首页 > 最新文献

Frontiers in genome editing最新文献

英文 中文
Knockout mutation in TaD27 enhances number of productive tillers in hexaploid wheat. TaD27的基因敲除突变可提高六倍体小麦的高产分蘖数量。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-10-14 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1455761
Muhammad Jawad Akbar Awan, Imran Amin, Awais Rasheed, Nasir A Saeed, Shahid Mansoor

Recent advances allow the deployment of cluster regularly interspaced short palindromic repeats (CRISPR)-associated endonucleases (Cas) system for the targeted mutagenesis in the genome with accuracy and precision for trait improvement in crops. CRISPR-Cas systems have been extensively utilized to induce knockout or frameshift mutations in the targeted sequence of mostly negative regulating genes for wheat improvement. However, most of the reported work has been done in non-commercial varieties of wheat and introgression of edited alleles into breeding population comes with the penalty of unwanted linkage-drag. Wheat yield is controlled by various genes such as positive and negative regulators. The TaD27 gene is described as a negative regulator of shoot branching or tillering and involved in the biosynthesis of strigolactones. In this study, we developed Tad27 knockout mutant lines of an elite wheat cultivar that showed a twofold increase in the number of tillers and 1.8-fold increase in the number of grains per plant. Subsequently, enhancing the grain yield without any morphological penalty in the architecture of the plants. The co-transformation of regeneration enhancing growth regulator, Growth Regulating Factor 4 (GRF4) and its cofactor GRF-Interacting Factor 1 (GIF1), under single T-DNA cassette improved the regeneration efficiency up to 6% of transgenic events from mature embryos of wheat. Our results indicate that the CRISPR-mediated targeted mutagenesis confers the potential to knockout yield-related negative regulators in elite cultivars of wheat that can substantially enhance grain yield per plant and this strategy can be harnessed for the improvement of future wheat.

最近的研究进展使人们能够利用集群有规则间隔短回文重复序列(CRISPR)-相关内切酶(Cas)系统对基因组进行精确的定向诱变,从而改良作物的性状。CRISPR-Cas 系统已被广泛用于诱导大部分负调控基因目标序列的基因敲除或框移突变,以改良小麦。然而,大多数报道的工作都是在非商业性小麦品种中进行的,将编辑过的等位基因导入育种群体会带来不必要的连接拖累。小麦产量受各种基因控制,如正向和负向调节因子。TaD27 基因被描述为芽分枝或分蘖的负调控因子,并参与绞股蓝内酯的生物合成。在这项研究中,我们培育了一个小麦精英栽培品种的 Tad27 基因敲除突变株系,其分蘖数量增加了两倍,单株籽粒数增加了 1.8 倍。随后,在不影响植株形态结构的情况下提高了谷物产量。在单个 T-DNA 基因盒下共转化可促进再生的生长调节因子--生长调节因子 4(GRF4)及其辅助因子--GRF-互作因子 1(GIF1),可使小麦成熟胚的转基因效率提高到 6%。我们的研究结果表明,CRISPR 介导的定向诱变具有敲除小麦优良栽培品种中与产量相关的负调控因子的潜力,可大幅提高单株谷物产量,这一策略可用于未来小麦的改良。
{"title":"Knockout mutation in <i>TaD27</i> enhances number of productive tillers in hexaploid wheat.","authors":"Muhammad Jawad Akbar Awan, Imran Amin, Awais Rasheed, Nasir A Saeed, Shahid Mansoor","doi":"10.3389/fgeed.2024.1455761","DOIUrl":"10.3389/fgeed.2024.1455761","url":null,"abstract":"<p><p>Recent advances allow the deployment of cluster regularly interspaced short palindromic repeats (CRISPR)-associated endonucleases (Cas) system for the targeted mutagenesis in the genome with accuracy and precision for trait improvement in crops. CRISPR-Cas systems have been extensively utilized to induce knockout or frameshift mutations in the targeted sequence of mostly negative regulating genes for wheat improvement. However, most of the reported work has been done in non-commercial varieties of wheat and introgression of edited alleles into breeding population comes with the penalty of unwanted linkage-drag. Wheat yield is controlled by various genes such as positive and negative regulators. The <i>TaD27</i> gene is described as a negative regulator of shoot branching or tillering and involved in the biosynthesis of strigolactones. In this study, we developed <i>Tad27</i> knockout mutant lines of an elite wheat cultivar that showed a twofold increase in the number of tillers and 1.8-fold increase in the number of grains per plant. Subsequently, enhancing the grain yield without any morphological penalty in the architecture of the plants. The co-transformation of regeneration enhancing growth regulator, G<i>rowth Regulating Factor 4</i> (<i>GRF4</i>) and its cofactor <i>GRF-Interacting Factor 1</i> (<i>GIF1</i>), under single T-DNA cassette improved the regeneration efficiency up to 6% of transgenic events from mature embryos of wheat. Our results indicate that the CRISPR-mediated targeted mutagenesis confers the potential to knockout yield-related negative regulators in elite cultivars of wheat that can substantially enhance grain yield per plant and this strategy can be harnessed for the improvement of future wheat.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11513295/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142523774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting DLBCL by mutation-specific disruption of cancer-driving oncogenes. 通过突变特异性地破坏致癌基因来靶向 DLBCL。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-10-14 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1427322
Najmeh Heshmatpour, S Maryam Kazemi, Niklas D Schmidt, Sarita R Patnaik, Patrick Korus, Bodo G C Wilkens, Arturo Macarrón Palacios

Diffuse large B cell lymphomas (DLBCL) are highly aggressive tumors. Their genetic complexity and heterogeneity have hampered the development of novel approaches for precision medicine. Our study aimed to develop a personalized therapy for DLBCL by utilizing the CRISPR/Cas system to induce knockouts (KO) of driver genes, thereby causing cancer cell death while minimizing side effects. We focused on OCI-LY3 cells, modeling DLBCL, and compared them with BJAB cells as controls. Analysis of whole exome sequencing revealed significant mutations in genes like PAX5, CD79B, and MYC in OCI-LY3 cells. CRISPR/Cas9-mediated KO of these genes resulted in reduced cancer cell viability. Subsequent single and dual gRNA targeting of PAX5 mutations inhibited proliferation specifically in OCI-LY3 cells. Moreover, dual gRNA targeting of PAX5 and MYC induced chromosomal rearrangements, reducing cell proliferation substantially. However, targeting single intronic mutations did not affect cell viability, highlighting the importance of disrupting protein function. Targeting multiple mutations simultaneously addresses intra-tumoral heterogeneity, and the transient delivery of CRISPR/Cas9 allows for permanent gene disruption. While challenges such as incomplete editing efficiency and delivery limitations exist, further optimization may enhance therapeutic efficacy. Overall, our findings demonstrate the efficacy of CRISPR/Cas9 in targeting oncogenic mutations, opening avenues for precision medicine in DLBCL treatment.

