首页 > 最新文献

Molecular Therapy. Nucleic Acids最新文献

英文 中文
First-in-class transactivator-free, doxycycline-inducible IL-18-engineered CAR-T cells for relapsed/refractory B cell lymphomas 治疗复发/难治性 B 细胞淋巴瘤的不含转录因子、强力霉素诱导的 IL-18 工程 CAR-T 细胞,属同类产品之首
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-08-15 DOI: 10.1016/j.omtn.2024.102308
Pedro Justicia-Lirio, María Tristán-Manzano, Noelia Maldonado-Pérez, Carmen Barbero-Jiménez, Marina Cortijo-Gutiérrez, Kristina Pavlovic, Francisco J. Molina-Estevez, Pilar Muñoz, Ana Hinckley-Boned, Juan R. Rodriguez-Madoz, Felipe Prosper, Carmen Griñán-Lison, Saúl A. Navarro-Marchal, Julia Muñoz-Ballester, Pedro A. González-Sierra, Concha Herrera, Juan A. Marchal, Francisco Martín
Although chimeric antigen receptor (CAR) T cell therapy has revolutionized type B cancer treatment, efficacy remains limited in various lymphomas and solid tumors. Reinforcing conventional CAR-T cells to release cytokines can improve their efficacy but also increase safety concerns. Several strategies have been developed to regulate their secretion using minimal promoters that are controlled by chimeric proteins harboring transactivators. However, these chimeric proteins can disrupt the normal physiology of T cells. Here, we present the first transactivator-free anti-CD19 CAR-T cells able to control IL-18 expression (iTRUCK19.18) under ultra-low doses of doxycycline and without altering cellular fitness. Interestingly, IL-18 secretion requires T cell activation in addition to doxycycline, allowing the external regulation of CAR-T cell potency. This effect was translated into an increased CAR-T cell antitumor activity against aggressive hematologic and solid tumor models. In a clinically relevant context, we generated patient-derived iTRUCK19.18 cells capable of eradicating primary B cells tumors in a doxycycline-dependent manner. Furthermore, IL-18-releasing CAR-T cells polarized pro-tumoral macrophages toward an antitumoral phenotype, suggesting potential for modulating the tumor microenvironment. In summary, we showed that our platform can generate exogenously controlled CAR-T cells with enhanced potency and in the absence of transactivators.
虽然嵌合抗原受体(CAR)T 细胞疗法彻底改变了 B 型癌症的治疗,但在各种淋巴瘤和实体瘤中的疗效仍然有限。加强传统 CAR-T 细胞释放细胞因子可以提高疗效,但也会增加安全性问题。目前已开发出几种策略,利用由携带转录因子的嵌合蛋白控制的最小启动子来调节细胞因子的分泌。然而,这些嵌合蛋白会破坏 T 细胞的正常生理机能。在这里,我们首次发现了不含转座因子的抗 CD19 CAR-T 细胞,它们能在超低剂量强力霉素作用下控制 IL-18 的表达(iTRUCK19.18),且不会改变细胞的适应性。有趣的是,IL-18 的分泌除了需要强力霉素外,还需要 T 细胞的活化,从而实现了对 CAR-T 细胞效力的外部调节。这种效应转化为 CAR-T 细胞对侵袭性血液肿瘤和实体肿瘤模型的抗肿瘤活性。在与临床相关的背景下,我们生成了源自患者的 iTRUCK19.18 细胞,它能够以多西环素依赖的方式根除原发性 B 细胞肿瘤。此外,释放IL-18的CAR-T细胞还能将亲肿瘤巨噬细胞极化为抗肿瘤表型,这表明CAR-T细胞具有调节肿瘤微环境的潜力。总之,我们的研究表明,我们的平台可以在没有反式激活剂的情况下生成外源控制的 CAR-T 细胞,并增强其效力。
{"title":"First-in-class transactivator-free, doxycycline-inducible IL-18-engineered CAR-T cells for relapsed/refractory B cell lymphomas","authors":"Pedro Justicia-Lirio, María Tristán-Manzano, Noelia Maldonado-Pérez, Carmen Barbero-Jiménez, Marina Cortijo-Gutiérrez, Kristina Pavlovic, Francisco J. Molina-Estevez, Pilar Muñoz, Ana Hinckley-Boned, Juan R. Rodriguez-Madoz, Felipe Prosper, Carmen Griñán-Lison, Saúl A. Navarro-Marchal, Julia Muñoz-Ballester, Pedro A. González-Sierra, Concha Herrera, Juan A. Marchal, Francisco Martín","doi":"10.1016/j.omtn.2024.102308","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102308","url":null,"abstract":"Although chimeric antigen receptor (CAR) T cell therapy has revolutionized type B cancer treatment, efficacy remains limited in various lymphomas and solid tumors. Reinforcing conventional CAR-T cells to release cytokines can improve their efficacy but also increase safety concerns. Several strategies have been developed to regulate their secretion using minimal promoters that are controlled by chimeric proteins harboring transactivators. However, these chimeric proteins can disrupt the normal physiology of T cells. Here, we present the first transactivator-free anti-CD19 CAR-T cells able to control IL-18 expression (iTRUCK19.18) under ultra-low doses of doxycycline and without altering cellular fitness. Interestingly, IL-18 secretion requires T cell activation in addition to doxycycline, allowing the external regulation of CAR-T cell potency. This effect was translated into an increased CAR-T cell antitumor activity against aggressive hematologic and solid tumor models. In a clinically relevant context, we generated patient-derived iTRUCK19.18 cells capable of eradicating primary B cells tumors in a doxycycline-dependent manner. Furthermore, IL-18-releasing CAR-T cells polarized pro-tumoral macrophages toward an antitumoral phenotype, suggesting potential for modulating the tumor microenvironment. In summary, we showed that our platform can generate exogenously controlled CAR-T cells with enhanced potency and in the absence of transactivators.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"40 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142222598","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Plug-and-play nucleic acid-mediated multimerization of biparatopic nanobodies for molecular imaging 即插即用的核酸介导的双原子纳米抗体多聚化,用于分子成像
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-08-15 DOI: 10.1016/j.omtn.2024.102305
Laura Teodori, Sarah K. Ochoa, Marjan Omer, Veronica L. Andersen, Pernille Bech, Junyi Su, Jessica Bridoux, Jesper S. Nielsen, Mathias B. Bertelsen, Sophie Hernot, Kurt V. Gothelf, Jørgen Kjems
In cancer molecular imaging, selecting binders with high specificity and affinity for biomarkers is paramount for achieving high-contrast imaging within clinical time frames. Nanobodies have emerged as potent candidates, surpassing antibodies in pre-clinical imaging due to their convenient production, rapid renal clearance, and deeper tissue penetration. Multimerization of nanobodies is a popular strategy to enhance their affinity and pharmacokinetics; however, traditional methods are laborious and may yield heterogeneous products. In this study, we employ a Holliday junction (HJ)-like nucleic acid-based scaffold to create homogeneous nanostructures with precise multivalent and multiparatopic nanobody displays. The plug-and-play assembly allowed the screening of several nanobody multimer configurations for the detection of the breast cancer biomarker, human epidermal growth factor receptor 2 (HER2). studies demonstrated significant improvements in binding avidity, particularly with the biparatopic construct exhibiting high sensitivity, surpassing that of traditional antibody-based cell binding. Furthermore, our HJ platform allowed for adaptation from fluorescence-based to nuclear imaging, as demonstrated in xenografted mice, thereby allowing for future applications. This work highlights the potential of nucleic acid-mediated multimerization to markedly enhance nanobody binding, by exploring synergistic combinations and offering versatility for both diagnostics and cancer molecular imaging with prospects for future theranostic applications.
