucleic acidmolecule.Proc.Natl.Acad.Sci.58,217–224(1967).78. Mills, D.R., Peterson, R.L. & Spiegelman, S. An extracellular Darwinian experiment with a self-duplicating nucleic acid molecule. Proc. Natl. Acad. Sci. 58, 217–224 (1967).

79.Ichihashi,N.et al.Darwinian evolution in a translation-coupledRNAreplication system within a cell-like compartment.Nat.Commun.4,2494(2013).79. Ichihashi, N. et al. Darwinian evolution in a translation-coupledRNAreplication system within a cell-like compartment. Nat. Commun. 4, 2494 (2013).

80.Dykstra,P.B.,Kaplan,M.&Smolke,C.D.Engineering synthetic RNAdevices for cell control.Nat.Rev.Genet.23,215–228(2022).80. Dykstra, P. B., Kaplan, M. & Smolke, C. D. Engineering synthetic RNA devices for cell control. Nat. Rev. Genet. 23, 215–228 (2022).

81.P.,Lusa,S.,Huylebroeck,D.&Garoff,H.In vitro mutagenesisof a full-length cDNA clone of Semliki Forest virus:the small6,000-molecular-weight membrane protein modulates virus release.J.Virol.65,4107–4113(1991).81. P., Lusa, S., Huylebroeck, D. & Garoff, H. In vitro mutagenesis of a full-length cDNA clone of Semliki Forest virus: the small6,000-molecular-weight membrane protein modulates virus release. J. Virol. 65, 4107–4113 (1991).

82.Huang,L.-K.&Wang,M.-J.J.Image thresholding by minimizing themeasures of fuzziness.Pattern Recognition 28,41–51(1995).82.Huang,L.-K.&Wang,M.-J.J.Image thresholding by minimizing the measures of fuzziness.Pattern Recognition 28,41–51(1995).

尽管本申请已经示例并描述了实施本发明的示例性实施方案,但本领域技术人员将理解的是,上述实施方案不应当被理解为对本发明内容进行任何限制,并且可在不背离本发明内容的精神、原则和范围的情况下进行变化、替换和修改,这些经修改的实施方案也在本发明的范围内。Although the present application has illustrated and described exemplary embodiments for implementing the present invention, those skilled in the art will appreciate that the above embodiments should not be construed as limiting the present invention in any way, and that changes, substitutions and modifications may be made without departing from the spirit, principle and scope of the present invention, and that these modified embodiments are also within the scope of the present invention.

Claims (32)