弥漫性大B细胞淋巴瘤(DLBCL)是一种侵袭性极强的肿瘤。它们的遗传复杂性和异质性阻碍了精准医疗新方法的开发。我们的研究旨在利用CRISPR/Cas系统诱导驱动基因敲除(KO),从而导致癌细胞死亡,同时将副作用降至最低,从而开发出针对DLBCL的个性化疗法。我们重点研究了模拟DLBCL的OCI-LY3细胞,并将其与作为对照的BJAB细胞进行了比较。全外显子组测序分析显示,OCI-LY3 细胞中的 PAX5、CD79B 和 MYC 等基因发生了显著突变。CRISPR/Cas9 介导的这些基因的 KO 可降低癌细胞的活力。随后,PAX5 突变的单gRNA靶向和双gRNA靶向可特异性抑制 OCI-LY3 细胞的增殖。此外,PAX5 和 MYC 的双 gRNA 靶向会诱导染色体重排,从而大幅降低细胞增殖。然而,靶向单个内含子突变并不会影响细胞活力,这突出了破坏蛋白质功能的重要性。同时靶向多个突变可解决瘤内异质性问题,CRISPR/Cas9的瞬时传递可实现永久性基因破坏。虽然存在编辑效率不完全和递送限制等挑战,但进一步优化可能会提高疗效。总之,我们的研究结果证明了 CRISPR/Cas9 在靶向致癌突变方面的功效,为 DLBCL 的精准医疗开辟了道路。
{"title":"Targeting DLBCL by mutation-specific disruption of cancer-driving oncogenes.","authors":"Najmeh Heshmatpour, S Maryam Kazemi, Niklas D Schmidt, Sarita R Patnaik, Patrick Korus, Bodo G C Wilkens, Arturo Macarrón Palacios","doi":"10.3389/fgeed.2024.1427322","DOIUrl":"10.3389/fgeed.2024.1427322","url":null,"abstract":"<p><p>Diffuse large B cell lymphomas (DLBCL) are highly aggressive tumors. Their genetic complexity and heterogeneity have hampered the development of novel approaches for precision medicine. Our study aimed to develop a personalized therapy for DLBCL by utilizing the CRISPR/Cas system to induce knockouts (KO) of driver genes, thereby causing cancer cell death while minimizing side effects. We focused on OCI-LY3 cells, modeling DLBCL, and compared them with BJAB cells as controls. Analysis of whole exome sequencing revealed significant mutations in genes like <i>PAX5</i>, <i>CD79B</i>, and <i>MYC</i> in OCI-LY3 cells. CRISPR/Cas9-mediated KO of these genes resulted in reduced cancer cell viability. Subsequent single and dual gRNA targeting of <i>PAX5</i> mutations inhibited proliferation specifically in OCI-LY3 cells. Moreover, dual gRNA targeting of <i>PAX5</i> and <i>MYC</i> induced chromosomal rearrangements, reducing cell proliferation substantially. However, targeting single intronic mutations did not affect cell viability, highlighting the importance of disrupting protein function. Targeting multiple mutations simultaneously addresses intra-tumoral heterogeneity, and the transient delivery of CRISPR/Cas9 allows for permanent gene disruption. While challenges such as incomplete editing efficiency and delivery limitations exist, further optimization may enhance therapeutic efficacy. Overall, our findings demonstrate the efficacy of CRISPR/Cas9 in targeting oncogenic mutations, opening avenues for precision medicine in DLBCL treatment.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11513324/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142523775","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The potential of HBV cure: an overview of CRISPR-mediated HBV gene disruption. 治愈 HBV 的潜力:CRISPR 介导的 HBV 基因干扰概述。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-10-09 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1467449
Zhi Q Yao, Madison B Schank, Juan Zhao, Mohamed El Gazzar, Ling Wang, Yi Zhang, Addison C Hill, Puja Banik, Jaeden S Pyburn, Jonathan P Moorman

Hepatitis B virus (HBV) infection is a common cause of liver disease worldwide. The current antiviral treatment using nucleotide analogues (NAs) can only suppress de novo HBV replication but cannot eliminate chronic HBV infection due to the persistence of covalently closed circular (ccc) DNA that sustains viral replication. The CRISPR/Cas9 system is a novel genome-editing tool that enables precise gene disruption and inactivation. With high efficiency and simplicity, the CRISPR/Cas9 system has been utilized in multiple studies to disrupt the HBV genome specifically, eliciting varying anti-HBV effects both in vitro and in vivo. Additionally, multi-locus gene targeting has shown enhanced antiviral activity, paving the way for combination therapy to disrupt and inactivate HBV cccDNA as well as integrated HBV DNA. Despite its promising antiviral effects, this technology faces several challenges that need to be overcome before its clinical application, i.e., off-target effects and in vivo drug delivery. As such, there is a need for improvement in CRISPR/Cas9 efficiency, specificity, versatility, and delivery. Here, we critically review the recent literature describing the tools employed in designing guide RNAs (gRNAs) targeting HBV genomes, the vehicles used for expressing and delivering CRISPR/Cas9 components, the models used for evaluating CRISPR-mediated HBV gene disruption, the methods used for assessing antiviral and off-target effects induced by CRISPR/Cas9-mediated HBV gene disruption, and the prospects of future directions and challenges in leveraging this HBV gene-editing approach, to advance the HBV treatment toward a clinical cure.

乙型肝炎病毒(HBV)感染是全球肝病的常见病因。目前使用核苷酸类似物(NAs)进行的抗病毒治疗只能抑制新的 HBV 复制,但无法消除慢性 HBV 感染,因为持续存在的共价闭合环状(ccc)DNA 可维持病毒复制。CRISPR/Cas9系统是一种新型基因组编辑工具,可以精确地破坏和灭活基因。CRISPR/Cas9 系统高效简便,已被用于多项研究,特异性地破坏 HBV 基因组,在体外和体内产生不同的抗 HBV 作用。此外,多焦点基因打靶也显示出更强的抗病毒活性,为破坏和灭活 HBV cccDNA 以及整合的 HBV DNA 的联合疗法铺平了道路。尽管该技术具有良好的抗病毒效果,但在临床应用前仍面临着一些需要克服的挑战,即脱靶效应和体内给药。因此,需要提高 CRISPR/Cas9 的效率、特异性、多功能性和给药方式。在此,我们对最近的文献进行了批判性的回顾,这些文献描述了设计靶向 HBV 基因组的引导 RNA(gRNA)所使用的工具、表达和递送 CRISPR/Cas9 成分所使用的载体、评估 CRISPR 介导的 HBV 基因破坏所使用的模型、评估 CRISPR/Cas9 介导的 HBV 基因破坏所诱导的抗病毒和脱靶效应所使用的方法,以及利用这种 HBV 基因编辑方法的未来方向和挑战的前景,从而推动 HBV 治疗走向临床治愈。
{"title":"The potential of HBV cure: an overview of CRISPR-mediated HBV gene disruption.","authors":"Zhi Q Yao, Madison B Schank, Juan Zhao, Mohamed El Gazzar, Ling Wang, Yi Zhang, Addison C Hill, Puja Banik, Jaeden S Pyburn, Jonathan P Moorman","doi":"10.3389/fgeed.2024.1467449","DOIUrl":"https://doi.org/10.3389/fgeed.2024.1467449","url":null,"abstract":"<p><p>Hepatitis B virus (HBV) infection is a common cause of liver disease worldwide. The current antiviral treatment using nucleotide analogues (NAs) can only suppress <i>de novo</i> HBV replication but cannot eliminate chronic HBV infection due to the persistence of covalently closed circular (ccc) DNA that sustains viral replication. The CRISPR/Cas9 system is a novel genome-editing tool that enables precise gene disruption and inactivation. With high efficiency and simplicity, the CRISPR/Cas9 system has been utilized in multiple studies to disrupt the HBV genome specifically, eliciting varying anti-HBV effects both <i>in vitro</i> and <i>in vivo</i>. Additionally, multi-locus gene targeting has shown enhanced antiviral activity, paving the way for combination therapy to disrupt and inactivate HBV cccDNA as well as integrated HBV DNA. Despite its promising antiviral effects, this technology faces several challenges that need to be overcome before its clinical application, i.e., off-target effects and <i>in vivo</i> drug delivery. As such, there is a need for improvement in CRISPR/Cas9 efficiency, specificity, versatility, and delivery. Here, we critically review the recent literature describing the tools employed in designing guide RNAs (gRNAs) targeting HBV genomes, the vehicles used for expressing and delivering CRISPR/Cas9 components, the models used for evaluating CRISPR-mediated HBV gene disruption, the methods used for assessing antiviral and off-target effects induced by CRISPR/Cas9-mediated HBV gene disruption, and the prospects of future directions and challenges in leveraging this HBV gene-editing approach, to advance the HBV treatment toward a clinical cure.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11496132/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142514123","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Use of paired Cas9-NG nickase and truncated sgRNAs for single-nucleotide microbial genome editing. 使用成对的 Cas9-NG 缺口酶和截短的 sgRNA 进行单核苷酸微生物基因组编辑。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-26 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1471720
Song Hee Jeong, Ho Joung Lee, Sang Jun Lee

The paired nickases approach, which utilizes clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated proteins (Cas) nickase and dual guide RNA, has the advantage of reducing off-target effects by being able to double the target sequence. In this study, our research utilized the Cas9-NG nickase variant to minimize PAM sequence constraints, enabling the generation of paired nicks at desired genomic loci. We performed a systematic investigation into the formation sites for double nicks and the design of donor DNA within a bacterial model system. Although we successfully identified the conditions necessary for the effective formation of double nicks in vivo, achieving single-nucleotide level editing directly at the target sites in the genome proved challenging. Nonetheless, our experiments revealed that efficient editing at the single-nucleotide level was achievable on target DNA sequences that are hybridized with 5'-end-truncated dual single-guide RNAs (sgRNAs). Our findings contribute to a deeper understanding of the paired nickases approach, offering a single-mismatch intolerance design strategy for accurate nucleotide editing. This strategy not only enhances the precision of genome editing but also marks a significant step forward in the development of nickase-derived genome editing technologies.

配对切口酶方法利用聚类规则间隔短回文重复序列(CRISPR)-CRISPR相关蛋白(Cas)切口酶和双引导RNA,其优点是能够加倍靶序列,从而减少脱靶效应。在本研究中,我们利用Cas9-NG切口酶变体最大程度地减少了PAM序列限制,从而在所需的基因组位点上生成了成对的切口。我们在细菌模型系统中对双缺口的形成位点和供体 DNA 的设计进行了系统研究。虽然我们成功地确定了在体内有效形成双缺口的必要条件,但直接在基因组的目标位点实现单核苷酸水平的编辑证明具有挑战性。不过,我们的实验表明,在与 5'-end-truncated 双单导 RNA(sgRNA)杂交的目标 DNA 序列上,可以实现单核苷酸水平的高效编辑。我们的研究结果有助于加深对成对缺口酶方法的理解,为精确的核苷酸编辑提供了一种单错配不容忍设计策略。这一策略不仅提高了基因组编辑的精确度,而且标志着镍酶衍生基因组编辑技术的发展向前迈出了重要一步。
{"title":"Use of paired Cas9-NG nickase and truncated sgRNAs for single-nucleotide microbial genome editing.","authors":"Song Hee Jeong, Ho Joung Lee, Sang Jun Lee","doi":"10.3389/fgeed.2024.1471720","DOIUrl":"10.3389/fgeed.2024.1471720","url":null,"abstract":"<p><p>The paired nickases approach, which utilizes clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated proteins (Cas) nickase and dual guide RNA, has the advantage of reducing off-target effects by being able to double the target sequence. In this study, our research utilized the Cas9-NG nickase variant to minimize PAM sequence constraints, enabling the generation of paired nicks at desired genomic loci. We performed a systematic investigation into the formation sites for double nicks and the design of donor DNA within a bacterial model system. Although we successfully identified the conditions necessary for the effective formation of double nicks <i>in vivo</i>, achieving single-nucleotide level editing directly at the target sites in the genome proved challenging. Nonetheless, our experiments revealed that efficient editing at the single-nucleotide level was achievable on target DNA sequences that are hybridized with 5'-end-truncated dual single-guide RNAs (sgRNAs). Our findings contribute to a deeper understanding of the paired nickases approach, offering a single-mismatch intolerance design strategy for accurate nucleotide editing. This strategy not only enhances the precision of genome editing but also marks a significant step forward in the development of nickase-derived genome editing technologies.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11464485/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142402119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Making gene editing accessible in resource limited environments: recommendations to guide a first-time user. 在资源有限的环境中实现基因编辑:指导首次使用者的建议。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-25 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1464531
Shivani Goolab, Janine Scholefield

The designer nuclease, CRISPR-Cas9 system has advanced the field of genome engineering owing to its programmability and ease of use. The application of these molecular scissors for genome engineering earned the developing researchers the Nobel prize in Chemistry in the year 2020. At present, the potential of this technology to improve global challenges continues to grow exponentially. CRISPR-Cas9 shows promise in the recent advances made in the Global North such as the FDA-approved gene therapy for the treatment of sickle cell anaemia and β-thalassemia and the gene editing of porcine kidney for xenotransplantation into humans affected by end-stage kidney failure. Limited resources, low government investment with an allocation of 1% of gross domestic production to research and development including a shortage of skilled professionals and lack of knowledge may preclude the use of this revolutionary technology in the Global South where the countries involved have reduced science and technology budgets. Focusing on the practical application of genome engineering, successful genetic manipulation is not easily accomplishable and is influenced by the chromatin landscape of the target locus, guide RNA selection, the experimental design including the profiling of the gene edited cells, which impacts the overall outcome achieved. Our assessment primarily delves into economical approaches of performing efficient genome engineering to support the first-time user restricted by limited resources with the aim of democratizing the use of the technology across low- and middle-income countries. Here we provide a comprehensive overview on existing experimental techniques, the significance for target locus analysis and current pitfalls such as the underrepresentation of global genetic diversity. Several perspectives of genome engineering approaches are outlined, which can be adopted in a resource limited setting to enable a higher success rate of genome editing-based innovations in low- and middle-income countries.

由于其可编程性和易用性,CRISPR-Cas9 系统这种设计型核酸酶推动了基因组工程领域的发展。这些分子剪刀在基因组工程中的应用为研发人员赢得了 2020 年的诺贝尔化学奖。目前,这项技术在改善全球挑战方面的潜力仍在成倍增长。CRISPR-Cas9 最近在全球北方地区取得的进展显示了其前景,如美国食品及药物管理局批准的用于治疗镰状细胞性贫血和β-地中海贫血症的基因疗法,以及用于异种移植到受终末期肾衰竭影响的人体内的猪肾基因编辑。资源有限、政府对研发的投资仅占国内生产总值的 1%,包括缺乏专业技术人员和知识,这些因素都可能阻碍这一革命性技术在全球南方国家的应用,因为这些国家的科技预算减少。就基因组工程的实际应用而言,成功的基因操作并非易事,它受到目标基因座染色质景观、指导 RNA 选择、实验设计(包括基因编辑细胞的分析)的影响,从而影响到所取得的总体成果。我们的评估主要深入研究高效基因组工程的经济方法,以支持受限于有限资源的首次使用者,目的是在低收入和中等收入国家普及该技术的使用。在此,我们全面概述了现有的实验技术、目标基因座分析的意义以及目前存在的缺陷,如全球遗传多样性代表性不足。我们还概述了基因组工程方法的几个方面,这些方法可以在资源有限的情况下采用,从而提高中低收入国家基于基因组编辑的创新的成功率。
{"title":"Making gene editing accessible in resource limited environments: recommendations to guide a first-time user.","authors":"Shivani Goolab, Janine Scholefield","doi":"10.3389/fgeed.2024.1464531","DOIUrl":"10.3389/fgeed.2024.1464531","url":null,"abstract":"<p><p>The designer nuclease, CRISPR-Cas9 system has advanced the field of genome engineering owing to its programmability and ease of use. The application of these molecular scissors for genome engineering earned the developing researchers the Nobel prize in Chemistry in the year 2020. At present, the potential of this technology to improve global challenges continues to grow exponentially. CRISPR-Cas9 shows promise in the recent advances made in the Global North such as the FDA-approved gene therapy for the treatment of sickle cell anaemia and β-thalassemia and the gene editing of porcine kidney for xenotransplantation into humans affected by end-stage kidney failure. Limited resources, low government investment with an allocation of 1% of gross domestic production to research and development including a shortage of skilled professionals and lack of knowledge may preclude the use of this revolutionary technology in the Global South where the countries involved have reduced science and technology budgets. Focusing on the practical application of genome engineering, successful genetic manipulation is not easily accomplishable and is influenced by the chromatin landscape of the target locus, guide RNA selection, the experimental design including the profiling of the gene edited cells, which impacts the overall outcome achieved. Our assessment primarily delves into economical approaches of performing efficient genome engineering to support the first-time user restricted by limited resources with the aim of democratizing the use of the technology across low- and middle-income countries. Here we provide a comprehensive overview on existing experimental techniques, the significance for target locus analysis and current pitfalls such as the underrepresentation of global genetic diversity. Several perspectives of genome engineering approaches are outlined, which can be adopted in a resource limited setting to enable a higher success rate of genome editing-based innovations in low- and middle-income countries.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11461239/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142395669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Global regulatory policies for animal biotechnology: overview, opportunities and challenges. 全球动物生物技术监管政策:概述、机遇与挑战。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-24 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1467080
Diane Wray-Cahen, Eric Hallerman, Mark Tizard

Genome editing (GnEd) has the potential to provide many benefits to animal agriculture, offering a means for achieving rapid growth, disease resistance, and novel phenotypes. The technology has the potential to be useful for rapidly incorporating traits into existing selectively bred animals without the need for crossbreeding and backcrossing. Yet only four products from animals created via biotechnology, all growth-enhanced fishes, have reached commercialization and only on a limited scale. The past failure of genetically engineered (or GM) products to reach conventional producers can largely be attributed to the high cost of meeting GMO regulatory requirements. We review the history of GMO regulations internationally, noting the influence of Codex Alimentarius on the development of many existing regulatory frameworks. We highlight new regulatory approaches for GnEd organisms, first developed by Argentina, and the adoption of similar approaches by other countries. Such new regulatory approaches allow GnEd organisms that could have been developed by conventional means to be regulated under the same rules as conventional organisms and in the future is likely to enhance the opportunity for biotech animals to enter production. Treating certain GnEd products as conventional has had a large impact on the variety of biotechnological innovations successfully navigating regulatory processes. We suggest that for the full potential of GnEd technologies to be realized, enabling public policies are needed to facilitate use of GnEd as a breeding tool to incorporate new traits within existing animal breeding programs, rather than only a tool to create distinct new products.

基因组编辑(GnEd)有可能为畜牧业带来许多益处,提供一种实现快速生长、抗病和新表型的手段。该技术有可能在无需杂交和回交的情况下,将性状快速融入现有的选育动物中。然而,目前只有四种通过生物技术培育的动物产品实现了商业化,而且规模有限,均为生长增强型鱼类。转基因产品过去未能进入传统生产商的视野,在很大程度上归因于满足转基因生物监管要求的高昂成本。我们回顾了国际转基因生物监管的历史,注意到《食品法典》对许多现有监管框架发展的影响。我们强调了阿根廷首先制定的针对转基因生物的新监管方法,以及其他国家对类似方法的采用。这种新的监管方法使本可通过传统方法开发的 GnEd 生物在与传统生物相同的规则下接受监管,未来很可能会增加生物技术动物进入生产领域的机会。将某些 GnEd 产品视为常规产品对成功通过监管程序的各种生物技术创新产生了很大影响。我们建议,为充分发挥 GnEd 技术的潜力,需要制定有利的公共政策,促进将 GnEd 作为育种工具,将新性状纳入现有的动物育种计划,而不仅仅是作为创造独特新产品的工具。
{"title":"Global regulatory policies for animal biotechnology: overview, opportunities and challenges.","authors":"Diane Wray-Cahen, Eric Hallerman, Mark Tizard","doi":"10.3389/fgeed.2024.1467080","DOIUrl":"https://doi.org/10.3389/fgeed.2024.1467080","url":null,"abstract":"<p><p>Genome editing (GnEd) has the potential to provide many benefits to animal agriculture, offering a means for achieving rapid growth, disease resistance, and novel phenotypes. The technology has the potential to be useful for rapidly incorporating traits into existing selectively bred animals without the need for crossbreeding and backcrossing. Yet only four products from animals created via biotechnology, all growth-enhanced fishes, have reached commercialization and only on a limited scale. The past failure of genetically engineered (or GM) products to reach conventional producers can largely be attributed to the high cost of meeting GMO regulatory requirements. We review the history of GMO regulations internationally, noting the influence of Codex Alimentarius on the development of many existing regulatory frameworks. We highlight new regulatory approaches for GnEd organisms, first developed by Argentina, and the adoption of similar approaches by other countries. Such new regulatory approaches allow GnEd organisms that could have been developed by conventional means to be regulated under the same rules as conventional organisms and in the future is likely to enhance the opportunity for biotech animals to enter production. Treating certain GnEd products as conventional has had a large impact on the variety of biotechnological innovations successfully navigating regulatory processes. We suggest that for the full potential of GnEd technologies to be realized, enabling public policies are needed to facilitate use of GnEd as a breeding tool to incorporate new traits within existing animal breeding programs, rather than only a tool to create distinct new products.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459211/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142395668","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The 4Fs of cotton: genome editing of cotton for fiber, food, feed, and fuel to achieve zero hunger. 棉花的 4Fs:基因组编辑棉花纤维、食品、饲料和燃料,实现零饥饿。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-09-12 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1401088
Muhammad Sulyman Saleem, Sultan Habibullah Khan, Aftab Ahmad, Iqrar Ahmad Rana, Zunaira Afzal Naveed, Azeem Iqbal Khan

Cotton is globally known for its high-priority cellulose-rich natural fiber. In addition to providing fiber for the textile industry, it is an important source material for edible oil, livestock feed, and fuel products. Global warming and the growing population are the major challenges to the world's agriculture and the potential risks to food security. In this context, improving output traits in cotton is necessary to achieve sustainable cotton production. During the last few years, high throughput omics techniques have aided in identifying crucial genes associated with traits of cotton fiber, seed, and plant architecture which could be targeted with more precision and efficiency through the CIRPSR/Cas-mediated genome editing technique. The various CRISPR/Cas systems such as CRISPR/Cas9, CRISPR/nCas9, and CRISPR/Cas12a have been employed to edit cotton genes associated with a wide range of traits including fiber length, flowering, leaf colour, rooting, seed oil, plant architecture, gossypol content, somatic embryogenesis, and biotic and abiotic stresses tolerance, highlighting its effectiveness in editing the cotton genome. Thus, CRISPR/Cas-mediated genome editing has emerged as a technique of choice to tailor crop phenotypes for better yield potential and environmental resilience. The review covers a comprehensive analysis of cotton phenotypic traits and their improvement with the help of the latest genome editing tools to improve fiber, food, feed, and fuel-associated genes of cotton to ensure food security.

棉花因其富含纤维素的天然纤维而闻名于世。除了为纺织业提供纤维外,它还是食用油、牲畜饲料和燃料产品的重要原料。全球变暖和人口增长是世界农业面临的主要挑战,也是粮食安全的潜在风险。在这种情况下,要实现棉花的可持续生产,就必须改善棉花的产出性状。在过去几年中,高通量组学技术帮助确定了与棉花纤维、种子和植株结构等性状相关的关键基因,通过 CIRPSR/Cas 介导的基因组编辑技术,可以更精确、更高效地确定这些基因。各种 CRISPR/Cas 系统(如 CRISPR/Cas9、CRISPR/nCas9 和 CRISPR/Cas12a)已被用于编辑与纤维长度、开花、叶色、生根、籽油、植株结构、棉酚含量、体细胞胚胎发生以及生物和非生物胁迫耐受性等多种性状相关的棉花基因,凸显了其在编辑棉花基因组方面的有效性。因此,CRISPR/Cas 介导的基因组编辑已成为定制作物表型以提高产量潜力和环境适应能力的首选技术。本综述全面分析了棉花的表型性状,并借助最新的基因组编辑工具对其进行改良,以改良棉花的纤维、食品、饲料和燃料相关基因,确保粮食安全。
{"title":"The 4Fs of cotton: genome editing of cotton for fiber, food, feed, and fuel to achieve zero hunger.","authors":"Muhammad Sulyman Saleem, Sultan Habibullah Khan, Aftab Ahmad, Iqrar Ahmad Rana, Zunaira Afzal Naveed, Azeem Iqbal Khan","doi":"10.3389/fgeed.2024.1401088","DOIUrl":"https://doi.org/10.3389/fgeed.2024.1401088","url":null,"abstract":"<p><p>Cotton is globally known for its high-priority cellulose-rich natural fiber. In addition to providing fiber for the textile industry, it is an important source material for edible oil, livestock feed, and fuel products. Global warming and the growing population are the major challenges to the world's agriculture and the potential risks to food security. In this context, improving output traits in cotton is necessary to achieve sustainable cotton production. During the last few years, high throughput omics techniques have aided in identifying crucial genes associated with traits of cotton fiber, seed, and plant architecture which could be targeted with more precision and efficiency through the CIRPSR/Cas-mediated genome editing technique. The various CRISPR/Cas systems such as CRISPR/Cas9, CRISPR/nCas9, and CRISPR/Cas12a have been employed to edit cotton genes associated with a wide range of traits including fiber length, flowering, leaf colour, rooting, seed oil, plant architecture, gossypol content, somatic embryogenesis, and biotic and abiotic stresses tolerance, highlighting its effectiveness in editing the cotton genome. Thus, CRISPR/Cas-mediated genome editing has emerged as a technique of choice to tailor crop phenotypes for better yield potential and environmental resilience. The review covers a comprehensive analysis of cotton phenotypic traits and their improvement with the help of the latest genome editing tools to improve fiber, food, feed, and fuel-associated genes of cotton to ensure food security.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11424549/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142333831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In vivo liver targeted genome editing as therapeutic approach: progresses and challenges. 作为治疗方法的体内肝脏靶向基因组编辑:进展与挑战。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-23 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1458037
Chiara Simoni, Elena Barbon, Andrés F Muro, Alessio Cantore

The liver is an essential organ of the body that performs several vital functions, including the metabolism of biomolecules, foreign substances, and toxins, and the production of plasma proteins, such as coagulation factors. There are hundreds of genetic disorders affecting liver functions and, for many of them, the only curative option is orthotopic liver transplantation, which nevertheless entails many risks and long-term complications. Some peculiar features of the liver, such as its large blood flow supply and the tolerogenic immune environment, make it an attractive target for in vivo gene therapy approaches. In recent years, several genome-editing tools mainly based on the clustered regularly interspaced short palindromic repeats associated protein 9 (CRISPR-Cas9) system have been successfully exploited in the context of liver-directed preclinical or clinical therapeutic applications. These include gene knock-out, knock-in, activation, interference, or base and prime editing approaches. Despite many achievements, important challenges still need to be addressed to broaden clinical applications, such as the optimization of the delivery methods, the improvement of the editing efficiency, and the risk of on-target or off-target unwanted effects and chromosomal rearrangements. In this review, we highlight the latest progress in the development of in vivo liver-targeted genome editing approaches for the treatment of genetic disorders. We describe the technological advancements that are currently under investigation, the challenges to overcome for clinical applicability, and the future perspectives of this technology.

肝脏是人体的重要器官,具有多种重要功能,包括代谢生物大分子、外来物质和毒素,以及生成凝血因子等血浆蛋白。影响肝脏功能的遗传性疾病有数百种,其中许多疾病的唯一治疗方法是肝移植,但肝移植存在许多风险和长期并发症。肝脏的一些特殊功能,如大量血流供应和耐受性免疫环境,使其成为体内基因治疗方法的一个有吸引力的靶点。近年来,几种主要基于聚类规则间隔短回文重复序列相关蛋白9(CRISPR-Cas9)系统的基因组编辑工具已成功应用于肝脏定向临床前或临床治疗。这些方法包括基因敲除、敲入、激活、干扰或碱基和质粒编辑方法。尽管取得了许多成就,但要扩大临床应用,仍需应对一些重要挑战,如优化传递方法、提高编辑效率、靶上或靶下意外效应和染色体重排的风险等。在本综述中,我们将重点介绍用于治疗遗传疾病的体内肝脏靶向基因组编辑方法的最新进展。我们介绍了目前正在研究的技术进展、临床应用需要克服的挑战以及这项技术的未来前景。
{"title":"<i>In vivo</i> liver targeted genome editing as therapeutic approach: progresses and challenges.","authors":"Chiara Simoni, Elena Barbon, Andrés F Muro, Alessio Cantore","doi":"10.3389/fgeed.2024.1458037","DOIUrl":"10.3389/fgeed.2024.1458037","url":null,"abstract":"<p><p>The liver is an essential organ of the body that performs several vital functions, including the metabolism of biomolecules, foreign substances, and toxins, and the production of plasma proteins, such as coagulation factors. There are hundreds of genetic disorders affecting liver functions and, for many of them, the only curative option is orthotopic liver transplantation, which nevertheless entails many risks and long-term complications. Some peculiar features of the liver, such as its large blood flow supply and the tolerogenic immune environment, make it an attractive target for <i>in vivo</i> gene therapy approaches. In recent years, several genome-editing tools mainly based on the clustered regularly interspaced short palindromic repeats associated protein 9 (CRISPR-Cas9) system have been successfully exploited in the context of liver-directed preclinical or clinical therapeutic applications. These include gene knock-out, knock-in, activation, interference, or base and prime editing approaches. Despite many achievements, important challenges still need to be addressed to broaden clinical applications, such as the optimization of the delivery methods, the improvement of the editing efficiency, and the risk of on-target or off-target unwanted effects and chromosomal rearrangements. In this review, we highlight the latest progress in the development of <i>in vivo</i> liver-targeted genome editing approaches for the treatment of genetic disorders. We describe the technological advancements that are currently under investigation, the challenges to overcome for clinical applicability, and the future perspectives of this technology.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11378722/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142157203","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genetic engineering and genome editing technologies as catalyst for Africa’s food security: the case of plant biotechnology in Nigeria 基因工程和基因组编辑技术是非洲粮食安全的催化剂:尼日利亚的植物生物技术案例
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-09 DOI: 10.3389/fgeed.2024.1398813
M. S. Adegbaju, Titilayo Ajose, Ifeoluwa Elizabeth Adegbaju, Temitayo Omosebi, Shakirat Oloruntoyin Ajenifujah-Solebo, Olaitan Yetunde Falana, Olufunke Bolatito Shittu, C. Adetunji, Olalekan Akinbo
Many African countries are unable to meet the food demands of their growing population and the situation is worsened by climate change and disease outbreaks. This issue of food insecurity may lead to a crisis of epic proportion if effective measures are not in place to make more food available. Thus, deploying biotechnology towards the improvement of existing crop varieties for tolerance or resistance to both biotic and abiotic stresses is crucial to increasing crop production. In order to optimize crop production, several African countries have implemented strategies to make the most of this innovative technology. For example, Nigerian government has implemented the National Biotechnology Policy to facilitate capacity building, research, bioresource development and commercialization of biotechnology products for over two decades. Several government ministries, research centers, universities, and agencies have worked together to implement the policy, resulting in the release of some genetically modified crops to farmers for cultivation and Commercialization, which is a significant accomplishment. However, the transgenic crops were only brought to Nigeria for confined field trials; the manufacturing of the transgenic crops took place outside the country. This may have contributed to the suspicion of pressure groups and embolden proponents of biotechnology as an alien technology. Likewise, this may also be the underlying issue preventing the adoption of biotechnology products in other African countries. It is therefore necessary that African universities develop capacity in various aspects of biotechnology, to continuously train indigenous scientists who can generate innovative ideas tailored towards solving problems that are peculiar to respective country. Therefore, this study intends to establish the role of genetic engineering and genome editing towards the achievement of food security in Africa while using Nigeria as a case study. In our opinion, biotechnology approaches will not only complement conventional breeding methods in the pursuit of crop improvements, but it remains a viable and sustainable means of tackling specific issues hindering optimal crop production. Furthermore, we suggest that financial institutions should offer low-interest loans to new businesses. In order to promote the growth of biotechnology products, especially through the creation of jobs and revenues through molecular farming.
许多非洲国家无法满足日益增长的人口对粮食的需求,而气候变化和疾病爆发又使情况更加恶化。如果不采取有效措施提供更多的粮食,粮食不安全问题可能会导致史诗般的危机。因此,利用生物技术改良现有作物品种,使其能够耐受或抵御生物和非生物压力,对于提高作物产量至关重要。为了优化作物生产,一些非洲国家已经实施了充分利用这一创新技术的战略。例如,尼日利亚政府二十多年来一直在实施国家生物技术政策,以促进生物技术产品的能力建设、研究、生物资源开发和商业化。多个政府部委、研究中心、大学和机构共同努力实施该政策,最终向农民发放了一些转基因作物,供其种植和商业化,这是一项重大成就。然而,转基因作物只是被运到尼日利亚进行有限的田间试验;转基因作物的生产是在国外进行的。这可能导致了压力集团的怀疑,并使生物技术的支持者更加胆大妄为,将其视为外来技术。同样,这也可能是阻碍其他非洲国家采用生物技术产品的根本问题。因此,非洲的大学有必要发展生物技术各方面的能力,不断培养本土科学家,使他们能够提出创新想法,解决各自国家特有的问题。因此,本研究以尼日利亚为案例,旨在确定基因工程和基因组编辑对实现非洲粮食安全的作用。我们认为,生物技术方法不仅可以补充传统的作物改良育种方法,而且仍然是解决阻碍作物优化生产的具体问题的可行和可持续的手段。此外,我们建议金融机构为新企业提供低息贷款。以促进生物技术产品的发展,特别是通过分子农业创造就业和收入。
{"title":"Genetic engineering and genome editing technologies as catalyst for Africa’s food security: the case of plant biotechnology in Nigeria","authors":"M. S. Adegbaju, Titilayo Ajose, Ifeoluwa Elizabeth Adegbaju, Temitayo Omosebi, Shakirat Oloruntoyin Ajenifujah-Solebo, Olaitan Yetunde Falana, Olufunke Bolatito Shittu, C. Adetunji, Olalekan Akinbo","doi":"10.3389/fgeed.2024.1398813","DOIUrl":"https://doi.org/10.3389/fgeed.2024.1398813","url":null,"abstract":"Many African countries are unable to meet the food demands of their growing population and the situation is worsened by climate change and disease outbreaks. This issue of food insecurity may lead to a crisis of epic proportion if effective measures are not in place to make more food available. Thus, deploying biotechnology towards the improvement of existing crop varieties for tolerance or resistance to both biotic and abiotic stresses is crucial to increasing crop production. In order to optimize crop production, several African countries have implemented strategies to make the most of this innovative technology. For example, Nigerian government has implemented the National Biotechnology Policy to facilitate capacity building, research, bioresource development and commercialization of biotechnology products for over two decades. Several government ministries, research centers, universities, and agencies have worked together to implement the policy, resulting in the release of some genetically modified crops to farmers for cultivation and Commercialization, which is a significant accomplishment. However, the transgenic crops were only brought to Nigeria for confined field trials; the manufacturing of the transgenic crops took place outside the country. This may have contributed to the suspicion of pressure groups and embolden proponents of biotechnology as an alien technology. Likewise, this may also be the underlying issue preventing the adoption of biotechnology products in other African countries. It is therefore necessary that African universities develop capacity in various aspects of biotechnology, to continuously train indigenous scientists who can generate innovative ideas tailored towards solving problems that are peculiar to respective country. Therefore, this study intends to establish the role of genetic engineering and genome editing towards the achievement of food security in Africa while using Nigeria as a case study. In our opinion, biotechnology approaches will not only complement conventional breeding methods in the pursuit of crop improvements, but it remains a viable and sustainable means of tackling specific issues hindering optimal crop production. Furthermore, we suggest that financial institutions should offer low-interest loans to new businesses. In order to promote the growth of biotechnology products, especially through the creation of jobs and revenues through molecular farming.","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141664011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advancing crop disease resistance through genome editing: a promising approach for enhancing agricultural production. 通过基因组编辑提高作物抗病性:提高农业产量的有效方法。
IF 4.9 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-06-26 eCollection Date: 2024-01-01 DOI: 10.3389/fgeed.2024.1399051
Subaya Manzoor, Sajad Un Nabi, Tariq Rasool Rather, Gousia Gani, Zahoor Ahmad Mir, Ab Waheed Wani, Sajad Ali, Anshika Tyagi, Nazia Manzar

Modern agriculture has encountered several challenges in achieving constant yield stability especially due to disease outbreaks and lack of long-term disease-resistant crop cultivars. In the past, disease outbreaks in economically important crops had a major impact on food security and the economy. On the other hand climate-driven emergence of new pathovars or changes in their host specificity further poses a serious threat to sustainable agriculture. At present, chemical-based control strategies are frequently used to control microbial pathogens and pests, but they have detrimental impact on the environment and also resulted in the development of resistant phyto-pathogens. As a replacement, cultivating engineered disease-resistant crops can help to minimize the negative impact of regular pesticides on agriculture and the environment. Although traditional breeding and genetic engineering have been instrumental in crop disease improvement but they have certain limitations such as labour intensity, time consumption, and low efficiency. In this regard, genome editing has emerged as one of the potential tools for improving disease resistance in crops by targeting multiple traits with more accuracy and efficiency. For instance, genome editing techniques, such as CRISPR/Cas9, CRISPR/Cas13, base editing, TALENs, ZFNs, and meganucleases, have proved successful in improving disease resistance in crops through targeted mutagenesis, gene knockouts, knockdowns, modifications, and activation of target genes. CRISPR/Cas9 is unique among these techniques because of its remarkable efficacy, low risk of off-target repercussions, and ease of use. Some primary targets for developing CRISPR-mediated disease-resistant crops are host-susceptibility genes (the S gene method), resistance genes (R genes) and pathogen genetic material that prevents their development, broad-spectrum disease resistance. The use of genome editing methods has the potential to notably ameliorate crop disease resistance and transform agricultural practices in the future. This review highlights the impact of phyto-pathogens on agricultural productivity. Next, we discussed the tools for improving disease resistance while focusing on genome editing. We provided an update on the accomplishments of genome editing, and its potential to improve crop disease resistance against bacterial, fungal and viral pathogens in different crop systems. Finally, we highlighted the future challenges of genome editing in different crop systems for enhancing disease resistance.

现代农业在实现持续稳产方面遇到了一些挑战,特别是由于疾病爆发和缺乏长期抗病的作物栽培品种。过去,重要经济作物爆发病害对粮食安全和经济造成了重大影响。另一方面,气候导致的新病原菌的出现或宿主特异性的改变进一步对可持续农业构成严重威胁。目前,人们经常使用化学防治策略来控制微生物病原体和害虫,但这些策略会对环境造成不利影响,还会导致抗性植物病原体的产生。作为替代,培育工程抗病作物有助于最大限度地减少常规杀虫剂对农业和环境的负面影响。虽然传统育种和基因工程在改良作物病害方面发挥了重要作用,但它们也有一定的局限性,如劳动强度大、耗时长、效率低等。为此,基因组编辑技术应运而生,它可以更准确、更高效地针对作物的多个性状,成为提高作物抗病性的潜在工具之一。例如,CRISPR/Cas9、CRISPR/Cas13、碱基编辑、TALENs、ZFNs 和巨核酸酶等基因组编辑技术,通过靶向诱变、基因敲除、敲除、修饰和激活目标基因,在提高作物抗病性方面取得了成功。CRISPR/Cas9 在这些技术中独树一帜,因为它功效显著、脱靶风险低、易于使用。开发 CRISPR 介导的抗病作物的一些主要目标是宿主易感基因(S 基因法)、抗性基因(R 基因)和阻止其发展的病原体遗传物质、广谱抗病性。基因组编辑方法的使用有可能显著改善作物的抗病性,并在未来改变农业生产方式。本综述强调了植物病原体对农业生产力的影响。接下来,我们讨论了提高抗病性的工具,同时重点介绍了基因组编辑。我们介绍了基因组编辑的最新成就,及其在不同作物系统中提高作物对细菌、真菌和病毒病原体的抗病性的潜力。最后,我们强调了基因组编辑在不同作物系统中提高抗病性的未来挑战。
{"title":"Advancing crop disease resistance through genome editing: a promising approach for enhancing agricultural production.","authors":"Subaya Manzoor, Sajad Un Nabi, Tariq Rasool Rather, Gousia Gani, Zahoor Ahmad Mir, Ab Waheed Wani, Sajad Ali, Anshika Tyagi, Nazia Manzar","doi":"10.3389/fgeed.2024.1399051","DOIUrl":"10.3389/fgeed.2024.1399051","url":null,"abstract":"<p><p>Modern agriculture has encountered several challenges in achieving constant yield stability especially due to disease outbreaks and lack of long-term disease-resistant crop cultivars. In the past, disease outbreaks in economically important crops had a major impact on food security and the economy. On the other hand climate-driven emergence of new pathovars or changes in their host specificity further poses a serious threat to sustainable agriculture. At present, chemical-based control strategies are frequently used to control microbial pathogens and pests, but they have detrimental impact on the environment and also resulted in the development of resistant phyto-pathogens. As a replacement, cultivating engineered disease-resistant crops can help to minimize the negative impact of regular pesticides on agriculture and the environment. Although traditional breeding and genetic engineering have been instrumental in crop disease improvement but they have certain limitations such as labour intensity, time consumption, and low efficiency. In this regard, genome editing has emerged as one of the potential tools for improving disease resistance in crops by targeting multiple traits with more accuracy and efficiency. For instance, genome editing techniques, such as CRISPR/Cas9, CRISPR/Cas13, base editing, TALENs, ZFNs, and meganucleases, have proved successful in improving disease resistance in crops through targeted mutagenesis, gene knockouts, knockdowns, modifications, and activation of target genes. CRISPR/Cas9 is unique among these techniques because of its remarkable efficacy, low risk of off-target repercussions, and ease of use. Some primary targets for developing CRISPR-mediated disease-resistant crops are host-susceptibility genes (the S gene method), resistance genes (R genes) and pathogen genetic material that prevents their development, broad-spectrum disease resistance. The use of genome editing methods has the potential to notably ameliorate crop disease resistance and transform agricultural practices in the future. This review highlights the impact of phyto-pathogens on agricultural productivity. Next, we discussed the tools for improving disease resistance while focusing on genome editing. We provided an update on the accomplishments of genome editing, and its potential to improve crop disease resistance against bacterial, fungal and viral pathogens in different crop systems. Finally, we highlighted the future challenges of genome editing in different crop systems for enhancing disease resistance.</p>","PeriodicalId":73086,"journal":{"name":"Frontiers in genome editing","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11234172/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141581734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Frontiers in genome editing
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