在癌症分子成像中,选择对生物标记物具有高特异性和亲和力的粘合剂对于在临床时限内实现高对比度成像至关重要。纳米抗体因其生产方便、肾脏清除快、组织穿透更深等优点,已成为临床前成像中超越抗体的有效候选药物。纳米抗体的多聚化是增强其亲和力和药代动力学的常用策略;然而,传统方法费时费力,而且可能产生异构产物。在这项研究中,我们采用了一种类似霍利迪接合点(HJ)的核酸基支架来创建具有精确多价和多异位纳米抗体显示的同质纳米结构。这种即插即用的组装方式可以筛选出几种纳米抗体多聚体配置,用于检测乳腺癌生物标志物人类表皮生长因子受体 2(HER2)。研究表明,结合热度有了显著提高,特别是双配位构建体表现出了高灵敏度,超过了传统的基于抗体的细胞结合。此外,正如在异种移植小鼠中展示的那样,我们的 HJ 平台允许从基于荧光到核成像的调整,从而为未来的应用提供了可能。这项工作强调了核酸介导的多聚化在显著增强纳米抗体结合方面的潜力,通过探索协同组合,为诊断和癌症分子成像提供了多功能性,为未来的治疗学应用提供了前景。
{"title":"Plug-and-play nucleic acid-mediated multimerization of biparatopic nanobodies for molecular imaging","authors":"Laura Teodori, Sarah K. Ochoa, Marjan Omer, Veronica L. Andersen, Pernille Bech, Junyi Su, Jessica Bridoux, Jesper S. Nielsen, Mathias B. Bertelsen, Sophie Hernot, Kurt V. Gothelf, Jørgen Kjems","doi":"10.1016/j.omtn.2024.102305","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102305","url":null,"abstract":"In cancer molecular imaging, selecting binders with high specificity and affinity for biomarkers is paramount for achieving high-contrast imaging within clinical time frames. Nanobodies have emerged as potent candidates, surpassing antibodies in pre-clinical imaging due to their convenient production, rapid renal clearance, and deeper tissue penetration. Multimerization of nanobodies is a popular strategy to enhance their affinity and pharmacokinetics; however, traditional methods are laborious and may yield heterogeneous products. In this study, we employ a Holliday junction (HJ)-like nucleic acid-based scaffold to create homogeneous nanostructures with precise multivalent and multiparatopic nanobody displays. The plug-and-play assembly allowed the screening of several nanobody multimer configurations for the detection of the breast cancer biomarker, human epidermal growth factor receptor 2 (HER2). studies demonstrated significant improvements in binding avidity, particularly with the biparatopic construct exhibiting high sensitivity, surpassing that of traditional antibody-based cell binding. Furthermore, our HJ platform allowed for adaptation from fluorescence-based to nuclear imaging, as demonstrated in xenografted mice, thereby allowing for future applications. This work highlights the potential of nucleic acid-mediated multimerization to markedly enhance nanobody binding, by exploring synergistic combinations and offering versatility for both diagnostics and cancer molecular imaging with prospects for future theranostic applications.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"30 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142222604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nanobody against SARS-CoV-2 non-structural protein Nsp9 inhibits viral replication in human airway epithelia 抗 SARS-CoV-2 非结构蛋白 Nsp9 的纳米抗体可抑制病毒在人气道上皮细胞中的复制
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-08-15 DOI: 10.1016/j.omtn.2024.102304
Tomas Venit, Jeremy Blavier, Sibusiso B. Maseko, Sam Shu, Lilia Espada, Christopher Breunig, Hans-Peter Holthoff, Sabrina C. Desbordes, Martin Lohse, Gennaro Esposito, Jean-Claude Twizere, Piergiorgio Percipalle
Nanobodies are emerging as critical tools for drug design. Several have been recently created to serve as inhibitors of severe acute respiratory syndrome coronavirus s (SARS-CoV-2) entry in the host cell by targeting surface-exposed spike protein. Here we have established a pipeline that instead targets highly conserved viral proteins made only after viral entry into the host cell when the SARS-CoV-2 RNA-based genome is translated. As proof of principle, we designed nanobodies against the SARS-CoV-2 non-structural protein (Nsp)9, which is required for viral genome replication. One of these anti-Nsp9 nanobodies, 2NSP23, previously characterized using immunoassays and nuclear magnetic resonance spectroscopy for epitope mapping, was expressed and found to block SARS-CoV-2 replication specifically. We next encapsulated 2NSP23 nanobody into lipid nanoparticles (LNPs) as mRNA. We show that this nanobody, hereby referred to as LNP-mRNA-2NSP23, is internalized and translated in cells and suppresses multiple SARS-CoV-2 variants, as seen by qPCR and RNA deep sequencing. These results are corroborated in three-dimensional reconstituted human epithelium kept at air-liquid interface to mimic the outer surface of lung tissue. These observations indicate that LNP-mRNA-2NSP23 is internalized and, after translation, it inhibits viral replication by targeting Nsp9 in living cells. We speculate that LNP-mRNA-2NSP23 may be translated into an innovative strategy to generate novel antiviral drugs highly efficient across coronaviruses.
纳米抗体正在成为药物设计的重要工具。最近有几种纳米抗体通过靶向表面暴露的尖峰蛋白,成为严重急性呼吸系统综合症冠状病毒(SARS-CoV-2)进入宿主细胞的抑制剂。在这里,我们建立了一个管道,它的目标是高度保守的病毒蛋白,这些蛋白只有在病毒进入宿主细胞后,当基于 SARS-CoV-2 RNA 的基因组被翻译时才会产生。作为原理验证,我们设计了抗SARS-CoV-2非结构蛋白(Nsp)9的纳米抗体,病毒基因组复制需要这种蛋白。其中一种抗 Nsp9 纳米抗体 2NSP23 以前曾利用免疫测定和核磁共振光谱绘制表位图对其进行过表征,我们表达了这种纳米抗体并发现它能特异性地阻断 SARS-CoV-2 的复制。接下来,我们将 2NSP23 纳米抗体作为 mRNA 封装到脂质纳米粒子(LNPs)中。通过 qPCR 和 RNA 深度测序,我们发现这种纳米抗体(在此称为 LNP-mRNA-2NSP23)能在细胞内被内化和翻译,并能抑制多种 SARS-CoV-2 变体。这些结果在模拟肺组织外表面的气液界面三维重建人体上皮细胞中得到了证实。这些观察结果表明,LNP-mRNA-2NSP23 被内化,翻译后通过靶向活细胞中的 Nsp9 抑制病毒复制。我们推测,LNP-mRNA-2NSP23 有可能被转化为一种创新策略,产生新型抗病毒药物,对冠状病毒非常有效。
{"title":"Nanobody against SARS-CoV-2 non-structural protein Nsp9 inhibits viral replication in human airway epithelia","authors":"Tomas Venit, Jeremy Blavier, Sibusiso B. Maseko, Sam Shu, Lilia Espada, Christopher Breunig, Hans-Peter Holthoff, Sabrina C. Desbordes, Martin Lohse, Gennaro Esposito, Jean-Claude Twizere, Piergiorgio Percipalle","doi":"10.1016/j.omtn.2024.102304","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102304","url":null,"abstract":"Nanobodies are emerging as critical tools for drug design. Several have been recently created to serve as inhibitors of severe acute respiratory syndrome coronavirus s (SARS-CoV-2) entry in the host cell by targeting surface-exposed spike protein. Here we have established a pipeline that instead targets highly conserved viral proteins made only after viral entry into the host cell when the SARS-CoV-2 RNA-based genome is translated. As proof of principle, we designed nanobodies against the SARS-CoV-2 non-structural protein (Nsp)9, which is required for viral genome replication. One of these anti-Nsp9 nanobodies, 2NSP23, previously characterized using immunoassays and nuclear magnetic resonance spectroscopy for epitope mapping, was expressed and found to block SARS-CoV-2 replication specifically. We next encapsulated 2NSP23 nanobody into lipid nanoparticles (LNPs) as mRNA. We show that this nanobody, hereby referred to as LNP-mRNA-2NSP23, is internalized and translated in cells and suppresses multiple SARS-CoV-2 variants, as seen by qPCR and RNA deep sequencing. These results are corroborated in three-dimensional reconstituted human epithelium kept at air-liquid interface to mimic the outer surface of lung tissue. These observations indicate that LNP-mRNA-2NSP23 is internalized and, after translation, it inhibits viral replication by targeting Nsp9 in living cells. We speculate that LNP-mRNA-2NSP23 may be translated into an innovative strategy to generate novel antiviral drugs highly efficient across coronaviruses.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"60 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142222634","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The changing scenario of drug discovery using AI to deep learning: Recent advancement, success stories, collaborations, and challenges 利用人工智能和深度学习进行药物发现的变化情况:最新进展、成功案例、合作与挑战
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-08-08 DOI: 10.1016/j.omtn.2024.102295
Chiranjib Chakraborty, Manojit Bhattacharya, Sang-Soo Lee, Zhi-Hong Wen, Yi-Hao Lo
Due to the transformation of artificial intelligence (AI) tools and technologies, AI-driven drug discovery has come to the forefront. It reduces the time and expenditure. Due to these advantages, pharmaceutical industries are concentrating on AI-driven drug discovery. Several drug molecules have been discovered using AI-based techniques and tools, and several newly AI-discovered drug molecules have already entered clinical trials. In this review, we first present the data and their resources in the pharmaceutical sector for AI-driven drug discovery and illustrated some significant algorithms or techniques used for AI and ML which are used in this field. We gave an overview of the deep neural network (NN) models and compared them with artificial NNs. Then, we illustrate the recent advancement of the landscape of drug discovery using AI to deep learning, such as the identification of drug targets, prediction of their structure, estimation of drug-target interaction, estimation of drug-target binding affinity, design of drug, prediction of drug toxicity, estimation of absorption, distribution, metabolism, excretion, toxicity; and estimation of drug-drug interaction. Moreover, we highlighted the success stories of AI-driven drug discovery and discussed several collaboration and the challenges in this area. The discussions in the article will enrich the pharmaceutical industry.
由于人工智能(AI)工具和技术的变革,AI 驱动的药物发现已走到前沿。它缩短了时间,减少了开支。由于这些优势,制药行业正专注于人工智能驱动的药物发现。一些药物分子已经利用基于人工智能的技术和工具被发现,一些新发现的人工智能药物分子已经进入临床试验阶段。在这篇综述中,我们首先介绍了制药领域用于人工智能驱动药物发现的数据及其资源,并说明了该领域使用的一些重要的人工智能和 ML 算法或技术。我们概述了深度神经网络(NN)模型,并将其与人工神经网络进行了比较。然后,我们阐述了利用人工智能和深度学习进行药物发现的最新进展,如药物靶点的识别、药物靶点结构的预测、药物与靶点相互作用的估计、药物与靶点结合亲和力的估计、药物设计、药物毒性的预测、吸收、分布、代谢、排泄、毒性的估计以及药物与药物相互作用的估计。此外,我们还重点介绍了人工智能驱动药物发现的成功案例,并讨论了这一领域的若干合作与挑战。文章中的讨论将丰富制药行业的内容。
{"title":"The changing scenario of drug discovery using AI to deep learning: Recent advancement, success stories, collaborations, and challenges","authors":"Chiranjib Chakraborty, Manojit Bhattacharya, Sang-Soo Lee, Zhi-Hong Wen, Yi-Hao Lo","doi":"10.1016/j.omtn.2024.102295","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102295","url":null,"abstract":"Due to the transformation of artificial intelligence (AI) tools and technologies, AI-driven drug discovery has come to the forefront. It reduces the time and expenditure. Due to these advantages, pharmaceutical industries are concentrating on AI-driven drug discovery. Several drug molecules have been discovered using AI-based techniques and tools, and several newly AI-discovered drug molecules have already entered clinical trials. In this review, we first present the data and their resources in the pharmaceutical sector for AI-driven drug discovery and illustrated some significant algorithms or techniques used for AI and ML which are used in this field. We gave an overview of the deep neural network (NN) models and compared them with artificial NNs. Then, we illustrate the recent advancement of the landscape of drug discovery using AI to deep learning, such as the identification of drug targets, prediction of their structure, estimation of drug-target interaction, estimation of drug-target binding affinity, design of drug, prediction of drug toxicity, estimation of absorption, distribution, metabolism, excretion, toxicity; and estimation of drug-drug interaction. Moreover, we highlighted the success stories of AI-driven drug discovery and discussed several collaboration and the challenges in this area. The discussions in the article will enrich the pharmaceutical industry.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"60 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142222616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhancing peptide and PMO delivery to mouse airway epithelia by chemical conjugation with the amphiphilic peptide S10 通过与两亲性肽 S10 化学共轭,增强向小鼠气道上皮细胞输送肽和 PMO 的能力
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-07-31 DOI: 10.1016/j.omtn.2024.102290
Maud Auger, Luis Sorroza-Martinez, Nadine Brahiti, Carole-Ann Huppé, Laurence Faucher-Giguère, Imen Arbi, Maxime Hervault, Xue Cheng, Bruno Gaillet, Frédéric Couture, David Guay, Al-Halifa Soultan
Delivery of antisense oligonucleotides (ASOs) to airway epithelial cells is arduous due to the physiological barriers that protect the lungs and the endosomal entrapment phenomenon, which prevents ASOs from reaching their intracellular targets. Various delivery strategies involving peptide-, lipid-, and polymer-based carriers are being investigated, yet the challenge remains. S10 is a peptide-based delivery agent that enables the intracellular delivery of biomolecules such as GFP, CRISPR-associated nuclease ribonucleoprotein (RNP), base editor RNP, and a fluorescent peptide into lung cells after intranasal or intratracheal administrations to mice, ferrets, and rhesus monkeys. Herein, we demonstrate that covalently attaching S10 to a fluorescently labeled peptide or a functional splice-switching phosphorodiamidate morpholino oligomer improves their intracellular delivery to airway epithelia in mice after a single intranasal instillation. Data reveal a homogeneous delivery from the trachea to the distal region of the lungs, specifically into the cells lining the airway. Quantitative measurements further highlight that conjugation via a disulfide bond through a pegylated (PEG) linker was the most beneficial strategy compared with direct conjugation (without the PEG linker) or conjugation via a permanent thiol-maleimide bond. We believe that S10-based conjugation provides a great strategy to achieve intracellular delivery of peptides and ASOs with therapeutic properties in lungs.
将反义寡核苷酸(ASO)输送到气道上皮细胞是一项艰巨的任务,这是因为保护肺部的生理屏障和内质体阻滞现象阻碍了反义寡核苷酸到达细胞内靶点。目前正在研究涉及肽基、脂基和聚合物基载体的各种递送策略,但挑战依然存在。S10 是一种基于肽的递送剂,它能在小鼠、雪貂和恒河猴的鼻腔内或气管内给药后,将 GFP、CRISPR 相关核酸酶核糖核蛋白(RNP)、碱基编辑 RNP 和荧光肽等生物大分子送入肺细胞内。在本文中,我们证明了将 S10 共价连接到荧光标记的多肽或功能性剪接转换磷酰二胺吗啉寡聚体上,可提高它们在小鼠一次鼻内灌注后向气道上皮细胞的胞内递送。数据显示,从气管到肺的远端区域,特别是气道内壁细胞,都能均匀地输送。定量测量结果进一步表明,与直接共轭(不含 PEG 连接体)或通过永久性硫醇-马来酰亚胺键共轭相比,通过聚乙二醇(PEG)连接体的二硫键共轭是最有效的策略。我们相信,基于 S10 的共轭为在肺部实现具有治疗特性的多肽和 ASO 的细胞内递送提供了一种很好的策略。
{"title":"Enhancing peptide and PMO delivery to mouse airway epithelia by chemical conjugation with the amphiphilic peptide S10","authors":"Maud Auger, Luis Sorroza-Martinez, Nadine Brahiti, Carole-Ann Huppé, Laurence Faucher-Giguère, Imen Arbi, Maxime Hervault, Xue Cheng, Bruno Gaillet, Frédéric Couture, David Guay, Al-Halifa Soultan","doi":"10.1016/j.omtn.2024.102290","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102290","url":null,"abstract":"Delivery of antisense oligonucleotides (ASOs) to airway epithelial cells is arduous due to the physiological barriers that protect the lungs and the endosomal entrapment phenomenon, which prevents ASOs from reaching their intracellular targets. Various delivery strategies involving peptide-, lipid-, and polymer-based carriers are being investigated, yet the challenge remains. S10 is a peptide-based delivery agent that enables the intracellular delivery of biomolecules such as GFP, CRISPR-associated nuclease ribonucleoprotein (RNP), base editor RNP, and a fluorescent peptide into lung cells after intranasal or intratracheal administrations to mice, ferrets, and rhesus monkeys. Herein, we demonstrate that covalently attaching S10 to a fluorescently labeled peptide or a functional splice-switching phosphorodiamidate morpholino oligomer improves their intracellular delivery to airway epithelia in mice after a single intranasal instillation. Data reveal a homogeneous delivery from the trachea to the distal region of the lungs, specifically into the cells lining the airway. Quantitative measurements further highlight that conjugation via a disulfide bond through a pegylated (PEG) linker was the most beneficial strategy compared with direct conjugation (without the PEG linker) or conjugation via a permanent thiol-maleimide bond. We believe that S10-based conjugation provides a great strategy to achieve intracellular delivery of peptides and ASOs with therapeutic properties in lungs.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"46 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141932108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of selective and non-selective C9ORF72 targeting in vivo active siRNAs 鉴定选择性和非选择性 C9ORF72 靶向体内活性 siRNA
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-07-31 DOI: 10.1016/j.omtn.2024.102291
James W. Gilbert, Zachary Kennedy, Bruno Godinho, Ashley Summers, Alexandra Weiss, Dimas Echeverria, Brianna Bramato, Nicholas McHugh, David Cooper, Ken Yamada, Matthew Hassler, Hélène Tran, Fen Biao Gao, Robert H. Brown Jr., Anastasia Khvorova
A hexanucleotide (GC) repeat expansion (HRE) within intron one of is the leading genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). haploinsufficiency, formation of RNA foci, and production of dipeptide repeat (DPR) proteins have been proposed as mechanisms of disease. Here, we report the first example of disease-modifying siRNAs for driven ALS/FTD. Using a combination of reporter assay and primary cortical neurons derived from a C9-ALS/FTD mouse model, we screened a panel of more than 150 fully chemically stabilized siRNAs targeting different transcriptional variants. We demonstrate the lack of correlation between siRNA efficacy in reporter assay versus native environment; repeat-containing mRNA variants are found to preferentially localize to the nucleus, and thus mRNA accessibility and intracellular localization have a dominant impact on functional RNAi. Using a C9-ALS/FTD mouse model, we demonstrate that divalent siRNAs targeting mRNA variants specifically or non-selectively reduce the expression of mRNA and significantly reduce DPR proteins. Interestingly, siRNA silencing all mRNA transcripts was more effective in removing intranuclear mRNA aggregates than targeting only HRE-containing mRNA transcripts. Combined, these data support RNAi-based degradation of as a potential therapeutic paradigm.
肌萎缩性脊髓侧索硬化症(ALS)和额颞叶痴呆症(FTD)的主要遗传病因是肌萎缩性脊髓侧索硬化症(ALS)和额颞叶痴呆症(FTD)内含子一的六核苷酸(GC)重复扩增(HRE)。在此,我们首次报道了用于驱动 ALS/FTD 的可改变疾病的 siRNA。我们结合使用报告分析法和从 C9-ALS/FTD 小鼠模型中提取的原发性皮层神经元,筛选出了 150 多种针对不同转录变体的完全化学稳定 siRNA。我们证明了 siRNA 在报告检测中的功效与原生环境之间缺乏相关性;含重复的 mRNA 变体被发现优先定位于细胞核,因此 mRNA 的可及性和细胞内定位对功能性 RNAi 有主要影响。我们利用 C9-ALS/FTD 小鼠模型证明,针对 mRNA 变异的二价 siRNA 可特异性或非选择性地降低 mRNA 的表达,并显著减少 DPR 蛋白。有趣的是,与只针对含 HRE 的 mRNA 转录本相比,沉默所有 mRNA 转录本的 siRNA 能更有效地清除核内 mRNA 聚集。综合来看,这些数据支持将基于 RNAi 的降解作为一种潜在的治疗范式。
{"title":"Identification of selective and non-selective C9ORF72 targeting in vivo active siRNAs","authors":"James W. Gilbert, Zachary Kennedy, Bruno Godinho, Ashley Summers, Alexandra Weiss, Dimas Echeverria, Brianna Bramato, Nicholas McHugh, David Cooper, Ken Yamada, Matthew Hassler, Hélène Tran, Fen Biao Gao, Robert H. Brown Jr., Anastasia Khvorova","doi":"10.1016/j.omtn.2024.102291","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102291","url":null,"abstract":"A hexanucleotide (GC) repeat expansion (HRE) within intron one of is the leading genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). haploinsufficiency, formation of RNA foci, and production of dipeptide repeat (DPR) proteins have been proposed as mechanisms of disease. Here, we report the first example of disease-modifying siRNAs for driven ALS/FTD. Using a combination of reporter assay and primary cortical neurons derived from a C9-ALS/FTD mouse model, we screened a panel of more than 150 fully chemically stabilized siRNAs targeting different transcriptional variants. We demonstrate the lack of correlation between siRNA efficacy in reporter assay versus native environment; repeat-containing mRNA variants are found to preferentially localize to the nucleus, and thus mRNA accessibility and intracellular localization have a dominant impact on functional RNAi. Using a C9-ALS/FTD mouse model, we demonstrate that divalent siRNAs targeting mRNA variants specifically or non-selectively reduce the expression of mRNA and significantly reduce DPR proteins. Interestingly, siRNA silencing all mRNA transcripts was more effective in removing intranuclear mRNA aggregates than targeting only HRE-containing mRNA transcripts. Combined, these data support RNAi-based degradation of as a potential therapeutic paradigm.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"2 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141932107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel gene therapy for drug-resistant melanoma: Synergistic combination of PTEN plasmid and BRD4 PROTAC-loaded lipid nanocarriers 耐药性黑色素瘤的新型基因疗法:PTEN 质粒与 BRD4 PROTAC 脂质纳米载体的协同组合
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-07-31 DOI: 10.1016/j.omtn.2024.102292
Aishwarya Saraswat, Hari Priya Vemana, Vikas Dukhande, Ketan Patel
Patients suffering from BRAF mutant melanoma have tumor recurrence within merely 7 months of treatment with a potent BRAF inhibitor (BRAFi) like vemurafenib. It has been proven that diverse molecular pathways driving BRAFi resistance converge to activation of c-Myc in melanoma. Therefore, we identified a novel combinatorial therapeutic strategy by targeting loss of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor gene and upregulated BRD4 oncoprotein as Myc-dependent vulnerabilities of drug-resistant melanoma. Being promising therapeutic targets, we decided to concomitantly deliver PTEN plasmid and BRD4 targeted PROteolysis-TArgeting Chimera (ARV) to drug the “undruggable” c-Myc in BRAFi-resistant melanoma. Since PTEN plasmid and ARV are distinct in their physicochemical properties, we fabricated PTEN-plasmid loaded lipid nanoparticles (PL-NANO) and ARV-825-loaded nanoliposomes (AL-NANO) to yield a mean particle size of less than 100 nm and greater than 99% encapsulation efficiency for each therapeutic payload. Combination of PL-NANO and AL-NANO displayed synergistic tumor growth inhibition and substantial apoptosis in two-dimensional and three-dimensional models. Importantly, simultaneous delivery of PL-NANO and AL-NANO achieved significant upregulation of PTEN expression levels and degradation of BRD4 protein to ultimately downregulate c-Myc levels in BRAFi-resistant melanoma cells. Altogether, lipid nanocarriers delivering this novel lethal cocktail stands as one-of-a-kind gene therapy to target undruggable c-Myc oncogene in BRAFi-resistant melanoma.
BRAF突变黑色素瘤患者在接受强效BRAF抑制剂(BRAFi)(如维莫非尼)治疗仅7个月后,肿瘤就会复发。事实证明,在黑色素瘤中,驱动BRAFi耐药性的多种分子通路都汇聚到了c-Myc的激活上。因此,我们发现了一种新的组合治疗策略,即针对10号染色体上缺失的磷酸酶和缺失的天丝同源物(PTEN)肿瘤抑制基因和上调的BRD4肿瘤蛋白,将其作为耐药黑色素瘤的Myc依赖性弱点。作为有希望的治疗靶点,我们决定同时提供 PTEN 质粒和 BRD4 靶向 PROteolysis-TArgeting Chimera(ARV),以治疗 BRAF 抗性黑色素瘤中 "无法治疗 "的 c-Myc。由于 PTEN 质粒和 ARV 的理化性质不同,我们制作了负载 PTEN 质粒的脂质纳米粒子(PL-NANO)和负载 ARV-825 的纳米脂质体(AL-NANO),使每种治疗载荷的平均粒径小于 100 纳米,封装效率大于 99%。在二维和三维模型中,PL-NANO 和 AL-NANO 的组合显示出协同的肿瘤生长抑制作用和显著的细胞凋亡作用。重要的是,PL-NANO 和 AL-NANO 的同时给药可显著上调 PTEN 的表达水平,并降解 BRD4 蛋白,最终下调 BRAFi 抗性黑色素瘤细胞中 c-Myc 的水平。总之,脂质纳米载体可提供这种新型致命鸡尾酒,是针对BRAF耐药黑色素瘤中无法治疗的c-Myc癌基因的独一无二的基因疗法。
{"title":"Novel gene therapy for drug-resistant melanoma: Synergistic combination of PTEN plasmid and BRD4 PROTAC-loaded lipid nanocarriers","authors":"Aishwarya Saraswat, Hari Priya Vemana, Vikas Dukhande, Ketan Patel","doi":"10.1016/j.omtn.2024.102292","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102292","url":null,"abstract":"Patients suffering from BRAF mutant melanoma have tumor recurrence within merely 7 months of treatment with a potent BRAF inhibitor (BRAFi) like vemurafenib. It has been proven that diverse molecular pathways driving BRAFi resistance converge to activation of c-Myc in melanoma. Therefore, we identified a novel combinatorial therapeutic strategy by targeting loss of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor gene and upregulated BRD4 oncoprotein as Myc-dependent vulnerabilities of drug-resistant melanoma. Being promising therapeutic targets, we decided to concomitantly deliver PTEN plasmid and BRD4 targeted PROteolysis-TArgeting Chimera (ARV) to drug the “undruggable” c-Myc in BRAFi-resistant melanoma. Since PTEN plasmid and ARV are distinct in their physicochemical properties, we fabricated PTEN-plasmid loaded lipid nanoparticles (PL-NANO) and ARV-825-loaded nanoliposomes (AL-NANO) to yield a mean particle size of less than 100 nm and greater than 99% encapsulation efficiency for each therapeutic payload. Combination of PL-NANO and AL-NANO displayed synergistic tumor growth inhibition and substantial apoptosis in two-dimensional and three-dimensional models. Importantly, simultaneous delivery of PL-NANO and AL-NANO achieved significant upregulation of PTEN expression levels and degradation of BRD4 protein to ultimately downregulate c-Myc levels in BRAFi-resistant melanoma cells. Altogether, lipid nanocarriers delivering this novel lethal cocktail stands as one-of-a-kind gene therapy to target undruggable c-Myc oncogene in BRAFi-resistant melanoma.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"39 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141932071","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DNA/RNA heteroduplex technology with cationic oligopeptide reduces class-related adverse effects of nucleic acid drugs 带有阳离子寡肽的 DNA/RNA 异源复合物技术可减少核酸药物的类相关不良反应
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-07-31 DOI: 10.1016/j.omtn.2024.102289
Masahiro Ohara, Tetsuya Nagata, Rintaro Iwata Hara, Kie Yoshida-Tanaka, Nozomi Toide, Kazunori Takagi, Kazuki Sato, Tomoya Takenaka, Masanori Nakakariya, Kenichi Miyata, Maeda Yusuke, Kazuko Toh, Takeshi Wada, Takanori Yokota
Antisense oligonucleotides (ASOs) are a therapeutic modality for incurable diseases. However, systemic injection of gapmer-type ASOs causes class-related toxicities, including prolongation of activated partial thromboplastin time (aPTT) and thrombocytopenia. We previously reported that cholesterol-conjugated DNA/RNA heteroduplex oligonucleotides (Chol-HDOs) exhibit significantly enhanced gene-silencing effects compared to ASOs, even in the central nervous system, by crossing the blood-brain barrier. In the present study, we initially evaluated the effect of the HDO structure on class-related toxicities. The HDO structure ameliorated the class-related toxicities associated with ASOs, but they remained to some extent. As a further antidote, we have developed artificial cationic oligopeptides, L-2,4-diaminobutanoic acid oligomers (DabOs), which bind to the phosphates in the major groove of the A-type double-helical structure of HDOs. The DabO/Chol-HDO complex showed significantly improved aPTT prolongation and thrombocytopenia in mice while maintaining gene-silencing efficacy. Moreover, the conjugation with DabOs effectively prevented cerebral infarction, a condition frequently observed in mice intravenously injected with high-dose Chol-HDO. These approaches, combining HDO technology with DabOs, offer distinct advantages over conventional strategies in reducing toxicities. Consequently, the DabO/HDO complex represents a promising platform for overcoming the class-related toxicities associated with therapeutic ASOs.
反义寡核苷酸(ASO)是一种治疗不治之症的方法。然而,全身注射间隙型反义寡核苷酸会引起与类相关的毒性,包括活化部分凝血活酶时间(aPTT)延长和血小板减少。我们以前曾报道过,胆固醇共轭 DNA/RNA 异质双链寡核苷酸(Chol-HDOs)通过穿越血脑屏障,即使在中枢神经系统中也能表现出比 ASOs 明显更强的基因沉默效应。在本研究中,我们初步评估了 HDO 结构对同类相关毒性的影响。HDO 结构改善了与 ASOs 相关的类相关毒性,但在一定程度上仍然存在。作为进一步的解毒剂,我们开发了人工阳离子寡肽--L-2,4-二氨基丁酸寡聚体(DabOs),它能与 HDOs 的 A 型双螺旋结构主要沟槽中的磷酸盐结合。DabO/Chol-HDO 复合物能显著改善小鼠的 aPTT 延长和血小板减少,同时保持基因沉默的功效。此外,与 DabOs 的共轭还能有效防止脑梗塞,而脑梗塞是静脉注射大剂量 Chol-HDO 的小鼠经常出现的症状。与传统策略相比,这些将 HDO 技术与 DabOs 相结合的方法在降低毒性方面具有明显优势。因此,DabO/HDO 复合物是克服与治疗性 ASO 有关的类相关毒性的一个前景广阔的平台。
{"title":"DNA/RNA heteroduplex technology with cationic oligopeptide reduces class-related adverse effects of nucleic acid drugs","authors":"Masahiro Ohara, Tetsuya Nagata, Rintaro Iwata Hara, Kie Yoshida-Tanaka, Nozomi Toide, Kazunori Takagi, Kazuki Sato, Tomoya Takenaka, Masanori Nakakariya, Kenichi Miyata, Maeda Yusuke, Kazuko Toh, Takeshi Wada, Takanori Yokota","doi":"10.1016/j.omtn.2024.102289","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102289","url":null,"abstract":"Antisense oligonucleotides (ASOs) are a therapeutic modality for incurable diseases. However, systemic injection of gapmer-type ASOs causes class-related toxicities, including prolongation of activated partial thromboplastin time (aPTT) and thrombocytopenia. We previously reported that cholesterol-conjugated DNA/RNA heteroduplex oligonucleotides (Chol-HDOs) exhibit significantly enhanced gene-silencing effects compared to ASOs, even in the central nervous system, by crossing the blood-brain barrier. In the present study, we initially evaluated the effect of the HDO structure on class-related toxicities. The HDO structure ameliorated the class-related toxicities associated with ASOs, but they remained to some extent. As a further antidote, we have developed artificial cationic oligopeptides, L-2,4-diaminobutanoic acid oligomers (DabOs), which bind to the phosphates in the major groove of the A-type double-helical structure of HDOs. The DabO/Chol-HDO complex showed significantly improved aPTT prolongation and thrombocytopenia in mice while maintaining gene-silencing efficacy. Moreover, the conjugation with DabOs effectively prevented cerebral infarction, a condition frequently observed in mice intravenously injected with high-dose Chol-HDO. These approaches, combining HDO technology with DabOs, offer distinct advantages over conventional strategies in reducing toxicities. Consequently, the DabO/HDO complex represents a promising platform for overcoming the class-related toxicities associated with therapeutic ASOs.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"98 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141932111","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Suppression of HSV-1 infection and viral reactivation by CRISPR-Cas9 gene editing in 2D and 3D culture models 在二维和三维培养模型中通过 CRISPR-Cas9 基因编辑抑制 HSV-1 感染和病毒再活化
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-07-19 DOI: 10.1016/j.omtn.2024.102282
Anna Bellizzi, Senem Çakır, Martina Donadoni, Rahsan Sariyer, Shuren Liao, Hong Liu, Guo-Xiang Ruan, Jennifer Gordon, Kamel Khalili, Ilker K. Sariyer
Although our understanding of herpes simplex virus type 1 (HSV-1) biology has been considerably enhanced, developing therapeutic strategies to eliminate HSV-1 in latently infected individuals remains a public health concern. Current antiviral drugs used for the treatment of HSV-1 complications are not specific and do not address latent infection. We recently developed a CRISPR-Cas9-based gene editing platform to specifically target the HSV-1 genome. In this study, we further used 2D Vero cell culture and 3D human induced pluripotent stem cell-derived cerebral organoid (CO) models to assess the effectiveness of our editing constructs targeting viral ICP0 or ICP27 genes. We found that targeting the ICP0 or ICP27 genes with AAV2-CRISPR-Cas9 vectors in Vero cells drastically suppressed HSV-1 replication. In addition, we productively infected COs with HSV-1, characterized the viral replication kinetics, and established a viral latency model. Finally, we discovered that ICP0- or ICP27-targeting AAV2-CRISPR-Cas9 vector significantly reduced viral rebound in the COs that were latently infected with HSV-1. In summary, our results suggest that CRISPR-Cas9 gene editing of HSV-1 is an efficient therapeutic approach to eliminate the latent viral reservoir and treat HSV-1-associated complications.
尽管我们对 1 型单纯疱疹病毒(HSV-1)生物学的了解已经大大加深,但制定治疗策略以消除潜伏感染者体内的 HSV-1 仍然是一个公共卫生问题。目前用于治疗 HSV-1 并发症的抗病毒药物不具有特异性,也不能解决潜伏感染问题。我们最近开发了一种基于 CRISPR-Cas9 的基因编辑平台,专门针对 HSV-1 基因组。在这项研究中,我们进一步使用二维Vero细胞培养和三维人类诱导多能干细胞衍生脑器官(CO)模型来评估我们针对病毒ICP0或ICP27基因的编辑构建物的有效性。我们发现,在Vero细胞中使用AAV2-CRISPR-Cas9载体靶向ICP0或ICP27基因能显著抑制HSV-1的复制。此外,我们还用HSV-1感染了CO,鉴定了病毒复制动力学,并建立了病毒潜伏模型。最后,我们发现以 ICP0 或 ICP27 为靶向的 AAV2-CRISPR-Cas9 载体能显著减少潜伏感染 HSV-1 的 COs 中的病毒反弹。总之,我们的研究结果表明,对HSV-1进行CRISPR-Cas9基因编辑是消除潜伏病毒库和治疗HSV-1相关并发症的有效治疗方法。
{"title":"Suppression of HSV-1 infection and viral reactivation by CRISPR-Cas9 gene editing in 2D and 3D culture models","authors":"Anna Bellizzi, Senem Çakır, Martina Donadoni, Rahsan Sariyer, Shuren Liao, Hong Liu, Guo-Xiang Ruan, Jennifer Gordon, Kamel Khalili, Ilker K. Sariyer","doi":"10.1016/j.omtn.2024.102282","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102282","url":null,"abstract":"Although our understanding of herpes simplex virus type 1 (HSV-1) biology has been considerably enhanced, developing therapeutic strategies to eliminate HSV-1 in latently infected individuals remains a public health concern. Current antiviral drugs used for the treatment of HSV-1 complications are not specific and do not address latent infection. We recently developed a CRISPR-Cas9-based gene editing platform to specifically target the HSV-1 genome. In this study, we further used 2D Vero cell culture and 3D human induced pluripotent stem cell-derived cerebral organoid (CO) models to assess the effectiveness of our editing constructs targeting viral ICP0 or ICP27 genes. We found that targeting the ICP0 or ICP27 genes with AAV2-CRISPR-Cas9 vectors in Vero cells drastically suppressed HSV-1 replication. In addition, we productively infected COs with HSV-1, characterized the viral replication kinetics, and established a viral latency model. Finally, we discovered that ICP0- or ICP27-targeting AAV2-CRISPR-Cas9 vector significantly reduced viral rebound in the COs that were latently infected with HSV-1. In summary, our results suggest that CRISPR-Cas9 gene editing of HSV-1 is an efficient therapeutic approach to eliminate the latent viral reservoir and treat HSV-1-associated complications.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"69 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141782960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immunoactive signatures of circulating tRNA- and rRNA-derived RNAs in chronic obstructive pulmonary disease 慢性阻塞性肺病中循环 tRNA 和 rRNA 衍生 RNA 的免疫活性特征
IF 8.8 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-07-19 DOI: 10.1016/j.omtn.2024.102285
Megumi Shigematsu, Takuya Kawamura, Deepak A. Deshpande, Yohei Kirino
Chronic obstructive pulmonary disease (COPD) is the most prevalent lung disease, and macrophages play a central role in the inflammatory response in COPD. We here report a comprehensive characterization of circulating short non-coding RNAs (sncRNAs) in plasma from patients with COPD. While circulating sncRNAs are increasingly recognized for their regulatory roles and biomarker potential in various diseases, the conventional RNA sequencing (RNA-seq) method cannot fully capture these circulating sncRNAs due to their heterogeneous terminal structures. By pre-treating the plasma RNAs with T4 polynucleotide kinase, which converts all RNAs to those with RNA-seq susceptible ends (5′-phosphate and 3′-hydroxyl), we comprehensively sequenced a wide variety of non-microRNA sncRNAs, such as 5′-tRNA halves containing a 2′,3′-cyclic phosphate. We discovered a remarkable accumulation of the 5′-half derived from tRNA in plasma from COPD patients, whereas the 5′-tRNA half is predominant in healthy donors. Further, the 5′-tRNA half activates human macrophages via Toll-like receptor 7 and induces cytokine production. Additionally, we identified circulating rRNA-derived fragments that were upregulated in COPD patients and demonstrated their ability to induce cytokine production in macrophages. Our findings provide evidence of circulating, immune-active sncRNAs in patients with COPD, suggesting that they serve as inflammatory mediators in the pathogenesis of COPD.
慢性阻塞性肺病(COPD)是发病率最高的肺部疾病,而巨噬细胞在慢性阻塞性肺病的炎症反应中起着核心作用。我们在此报告了慢性阻塞性肺病患者血浆中循环短非编码 RNA(sncRNA)的综合特征。尽管循环中的 sncRNAs 因其在各种疾病中的调控作用和生物标志物潜力而被越来越多的人所认识,但由于其末端结构的异质性,传统的 RNA 测序(RNA-seq)方法无法完全捕获这些循环中的 sncRNAs。T4 多核苷酸激酶能将所有 RNA 转化为 RNA-seq 易感末端(5′-磷酸和 3′-羟基),通过对血浆 RNA 进行预处理,我们全面测序了多种非微小 RNA sncRNA,如含有 2′,3′-环磷酸的 5′-tRNA。我们发现,在慢性阻塞性肺病患者的血浆中,由 tRNA 衍生的 5′-半部分明显累积,而在健康供体中,5′-tRNA 半部分占主导地位。此外,5′-tRNA 的一半通过 Toll 样受体 7 激活人类巨噬细胞,并诱导细胞因子的产生。此外,我们还发现了慢性阻塞性肺病患者体内上调的循环 rRNA 衍生片段,并证明了它们诱导巨噬细胞产生细胞因子的能力。我们的研究结果提供了慢性阻塞性肺病患者体内具有免疫活性的循环 sncRNA 的证据,表明它们在慢性阻塞性肺病的发病机制中充当了炎症介质。
{"title":"Immunoactive signatures of circulating tRNA- and rRNA-derived RNAs in chronic obstructive pulmonary disease","authors":"Megumi Shigematsu, Takuya Kawamura, Deepak A. Deshpande, Yohei Kirino","doi":"10.1016/j.omtn.2024.102285","DOIUrl":"https://doi.org/10.1016/j.omtn.2024.102285","url":null,"abstract":"Chronic obstructive pulmonary disease (COPD) is the most prevalent lung disease, and macrophages play a central role in the inflammatory response in COPD. We here report a comprehensive characterization of circulating short non-coding RNAs (sncRNAs) in plasma from patients with COPD. While circulating sncRNAs are increasingly recognized for their regulatory roles and biomarker potential in various diseases, the conventional RNA sequencing (RNA-seq) method cannot fully capture these circulating sncRNAs due to their heterogeneous terminal structures. By pre-treating the plasma RNAs with T4 polynucleotide kinase, which converts all RNAs to those with RNA-seq susceptible ends (5′-phosphate and 3′-hydroxyl), we comprehensively sequenced a wide variety of non-microRNA sncRNAs, such as 5′-tRNA halves containing a 2′,3′-cyclic phosphate. We discovered a remarkable accumulation of the 5′-half derived from tRNA in plasma from COPD patients, whereas the 5′-tRNA half is predominant in healthy donors. Further, the 5′-tRNA half activates human macrophages via Toll-like receptor 7 and induces cytokine production. Additionally, we identified circulating rRNA-derived fragments that were upregulated in COPD patients and demonstrated their ability to induce cytokine production in macrophages. Our findings provide evidence of circulating, immune-active sncRNAs in patients with COPD, suggesting that they serve as inflammatory mediators in the pathogenesis of COPD.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":"20 1","pages":""},"PeriodicalIF":8.8,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141872466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Therapy. Nucleic Acids
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1