1.一种RNA复制酶辅助的连续体内进化(REPLACE)系统,其包含自主复制RNA和宿主细胞,1. An RNA replicase-assisted continuous in vivo evolution (REPLACE) system comprising autonomously replicating RNA and a host cell, 其中所述自主复制RNA为repRNA-v4,wherein the autonomously replicating RNA is repRNA-v4, 其中所述自主复制RNA缺失RNA复制酶,并且所述宿主细胞组成型表达所述缺失的RNA复制酶。Wherein the autonomously replicating RNA lacks RNA replicase, and the host cell constitutively expresses the lacked RNA replicase. 其中所述宿主细胞为真核生物宿主细胞。The host cell is a eukaryotic host cell. 2.根据权利要求1所述的REPLACE系统,所述宿主细胞为动物宿主细胞。2. The REPLACE system according to claim 1, wherein the host cell is an animal host cell. 3.根据权利要求1所述的REPLACE系统,所述宿主细胞为哺乳动物宿主细胞。The REPLACE system according to claim 1 , wherein the host cell is a mammalian host cell. 4.根据权利要求1所述的REPLACE系统,所述宿主细胞选自仓鼠成纤维细胞系或衍生自仓鼠成纤维细胞系。4. The REPLACE system according to claim 1, wherein the host cell is selected from a hamster fibroblast cell line or is derived from a hamster fibroblast cell line. 5.根据权利要求1所述的REPLACE系统,所述宿主细胞选自BHK-21或衍生自BHK-21。The REPLACE system according to claim 1 , wherein the host cell is selected from BHK-21 or is derived from BHK-21. 6.根据权利要求1所述的REPLACE系统,所述宿主细胞为复制酶有限的真核生物宿主细胞。The REPLACE system according to claim 1 , wherein the host cell is a replicase-limited eukaryotic host cell. 7.根据权利要求1所述的REPLACE系统,所述宿主细胞为复制酶有限的动物宿主细胞。The REPLACE system according to claim 1 , wherein the host cell is a replicase-limited animal host cell. 8.根据权利要求1所述的REPLACE系统,所述宿主细胞为复制酶有限的哺乳动物宿主细胞。The REPLACE system according to claim 1 , wherein the host cell is a replicase-limited mammalian host cell. 9.根据权利要求1所述的REPLACE系统,所述宿主细胞选自复制酶有限的仓鼠成纤维细胞系或衍生自复制酶有限的仓鼠成纤维细胞系。9. The REPLACE system of claim 1, wherein the host cell is selected from a replicase-limited hamster fibroblast cell line or a cell line derived from a replicase-limited hamster fibroblast cell line. 10.根据权利要求1所述的REPLACE系统,所述宿主细胞选自复制酶有限的BHK-21或衍生自复制酶有限的BHK-21。10. The REPLACE system according to claim 1, wherein the host cell is selected from the group consisting of replicase-limited BHK-21 or derived from replicase-limited BHK-21. 11.根据权利要求1所述的REPLACE系统,其还包含诱导突变的诱变剂。The REPLACE system according to claim 1 , further comprising a mutagen that induces mutation. 12.根据权利要求11所述的REPLACE系统,其中所述诱变剂选自小分子诱变剂、核苷类似物中的任一种。12 . The REPLACE system according to claim 11 , wherein the mutagen is selected from any one of a small molecule mutagen and a nucleoside analog. 13.根据权利要求11所述的REPLACE系统,其中所述诱变剂选自favipiravir、molnupiravir中的任一种。13. The REPLACE system according to claim 11, wherein the mutagen is selected from any one of favipiravir and molnupiravir. 14.一种RNA复制酶辅助的连续体内进化(REPLACE)方法,其包括:14. An RNA replicase-assisted continuous in vivo evolution (REPLACE) method comprising: 提供自主复制RNA;Provides autonomously replicating RNA; 提供宿主细胞;providing a host cell; 使自主复制RNA在宿主细胞内连续进化;Enables continuous evolution of autonomously replicating RNA within host cells; 其中所述自主复制RNA为repRNA-v4,wherein the autonomously replicating RNA is repRNA-v4, 其中所述自主复制RNA缺失RNA复制酶,并且所述宿主细胞组成型表达所述缺失的RNA复制酶,wherein the autonomously replicating RNA lacks RNA replicase, and the host cell constitutively expresses the lacked RNA replicase, 其中所述宿主细胞为真核生物宿主细胞。The host cell is a eukaryotic host cell. 15.根据权利要求14所述的REPLACE方法,所述宿主细胞为动物宿主细胞。The REPLACE method according to claim 14 , wherein the host cell is an animal host cell. 16.根据权利要求14所述的REPLACE方法,所述宿主细胞为哺乳动物宿主细胞。The REPLACE method according to claim 14 , wherein the host cell is a mammalian host cell. 17.根据权利要求14所述的REPLACE方法,所述宿主细胞选自仓鼠成纤维细胞系或衍生自仓鼠成纤维细胞系。17. The REPLACE method according to claim 14, wherein the host cell is selected from a hamster fibroblast cell line or is derived from a hamster fibroblast cell line. 18.根据权利要求14所述的REPLACE方法,所述宿主细胞选自BHK-21或衍生自BHK-21。18. The REPLACE method according to claim 14, wherein the host cell is selected from BHK-21 or is derived from BHK-21. 19.根据权利要求14所述的REPLACE方法,所述宿主细胞为复制酶有限的真核生物宿主细胞。The REPLACE method according to claim 14 , wherein the host cell is a replicase-limited eukaryotic host cell. 20.根据权利要求14所述的REPLACE方法,所述宿主细胞为复制酶有限的动物宿主细胞。20. The REPLACE method according to claim 14, wherein the host cell is a replicase-limited animal host cell. 21.根据权利要求14所述的REPLACE方法,所述宿主细胞为复制酶有限的哺乳动物宿主细胞。21. The REPLACE method according to claim 14, wherein the host cell is a replicase-limited mammalian host cell. 22.根据权利要求14所述的REPLACE方法,所述宿主细胞选自复制酶有限的仓鼠成纤维细胞系或衍生自复制酶有限的仓鼠成纤维细胞系。22. The REPLACE method of claim 14, wherein the host cell is selected from a replicase-limited hamster fibroblast cell line or a cell line derived from a replicase-limited hamster fibroblast cell line. 23.根据权利要求14所述的REPLACE方法,所述宿主细胞选自复制酶有限的BHK-21或衍生自复制酶有限的BHK-21。23. The REPLACE method according to claim 14, wherein the host cell is selected from the group consisting of replicase-limited BHK-21 or derived from replicase-limited BHK-21. 24.根据权利要求14所述的REPLACE方法,所述方法还包括提供诱导突变的诱变剂。24. The REPLACE method of claim 14, further comprising providing a mutagenic agent that induces mutation. 25.根据权利要求24所述的REPLACE方法,其中所述诱变剂选自小分子诱变剂、核苷类似物中的任一种。The REPLACE method according to claim 24 , wherein the mutagen is selected from any one of a small molecule mutagen and a nucleoside analog. 26.根据权利要求24所述的REPLACE方法,所述诱变剂选自favipiravir、molnupiravir中的任一种。26. The REPLACE method according to claim 24, wherein the mutagen is selected from any one of favipiravir and molnupiravir. 27.权利要求1至13中任一项所述的REPLACE系统在制造和优化生物大分子、疫苗、药物中的用途、在产生突变体库中的用途或者在实现达尔文式适应中的用途。27. Use of the REPLACE system of any one of claims 1 to 13 in the manufacture and optimization of biomacromolecules, vaccines, drugs, in the generation of mutant libraries, or in achieving Darwinian adaptation. 28.一种载体,其用于表达权利要求1至13中任一项所述的自主复制RNA。28. A vector for expressing the autonomously replicating RNA according to any one of claims 1 to 13. 29.一种宿主细胞,其包含表达载体,所述表达载体用于表达权利要求1至13中任一项所述的自主复制RNA。29. A host cell comprising an expression vector for expressing the autonomously replicating RNA according to any one of claims 1 to 13. 30.一种疫苗组合物,其包含通过或者可通过权利要求1至13中任一项所述的REPLACE系统产生的RNA。30. A vaccine composition comprising RNA produced or producible by the REPLACE system of any one of claims 1 to 13. 31.一种药物组合物,其包含通过或者可通过权利要求1至13中任一项所述的REPLACE系统产生的RNA。31. A pharmaceutical composition comprising RNA produced or producible by the REPLACE system of any one of claims 1 to 13. 32.一种递送载体,其包含通过或者可通过权利要求1至13中任一项所述的REPLACE系统产生的RNA。32. A delivery vector comprising RNA produced or produced by the REPLACE system of any one of claims 1 to 13.
CN202310078321.2A 2023-01-17 2023-01-17 Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA Active CN116064597B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202310078321.2A CN116064597B (en) 2023-01-17 2023-01-17 Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA
PCT/CN2024/072737 WO2024153118A1 (en) 2023-01-17 2024-01-17 Implementation of directed evolution and darwinian adaptation in mammalian cells by means of replicable rna

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310078321.2A CN116064597B (en) 2023-01-17 2023-01-17 Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA

Publications (2)

Publication Number Publication Date
CN116064597A CN116064597A (en) 2023-05-05
CN116064597B true CN116064597B (en) 2024-04-26

Family

ID=86176575

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310078321.2A Active CN116064597B (en) 2023-01-17 2023-01-17 Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA

Country Status (2)

Country Link
CN (1) CN116064597B (en)
WO (1) WO2024153118A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116064597B (en) * 2023-01-17 2024-04-26 北京大学 Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1081715A (en) * 1992-04-06 1994-02-09 佛罗里达大学研究基金会有限公司 Stem cell proliferation factor
CN1238806A (en) * 1996-07-17 1999-12-15 津莫吉尼蒂克斯公司 Preparation of pichia methanolica auxotrophic mutants
CN101023172A (en) * 2004-08-05 2007-08-22 协和发酵工业株式会社 Method of producing glycoprotein composition
WO2017188797A1 (en) * 2016-04-28 2017-11-02 연세대학교 산학협력단 Method for evaluating, in vivo, activity of rna-guided nuclease in high-throughput manner
CN111163799A (en) * 2017-07-28 2020-05-15 扬森疫苗与预防公司 Methods and compositions for heterologous repRNA immunization

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102005037351B3 (en) * 2005-08-08 2007-01-11 Geneart Ag In vitro method for directed evolution of proteins, useful e.g. in pharmaceutical development, uses expression system for performing translation, transcription and reverse transcription
CN107418964B (en) * 2016-05-24 2021-04-20 中国科学院深圳先进技术研究院 A phage-assisted multibacterial continuous directed evolution system and method
WO2019023680A1 (en) * 2017-07-28 2019-01-31 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (pace)
CN116064597B (en) * 2023-01-17 2024-04-26 北京大学 Directed evolution and darwinian adaptation in mammalian cells by autonomous replication of RNA

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1081715A (en) * 1992-04-06 1994-02-09 佛罗里达大学研究基金会有限公司 Stem cell proliferation factor
CN1238806A (en) * 1996-07-17 1999-12-15 津莫吉尼蒂克斯公司 Preparation of pichia methanolica auxotrophic mutants
CN101023172A (en) * 2004-08-05 2007-08-22 协和发酵工业株式会社 Method of producing glycoprotein composition
WO2017188797A1 (en) * 2016-04-28 2017-11-02 연세대학교 산학협력단 Method for evaluating, in vivo, activity of rna-guided nuclease in high-throughput manner
CN111163799A (en) * 2017-07-28 2020-05-15 扬森疫苗与预防公司 Methods and compositions for heterologous repRNA immunization

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
miR-483-5p通过上调IGF2基因转录促进肝癌细胞生长、迁移与侵袭;马彦;朱翠萍;胡建军;李悅行;王敏;汤绍辉;;中国病理生理杂志(第04期);全文 *

Also Published As

Publication number Publication date
CN116064597A (en) 2023-05-05
WO2024153118A1 (en) 2024-07-25

Similar Documents

Publication Publication Date Title
Odon et al. The role of ZAP and OAS3/RNAseL pathways in the attenuation of an RNA virus with elevated frequencies of CpG and UpA dinucleotides
Chiu et al. A bipotential organoid model of respiratory epithelium recapitulates high infectivity of SARS-CoV-2 Omicron variant
Jha et al. Trans-kingdom mimicry underlies ribosome customization by a poxvirus kinase
Lussignol et al. Proteomics of HCV virions reveals an essential role for the nucleoporin Nup98 in virus morphogenesis
Stroulios et al. Apical-out airway organoids as a platform for studying viral infections and screening for antiviral drugs
Dong et al. Establishment of a highly efficient virus-inducible CRISPR/Cas9 system in insect cells
Avgousti et al. Adenovirus core protein VII downregulates the DNA damage response on the host genome
Yi et al. Affinity purification of the hepatitis C virus replicase identifies valosin-containing protein, a member of the ATPases associated with diverse cellular activities family, as an active virus replication modulator
Lostroh Molecular and cellular biology of viruses
WO2024153118A1 (en) Implementation of directed evolution and darwinian adaptation in mammalian cells by means of replicable rna
CN110799643A (en) Compositions and methods for multiplex quantitative analysis of cell lineages
Ohlson et al. Genome-scale CRISPR screening reveals host factors required for ribosome formation and viral replication
Csibra et al. Modulation of stop codon read-through efficiency and its effect on the replication of murine leukemia virus
Frolova et al. Alphavirus-induced transcriptional and translational shutoffs play major roles in blocking the formation of stress granules
Hashizume et al. A small stem-loop-forming region within the 3′-UTR of a nonpolyadenylated LCMV mRNA promotes translation
Ruetz et al. In vitro and in vivo CRISPR-Cas9 screens reveal drivers of aging in neural stem cells of the brain
Ma et al. Orthogonal RNA replication enables directed evolution and Darwinian adaptation in mammalian cells
Zhu et al. Ebola virus sequesters IRF3 in viral inclusion bodies to evade host antiviral immunity
Sullender et al. Selective polyprotein processing determines norovirus sensitivity to trim7
JP2022525941A (en) Methods and systems for CRISPR selection
Zhang et al. Establishment and transcriptomic features of an immortalized hepatic cell line of the Chinese tree shrew
Yuan et al. Single-cell sequencing yields insights in the evolution of foot-and-mouth disease virus persistent infection
Hickson et al. RNA structures within Venezuelan equine encephalitis virus E1 alter macrophage replication fitness and contribute to viral emergence
Molina Molina et al. Targeting eEF1A reprograms translation and uncovers broad-spectrum antivirals against cap or m6A protein synthesis routes
WO2020196737A1 (en) Pharmaceutical composition for inhibiting production of hepatitis b virus protein, and method for screening same

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant