Acessibilidade / Reportar erro

Genome editing in maize: Toward improving complex traits in a global crop

Abstract

Recent advances in genome editing have enormously enhanced the effort to develop biotechnology crops for more sustainable food production. CRISPR/Cas, the most versatile genome-editing tool, has shown the potential to create genome modifications that range from gene knockout and gene expression pattern modulations to allele-specific changes in order to design superior genotypes harboring multiple improved agronomic traits. However, a frequent bottleneck is the delivery of CRISPR/Cas to crops that are less amenable to transformation and regeneration. Several technologies have recently been proposed to overcome transformation recalcitrance, including HI-Edit/IMGE and ectopic/transient expression of genes encoding morphogenic regulators. These technologies allow the eroding of the barriers that make crops inaccessible for genome editing. In this review, we discuss the advances in genome editing in crops with a particular focus on the use of technologies to improve complex traits such as water use efficiency, drought stress, and yield in maize.

Keywords:
CRISPR/Cas; maize transformation; transformation recalcitrance; multiplex genome editing; promoter editing

Introduction

As the global population grows, there is an increasing urgency for large-scale sustainable food production. The world population will reach 10 billion people by 2050, and although food production needs to increase proportionally, the ever-increasing effects of climate change threaten modern agriculture in an unprecedented manner (Gornall et al., 2010Gornall J, Betts R, Burke E, Clark R, Camp J, Willett K and Wiltshire A (2010) Implications of climate change for agricultural productivity in the early twenty-first century. Philos Trans R Soc Lond B Biol Sci 365:2973-2989.; Brás et al., 2021Brás TA, Seixas J, Carvalhais N and Jagermeyr J (2021) Severity of drought and heatwave crop losses tripled over the last five decades in Europe. Environ Res Lett 16:065012.). Maize (Zea mays L.) is one of the most important crops in the world, extensively used as food, feed, fuel, and raw material by several industries (Andorf et al., 2019Andorf C, Beavis WD, Hufford M, Smith S, Suza WP, Wang K, Woodhouse M, Yu J and Lübberstedt T (2019) Technological advances in maize breeding: past, present and future. Theor Appl Genet 132:817-849.). Importantly, variations in temperature, precipitation, and their interaction historically have had a large impact on global yields of maize and most other major crops (Lobell et al., 2011Lobell DB, Schlenker W and Costa-Roberts J (2011) Climate trends and global crop production since 1980. Science 333:616-620.; Ray et al., 2015Ray DK, Gerber JS, MacDonald GK and West PC (2015) Climate variation explains a third of global crop yield variability. Nat Commun 6:5989.; Daryanto et al., 2016Daryanto S, Wang L and Jacinthe PA (2016) Global synthesis of drought effects on maize and wheat production. PLoS One 11:e0156362.). As significant and recent examples, Brazil (the world’s third largest maize producer) experienced a reduction in its maize production of approximately 18 Mt and 23 Mt in the 2015/16 and 2020/21 growing seasons, corresponding to losses of approximately 21% compared to the 2014/15 and 2019/20 seasons, respectively (CONAB, 2021CONAB - Companhia Nacional de Abastecimento (2021) Acompanhamento de safra brasileira, CONAB - Companhia Nacional de Abastecimento (2021) Acompanhamento de safra brasileira, https://www.conab.gov.br/info-agro/safras/graos (accessed 12 Oct 2021).
https://www.conab.gov.br/info-agro/safra...
). Considering the average maize price from 2018 to 2021 (CEPEA-ESALQ, 2021CEPEA-ESALQ - Centro de Estudos Avançados em Economia Aplicada ESALQ (2021) Indicador do Milho Esalq/BM&FBovespa, CEPEA-ESALQ - Centro de Estudos Avançados em Economia Aplicada ESALQ (2021) Indicador do Milho Esalq/BM&FBovespa, https://www.cepea.esalq.usp.br/br/indicador/milho.aspx (accessed 12 Oct 2021).
https://www.cepea.esalq.usp.br/br/indica...
) these unrealized yields correspond to economic losses of approximately USD 3 and USD 5 billion. These crop failures occurred in years marked by pronounced drought (INMET, 2021INMET - Instituto Nacional de Meteorologia (2021) SPI - Índice de Precipitação Padronizada, INMET - Instituto Nacional de Meteorologia (2021) SPI - Índice de Precipitação Padronizada, https://portal.inmet.gov.br/servicos/spi-índice-de-precipitação-padronizada (accessed 12 Oct 2021).
https://portal.inmet.gov.br/servicos/spi...
), and resulted in poor yields in many of the largest producer geographies. Likewise, the 2012 drought in the U.S. (the world’s largest producer) resulted in similar yield reductions and spiking prices (Boyer et al., 2013Boyer JS, Schlegel A, Delmer D, Porter D, Warner D, Gruis F, Gaffney J, Habben J, Schussler J, Shanahan J et al. (2013) The U.S. drought of 2012 in perspective: a call to action. Glob Food Sec 2:139-143.). Thus, the continuous development of new maize cultivars aiming at better genetic adaptation, along with the adoption of improved agricultural practices, is crucial to minimize future losses resulting from the increased frequency, severity, and duration of stresses associated with global climate change (IPCC, 2022IPCC - The Intergovernmental Panel on Climate Change (2022) Climate Change 2022: Impacts, adaptation, and vulnerability. Sixth Assessment Report, Sixth Assessment Report, https://www.ipcc.ch/report/ar6/wg2/ (accessed 12 Oct 2021).
https://www.ipcc.ch/report/ar6/wg2/...
).

Yield and abiotic stress tolerance are complex traits usually strongly affected by the environment and associated with small-effect genomic loci. This complexity challenges dissecting the molecular mechanisms of gene actions and accurately measuring phenotypes, making it difficult to use genomic engineering tools to develop superior cultivars for those traits. Transgenic maize cultivars aiming at increased insect and herbicide tolerance have been on the market for decades, in contrast to only a few examples developed for complex traits (Yassitepe et al., 2021Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.). The difficulty of applying a transgenic approach to manipulate complex traits stable in several environments has limited the development of biotech cultivars that could be widely used (Simmons et al., 2021Simmons CR, Lafitte HR, Reimann KS, Brugière N, Roesler K, Albertsen MC, Greene TW and Habben JE (2021) Successes and insights of an industry biotech program to enhance maize agronomic traits. Plant Sci 307:110899.). However, recent advances in genome editing based on CRISPR/Cas (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR/associated proteins) technologies allow simultaneous mutations on multiple genes. Additionally, coupling genome editing with haploid induction increases the possibilities for genetically engineering complex traits across germplasms used in plant breeding programs.

Most genome editing studies in maize have been performed using genetic transformation protocols based on Agrobacterium tumefaciens or biolistic delivery methods in a couple of temperate genotypes suitable for Agrobacterium infection and regeneration (Kausch et al., 2021aKausch AP, Nelson-Vasilchik K, Tilelli M and Hague JP (2021a) Maize tissue culture, transformation, and genome editing. In Vitro Cell Dev Biol Plant 57:653-671.; Yassitepe et al., 2021Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.). However, the lack of a transformation protocol applicable to a wider range of genotypes and the typical low transformation efficiency hamper the broad application of genome editing in maize. Recently, solutions to overcome these constraints have been proposed, such as using morphogenic regulators (MRs) and in trans genome editing. Such approaches could be broadly used in maize breeding programs worldwide, including programs based on tropical germplasm. In this review, we discuss current advances in multiplex genome editing, base and prime editing, morphogenic regulators, and in trans genome editing tools that could potentially be applied to engineer complex traits.

Powerful genome editing toolkits for plant breeding

Although plant breeders have relied on molecular biology tools for introducing relevant agronomic traits into elite germplasm, transgenic approaches have limitations, such as the integration of foreign DNA into random sites of the host genome, which may raise regulatory concerns. Furthermore, developing new commercial genetically modified (GM) cultivars often requires a long and costly deregulation process, limiting this endeavor mostly to large multinational companies (Schmidt et al., 2020Schmidt SM, Belisle M and Frommer WB (2020) The evolving landscape around genome editing in agriculture. EMBO Rep 21:e50680.; Whelan et al., 2020Whelan AI, Gutti P and Lema MA (2020) Gene editing regulation and innovation economics. Front Bioeng Biotechnol 8:303.). Additionally, despite their significant beneficial impact on modern agriculture, the public still strongly rejects transgenic crops (Schmidt et al., 2020Schmidt SM, Belisle M and Frommer WB (2020) The evolving landscape around genome editing in agriculture. EMBO Rep 21:e50680.; Woźniak et al., 2021Woźniak E, Tyczewska A and Twardowski T (2021) A shift towards biotechnology: Social opinion in the EU. Trends Biotechnol 39:214-218.).

In this complex scenario, genome editing (GE) via CRISPR/Cas systems stands out as the most promising tool for the rapid development of new improved crop cultivars (Chen et al., 2019Chen K, Wang Y, Zhang R, Zhang H and Gao C (2019) CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu Rev Plant Biol 70:667-697.). This system’s accuracy in targeting specific sites, the opportunity to concomitantly alter multiple genes, and the possibility of segregating the CRISPR machinery by crossing while maintaining the edited loci are transforming plant breeding. In addition, CRISPR-based GE presents advantages over classical breeding and transgenic approaches, such as the opportunity to avoid GMO regulation by creating alleles indistinguishable from those produced by natural means, the possibility of rapidly developing stable homozygous lines mutated at precise loci, and relatively easy stacking of multiple advantageous traits via multiplex strategies.

Even though the original CRISPR/Cas system became a solid toolkit that revolutionized plant research and breeding, limitations such as the stochastic nature of the induced mutations prevented its application to specific cases. However, novel CRISPR/Cas-based technologies are constantly being developed, including the use of other Cas nucleases, such as Cas12a (also known as Cpf1), modified Cas nucleases with increased efficiency, deactivated Cas (dCas) or Cas nickases (nCas), chemically modified sgRNAs, and fusion of nCas or dCas to functional domains of other proteins (Chen et al., 2019Chen K, Wang Y, Zhang R, Zhang H and Gao C (2019) CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu Rev Plant Biol 70:667-697.; Anzalone et al., 2020Anzalone AV, Koblan LW and Liu DR (2020) Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 38:824-844.; Gao, 2021Gao C (2021) Genome engineering for crop improvement and future agriculture. Cell 184:1621-1635.). In addition, some CRISPR-based technologies such as base and prime editing allow for specific and precise modifications at the target locus, minimizing the randomness of indels caused by the non-homologous end joining (NHEJ) pathway (Anzalone et al., 2020Anzalone AV, Koblan LW and Liu DR (2020) Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 38:824-844.; Molla et al., 2021Molla KA, Sretenovic S, Bansal KC and Qi Y (2021) Precise plant genome editing using base editors and prime editors. Nat Plants 7:1166-1187.).

Additionally, improving CRISPR/Cas-based methods involves creating new modes of genome editing and broadening their applicability to plant breeding, allowing the improvement of traits controlled by multiple genes or even developing multiple traits at once. Next, we discuss some of these techniques and their potential application for plant breeding, summarized in Table 1.

Table 1 -
Advantages and limitations of diverse genome editing strategies and examples of their application in maize.

Multiplex genome editing

Small contributions of several genes determine important agronomic traits such as yield. Improvement of these complex quantitative traits requires hard and time-consuming selection and multi-crossing programs, which can take years before resulting in the development of new elite cultivars, even using modern techniques such as Genome Prediction, for example. CRISPR/Cas-based multiplex genome editing (MGE) enables the creation of new lines carrying multiple genome modifications in a few generations. This possibility represents an enormous advance over traditional plant breeding and transgenics, facilitating the stacking of advantageous traits (Najera et al., 2019Najera VA, Twyman RM, Christou P and Zhu C (2019) Applications of multiplex genome editing in higher plants. Curr Opin Biotechnol 59:93-102.).

MGE can be used to target similar and/or dissimilar sequences. For example, a single sgRNA can be used to target multiple genes with conserved regions (Figure 1A), whereas more than one sgRNA can target multiple genes (Figure 1B) or even multiple sites on the same gene (Figure 1C) (Najera et al., 2019Najera VA, Twyman RM, Christou P and Zhu C (2019) Applications of multiplex genome editing in higher plants. Curr Opin Biotechnol 59:93-102.).

Figure 1 -
Multiplex approaches for genome editing of multiple genes or multiple sites of a single gene. A. A single sgRNA is designed to target multiple genes with conserved domains/sequences (green boxes). B. Multiple sgRNAs are designed and simultaneously delivered, targeting sequences in different genes (colored boxes). C. Multiple sgRNAs may target different sections of a single gene, resulting in the deletion of specific regions between the target sites.

MGE is one of the most promising GE methods to improve complex traits. For example, by simultaneously knocking out the genes GW2, GW5, and TGW6, which are responsible for decreasing grain weight, new rice lines were created with ~30% higher grain weight (Xu et al., 2016Xu R, Yang Y, Qin R, Li H, Qiu C, Li L, Wei P and Yang J (2016) Rapid improvement of grain weight via highly efficient CRISPR/Cas9-mediated multiplex genome editing in rice. J Genet Genomics 43:529-532.). Additionally, by segregating the CRISPR machinery through crossing, transgene-free GE progeny was obtained, highlighting the advantage of this technique in developing new plant cultivars (Xu et al., 2016Xu W, Zhang C, Yang Y, Zhao S, Kang G, He X, Song J and Yang J (2020) Versatile nucleotides substitution in plant using an improved prime editing system. Mol Plant 13:675-678.). In another example, wheat grain length and weight were increased by targeting a conserved region of three homologs of the TaGASR7 gene with a single sgRNA. Transgene-free plants knocked out for all six alleles were obtained in a single generation using a transient expression setting for the CRISPR machinery (Zhang et al., 2016Zhang Y, Liang Z, Zong Y, Wang Y, Liu J, Chen K, Qiu JL and Gao C (2016) Efficient and transgene-free genome editing in wheat through transient expression of CRISPR/Cas9 DNA or RNA. Nat Commun 7:1-8.).

Different MGE strategies have been used in maize. For instance, Qi et al. (2016Qi W, Zhu T, Tian Z, Li C, Zhang W and Song R (2016) High-efficiency CRISPR/Cas9 multiplex gene editing using the glycine tRNA-processing system-based strategy in maize. BMC Biotechnol 16:58.) proposed an optimized tRNA-processing system-based method in maize in which up to four sgRNAs can be inserted into an array. The endogenous tRNA-processing system not only successfully processes the primary transcript from a more compact expression cassette but also seems to boost editing efficiency (85.7%-100%). The increase in editing efficiencies may be due to the A- and B-boxes in the tRNA sequences, which recruit transcription factors (White 2011White RJ (2011) Transcription by RNA polymerase III: More complex than we thought. Nat Rev Genet 12:459-463.; Xie et al., 2015Xie K, Minkenberg B and Yang Y (2015) Boosting CRISPR/Cas9 multiplex editing capability with the endogenous tRNA-processing system. Proc Natl Acad Sci 112:3570-3575.; Minkenberg et al., 2017Minkenberg B, Wheatley M and Yang Y (2017) CRISPR/Cas9-enabled multiplex genome editing and its application. Prog Mol Biol Transl Sci 149:111-132.). This result is interesting for maize GE since high editing efficiency is crucial when crop transformation efficiency is low, mitigating this considerable bottleneck. MGE in maize can also be performed with a larger number of sgRNAs. For instance, vectors harboring up to twelve individual sgRNA expression cassettes have been successfully used for the transformation of an “editor” (i.e., Cas9-expressing) maize line (Lorenzo et al., 2022Lorenzo CD, Debray K, Herwegh D, Develtere W, Impens L, Schaumont D, Vandeputte W, Aesaert S, Coussens G, Boe Y de et al. (2022) BREEDIT: A novel multiplex genome editing strategy to improve complex quantitative traits in maize. Plant Cell 35:218-238.). Gong et al. (2021Gong C, Huang S, Song R and Qi W (2021) Comparative study between the CRISPR/Cpf1 (Cas12a) and CRISPR/Cas9 systems for multiplex gene editing in maize. Agriculture 11:429.) compared the CRISPR/Cas12a versus the CRISPR/Cas9 system for MGE targeting the maize bZIP transcription factor Opaque2 (O2). Although CRISPR/Cas12a showed lower editing efficiency than CRISPR/Cas9 in the T0 and T1 generations, it led to a greater mutation variety in T2. In addition, the editing efficiency of the Cas12a-based system was positively correlated with the nuclease expression level, proving to be a valuable alternative for MGE in maize.

Although not yet broadly applied to maize, there are some reports of MGE applied to investigating and/or improving complex traits in this crop. For example, plant stature significantly impacts crop production, so the development of short-stature cereals was the foundation for the Green Revolution (GR) (Peng et al., 1999Peng J, Richards DE, Hartley NM, Murphy GP, Devos KM, Flintham JE, Beales J, Fish LJ, Worland AJ, Pelica F et al. (1999) “Green revolution” genes encode mutant gibberellin response modulators. Nature 400:256-261.). The dwarf and semidwarf cultivars have been shown to present several benefits, such as increased resistance to lodging caused by wind and rain, easy management in the field since plants are shorter and thus more accessible, and increased yield given that plants are more compact and require a smaller cultivated area. It has been shown that the application of gibberellin biosynthesis inhibitors during maize development leads to reduced plant height and improves water use efficiency and harvest index (grain mass to aboveground total mass ratio) (Hütsch and Schubert 2018Hütsch BW and Schubert S (2018) Maize harvest index and water use efficiency can be improved by inhibition of gibberellin biosynthesis. J Agron Crop Sci 204:209-218.). However, developing new plant lines with short stature by traditional breeding can be an overly long process. For example, 10 generations of directional selection were necessary to achieve maize plants with desirable heights (Teixeira et al., 2015Teixeira JEC, Weldekidan T, de Leon N, Flint-Garcia S, Holland JB, Lauter N, Murray SC, Xu W, Hessel DA, Kleintop AE et al. (2015) Hallauer’s Tusón: A decade of selection for tropical-to-temperate phenological adaptation in maize. Heredity (Edinb) 114:229-240.). In a simpler approach, semidwarf maize was generated by the knockout of ZmGA20ox3 using two sgRNAs (Zhang et al., 2020Zhang J, Zhang X, Chen R, Yang L, Fan K, Liu Y, Wang G, Ren Z and Liu Y (2020) Generation of transgene-free semidwarf maize plants by gene editing of GIBBERELLIN-OXIDASE20-3 using CRISPR/Cas9. Front Plant Sci 11:1048.).

MGE was also used for targeting multiple genes in maize. Because most cultivated maize plants are hybrids, detasseling is important for preventing self-pollination. Thus, identifying potential genes leading to male sterility is of great interest. Although several homologs of such genes are already known in maize, they often belong to families comprising genes with redundant functions. Through an MGE approach, Liu et al. (2022Liu X, Zhang S, Jiang Y, Yan T, Fang C, Hou Q, Wu S, Xie K, An X and Wan X (2022) Use of CRISPR/Cas9-based gene editing to simultaneously mutate multiple homologous genes required for pollen development and male fertility in maize. Cells 11:439.) were able to identify genes that can lead to male sterility when knocked out either individually or in combination with others. This study opens the possibility of selecting specific combinations of gene knockouts for establishing commercial lines.

Another interesting approach for MGE in maize has been recently described. The technique, dubbed BREEDIT, aims at improving complex traits controlled by many genes, such as drought resistance and yield. In BREEDIT, populations transformed with different sets of sgRNAs (twelve different sgRNAs on each set) are then crossed, stacking up mutations in an increasing number of genes. This approach is useful for both trait improvement as well as for the discovery of new genes contributing to a given trait of interest (Lorenzo et al., 2022Lorenzo CD, Debray K, Herwegh D, Develtere W, Impens L, Schaumont D, Vandeputte W, Aesaert S, Coussens G, Boe Y de et al. (2022) BREEDIT: A novel multiplex genome editing strategy to improve complex quantitative traits in maize. Plant Cell 35:218-238.).

Taken together, these works demonstrate the potential of MGE for the rapid improvement of complex traits, especially those controlled by numerous genes. It is important, however, to keep in mind the limitations of this approach. For example, designing a set of sgRNAs for specific genes belonging to conserved families can be challenging. Genotyping many genes can also require more sophisticated sequencing approaches, such as amplicon sequencing. Adding many sgRNAs into one vector can also potentially reduce transformation efficiency. An alternative method that circumvents the obstacles of managing excessively large plasmids is the direct delivery of ribonucleoprotein (RNP) complexes. Such RNPs are synthesized in vitro by combining Cas9 with mature sgRNA molecules (Woo et al., 2015Woo JWW, Kim JJSJJ-S, Kwon SI Il, Corvalán C, Cho SWW, Kim H, Kim S-TS-GTG, Kim S-TS-GTG, Choe S and Kim JJSJJ-S (2015) DNA-free genome editing in plants with preassembled CRISPR-Cas9 ribonucleoproteins. Nat Biotechnol 33:1162-1164.; Svitashev et al., 2016Svitashev S, Schwartz C, Lenderts B, Young JK and Mark Cigan A (2016) Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat Commun 7:13274.; Liang et al., 2017Liang Z, Chen K, Li T, Zhang Y, Wang Y, Zhao Q, Liu J, Zhang H, Liu C, Ran Y et al. (2017) Efficient DNA-free genome editing of bread wheat using CRISPR/Cas9 ribonucleoprotein complexes. Nat Commun 8:14261.). This approach is transgene-free since no foreign DNA is delivered to the plant. Successful RNP-mediated genome editing has also been shown for maize immature embryos co-bombarded with sgRNA:Cas RNPs, along with transformation-enhancing genes (morphogenic regulators) and a selectable/visible marker (MoPAT-DsRED) (Svitashev et al., 2016Svitashev S, Schwartz C, Lenderts B, Young JK and Mark Cigan A (2016) Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat Commun 7:13274.; Liang et al., 2017Liang Z, Chen K, Li T, Zhang Y, Wang Y, Zhao Q, Liu J, Zhang H, Liu C, Ran Y et al. (2017) Efficient DNA-free genome editing of bread wheat using CRISPR/Cas9 ribonucleoprotein complexes. Nat Commun 8:14261.). This promising result indicates the potential of using RNPs for MGE in maize while paving the way for its application in crops not prone to protoplast regeneration. Even though other CRISPR/Cas methods may generate transgene-free cultivars, crosses are needed to eliminate the exogenous editing system. In the case of RNPs, edited events will most likely bypass regulatory hurdles without additional efforts.

Promoter editing

Although coding sequences are usually the targets of choice for GE, other genetic elements, such as regulatory sequences, can also be targeted to modulate spatiotemporal gene expression patterns. For instance, genes playing essential roles in plant domestication have more stable expression patterns in cultivated species than in their wild relatives, suggesting that specific cis-regulatory elements (CREs) were selected during domestication (Lemmon et al., 2014Lemmon ZH, Bukowski R, Sun Q and Doebley JF (2014) The role of cis regulatory evolution in maize domestication. PLoS Genet 10:e1004745.; Swinnen et al., 2016Swinnen G, Goossens A and Pauwels L (2016) Lessons from domestication: targeting cis-regulatory elements for crop improvement. Trends Plant Sci 21:506-515.). Thus, regulatory sequences can be targeted by GE to fine-tune gene expression levels and tissue preference to create an array of subtle phenotypes (Figure 2) (Wittkopp and Kalay, 2012Wittkopp PJ and Kalay G (2012) Cis-regulatory elements: Molecular mechanisms and evolutionary processes underlying divergence. Nat Rev Genet 13:59-69.; Swinnen et al., 2016Swinnen G, Goossens A and Pauwels L (2016) Lessons from domestication: targeting cis-regulatory elements for crop improvement. Trends Plant Sci 21:506-515.; Ganguly et al., 2022Ganguly DR, Hickey LT and Crisp PA (2022) Harnessing genetic variation at regulatory regions to fine-tune traits for climate-resilient crops. Mol Plant 15:222-224.).

Figure 2 -
Using CRISPR/Cas to edit the promoter region of target genes. A. Cis -regulatory elements (CREs) present upstream of a given gene may act as enhancers (purple, red, and orange boxes) or repressors (green and blue boxes), modulating gene expression. B. Multiplex genome editing approach: multiple sgRNAs targeting different CREs may result in stochastic mutations in the promoter region, resulting in alleles with different expression patterns/levels. This method may ultimately lead to lines with a phenotypic gradient.

Other non-coding regions can also be targeted for regulating expression, such as introns and upstream open reading frames. Nevertheless, non-coding region editing is far from trivial. Given the complexity of the CRE landscape and mode of action, phenotypic effects resulting from mutations within promoter regions are hardly predictable (Rodríguez-Leal et al., 2017Rodríguez-Leal D, Lemmon ZH, Man J, Bartlett ME and Lippman ZB (2017) Engineering quantitative trait variation for crop improvement by genome editing. Cell 171:470-480.e8.). Compensation effects such as those from enhancers and repressors and the distance between different CREs contribute to this lack of predictability. Another layer of complexity comes from the chromatin conformation in the promoter region, with epigenetic modifications and chromatin accessibility also controlling gene expression levels (Rodgers-Melnick et al., 2016Rodgers-Melnick E, Vera DL, Bass HW and Buckler ES (2016) Open chromatin reveals the functional maize genome. Proc Natl Acad Sci U S A 113:3177-3184.; Schmitz et al., 2022Schmitz RJ, Grotewold E and Stam M (2022) Cis-regulatory sequences in plants: Their importance, discovery, and future challenges. Plant Cell 34:718-741.).

Even though editing promoters being challenging, it holds great potential for plant breeding by facilitating the approval of new commercial varieties. Promoter editing allows manipulating a small number of nucleotides in non-coding regions without exogenous DNA in the final product; therefore, it may overcome the lengthy and costly regulatory processes and social rejection hurdles that come with transgenic events (Lassoued et al., 2019Lassoued R, Macall DM, Hesseln H, Phillips PWB and Smyth SJ (2019) Benefits of genome-edited crops: expert opinion. Transgenic Res 28:247-256.). Furthermore, promoter GE may allow manipulating quantitative traits, adding diversity to plant breeding. As an example, tomato lines with fruits presenting a gradient in the number of locules were developed through MGE targeting the promoter region of the SlCLV3 gene (Rodríguez-Leal et al., 2017Rodríguez-Leal D, Lemmon ZH, Man J, Bartlett ME and Lippman ZB (2017) Engineering quantitative trait variation for crop improvement by genome editing. Cell 171:470-480.e8.). Similarly, an allelic series of the maize ZmCLE7 gene was generated by promoter GE, resulting in lines presenting variability for inflorescence meristem size, ultimately leading to enhanced grain-yield-related traits (Liu et al., 2021Liu L, Gallagher J, Arevalo ED, Chen R, Skopelitis T, Wu Q, Bartlett M and Jackson D (2021) Enhancing grain-yield-related traits by CRISPR-Cas9 promoter editing of maize CLE genes. Nat Plants 7:287-294.). Grain yield per ear was also recently improved by both knocking out and promoter editing of the ZmACO2 gene. In this case, the reduced expression / loss-of-function of ZmACO2 resulted in increased yield in different genetic backgrounds, including hybrids (Ning et al., 2021Ning Q, Jian Y, Du Y, Li Y, Shen X, Jia H, Zhao R, Zhan J, Yang F, Jackson D et al. (2021) An ethylene biosynthesis enzyme controls quantitative variation in maize ear length and kernel yield. Nat Commun 12:5832.).

In a noteworthy example addressing a highly complex trait, maize plants with increased drought tolerance were generated by either introducing or swapping the promoter region of the ARGOS8 gene with the endogenous and stronger promoter from GOS2, which contains drought-responsive cis-elements (Shi et al., 2017Shi J, Gao H, Wang H, Lafitte HR, Archibald RL, Yang M, Hakimi SM, Mo H and Habben JE (2017) ARGOS8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnol J 15:207-216.). ARGOS8 negatively regulates ethylene responses (Shi et al., 2016Shi J, Drummond BJ, Wang H, Archibald RL and Habben JE (2016) Maize and Arabidopsis ARGOS proteins interact with ethylene receptor signaling complex, supporting a regulatory role for ARGOS in ethylene signal transduction. Plant Physiol 171:2783-2797.); therefore, its overexpression promotes cell expansion and/or division, enhancing plant growth and mitigating yield loss under drought (Shi et al., 2017Shi J, Gao H, Wang H, Lafitte HR, Archibald RL, Yang M, Hakimi SM, Mo H and Habben JE (2017) ARGOS8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnol J 15:207-216.). Importantly, similar increases in grain yield had already been reported in transgenic maize plants overexpressing ARGOS8 when submitted to drought conditions (Shi et al., 2015Shi J, Habben JE, Archibald RL, Drummond BJ, Chamberlin MA, Williams RW, Renee Lafitte H, Weers BP, Lafitte HR and Weers BP (2015) Overexpression of ARGOS genes modifies plant sensitivity to ethylene, leading to improved drought tolerance in both arabidopsis and maize. Plant Physiol 169:266-282.), underscoring the potential of promoter-editing strategies to mimic desirable phenotypic effects originated from a transgene-mediated expression of target genes.

The environment has a great impact on the expression patterns of genes underlying complex traits, such as enhanced yield and tolerance to various stresses, but the same genes and pathways they act upon are also often required for plant growth and development. Because classical biotechnological approaches for the manipulation of such genes rely on overexpression or loss-of-function knockouts, they frequently result in pleiotropic effects and undesirable tradeoffs (Huot et al., 2014Huot B, Yao J, Montgomery BL and He SY (2014) Growth-defense tradeoffs in plants: a balancing act to optimize fitness. Mol Plant 7:1267-1287.). Thus, fine-tuning their expression patterns via GE of regulatory sequences holds great potential for improving complex traits.

Base and prime editing

Other CRISPR/Cas strategies at the forefront of plant science that have great potential for breeding applications regard base and prime editors. Base editing relies on a fusion between a catalytically impaired Cas9 (nCas9 or dCas9) with a cytosine or adenosine deaminase. The modified Cas9 guides and anchors the fused protein to the target sequence driven by the sgRNA. Then, the fused protein can change the DNA sequence in a programmable manner without creating double-strand breaks (DSBs) (Komor et al., 2016Komor AC, Kim YB, Packer MS, Zuris JA and Liu DR (2016) Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420-424.; Gaudelli et al., 2017Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI and Liu DR (2017) Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551:464-471.; Anzalone et al., 2019Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A et al. (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149-157.).

Among the first tools for base editing are the adenine base editors (ABEs), which allow A-to-G (or T-to-C) transitions (Gaudelli et al., 2017Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI and Liu DR (2017) Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551:464-471.), and cytidine base editors (CBEs), which promote C-to-T (or G-to-A) conversions (Komor et al., 2016Komor AC, Kim YB, Packer MS, Zuris JA and Liu DR (2016) Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420-424.). New recently developed base editors allow C-to-G (CGBEs) and C-to-A conversions in mammalian and bacterial cells, respectively (Molla et al., 2021Molla KA, Sretenovic S, Bansal KC and Qi Y (2021) Precise plant genome editing using base editors and prime editors. Nat Plants 7:1166-1187.). While C-to-A conversions are still restricted to bacterial cells, CGBEs have been tested in rice, tomato, and poplar (Sretenovic et al., 2021Sretenovic S, Liu S, Li G, Cheng Y, Fan T, Xu Y, Zhou J, Zheng X, Coleman G, Zhang Y et al. (2021) Exploring C-to-G base editing in rice, tomato, and poplar. Front Genome Ed 3:756766.), but efficiencies are still very low.

Base editing has already been used in many crops, such as rice, maize, wheat, potato, tomato, watermelon, and cotton (Zong et al., 2017Zong Y, Wang Y, Li C, Zhang R, Chen K, Ran Y, Qiu JL, Wang D and Gao C (2017) Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat Biotechnol 35:438-440.; Li et al., 2018Li C, Zong Y, Wang Y, Jin S, Zhang D, Song Q, Zhang R and Gao C (2018) Expanded base editing in rice and wheat using a Cas9-adenosine deaminase fusion. Genome Biol 19:59.; Mishra et al., 2020Mishra R, Joshi RK and Zhao K (2020) Base editing in crops: current advances, limitations and future implications. Plant Biotechnol J 18:20-31.; Qin et al., 2020Qin L, Li J, Wang Q, Xu Z, Sun L, Alariqi M, Manghwar H, Wang G, Li B, Ding X et al. (2020) High‐efficient and precise base editing of C•G to T•A in the allotetraploid cotton (Gossypium hirsutum) genome using a modified CRISPR /Cas9 system. Plant Biotechnol J 18:45-56.). For maize, targeted conversion of C-to-T in ZmALS1 and ZmALS2 genes, with an efficiency of up to 14%, generated transgene-free edited plants harboring a homozygous mutation for ALS1 or double mutation for the two ALS genes, leading to herbicide-tolerant plants (Li et al., 2020Li Y, Zhu J, Wu H, Liu C, Huang C, Lan J, Zhao Y and Xie C (2020) Precise base editing of non-allelic acetolactate synthase genes confers sulfonylurea herbicide resistance in maize. Crop J 8:449-456.).

Further possible programmed and precise sequence alterations are possible with prime editing, which allows insertions (up to 44 bp), deletions (up to 90 bp), and single base alterations, including all 12 possible base-to-base conversions, without requiring DSBs or donor DNA templates (Anzalone et al., 2019Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A et al. (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149-157.; Zong et al., 2022Zong Y, Liu Y, Xue C, Li B, Li X, Wang Y, Li J, Liu G, Huang X, Cao X et al. (2022) An engineered prime editor with enhanced editing efficiency in plants. Nat Biotechnol 40:1394-1402.). The prime editing system is based on a Cas nickase fused to an engineered reverse transcriptase and programmed with a prime editing guide RNA (pegRNA). The pegRNA both specifies the intended cut site (primes with the target DNA) and acts as a template (encodes the desired edit) for precise editing at the target genomic locus (Anzalone et al., 2019Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A et al. (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149-157.; 2020Anzalone AV, Koblan LW and Liu DR (2020) Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 38:824-844.). A reverse transcriptase extends the target DNA sequence based on the pegRNA and this new strand competes with the original one to bind with the non-target DNA strand. If the edited strand anneals, a mismatch occurs. Since the non-target strand is nicked by nCas9, it is more likely that the cell will copy the newly edited strand to repair the damaged DNA (Marzec et al., 2020Marzec M, Brąszewska-Zalewska A and Hensel G (2020) Prime Editing: a new way for genome editing. Trends Cell Biol 30:257-259.).

Prime editing, first proposed by Anzalone et al. (2019Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A et al. (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149-157.) to edit the human genome, has already been successfully employed to edit plant genomes, although with very low efficiencies (Tang et al., 2020Tang X, Sretenovic S, Ren Q, Jia X, Li M, Fan T, Yin D, Xiang S, Guo Y, Liu L et al. (2020) Plant prime editors enable precise gene editing in rice cells. Mol Plant 13:667-670.; Xu R et al., 2020Xu R, Li J, Liu X, Shan T, Qin R and Wei P (2020) Development of plant prime-editing systems for precise genome editing. Plant Commun 1:100043.; Xu R et al., 2020Xu R, Li J, Liu X, Shan T, Qin R and Wei P (2020) Development of plant prime-editing systems for precise genome editing. Plant Commun 1:100043.; Lin et al., 2020Lin Q, Zong Y, Xue C, Wang S, Jin S, Zhu Z, Wang Y, Anzalone A V., Raguram A, Doman JL et al. (2020) Prime genome editing in rice and wheat. Nat Biotechnol 38:582-585.; 2021Lin Q, Jin S, Zong Y, Yu H, Zhu Z, Liu G, Kou L, Wang Y, Qiu J-L, Li J et al. (2021) High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat Biotechnol 39:923-927.). Improvements such as modifications of the reverse transcriptase functional domains, which lead to an average of 5.8-fold increase in editing efficiency compared to the original prime editor, have been recently reported (Zong et al., 2022Zong Y, Liu Y, Xue C, Li B, Li X, Wang Y, Li J, Liu G, Huang X, Cao X et al. (2022) An engineered prime editor with enhanced editing efficiency in plants. Nat Biotechnol 40:1394-1402.). An independent improvement was achieved in rice, maize, and human cells by fusing the reverse transcriptase to the Cas9 N-terminal instead of the C-terminal and multiple-nucleotide substitutions in the reverse transcriptase template to enhance prime editing efficiency (Xu et al., 2022Xu W, Yang Y, Yang B, Krueger CJ, Xiao Q, Zhao S, Zhang L, Kang G, Wang F, Yi H et al. (2022) A design optimized prime editor with expanded scope and capability in plants. Nat Plants 8:45-52.).

Base and prime editing are of special convenience for developing traits known to arise from point mutations. For example, prime editing was applied to generate sulfonylurea herbicide-resistant maize (Li et al., 2020Li Y, Zhu J, Wu H, Liu C, Huang C, Lan J, Zhao Y and Xie C (2020) Precise base editing of non-allelic acetolactate synthase genes confers sulfonylurea herbicide resistance in maize. Crop J 8:449-456.), tomato, potato (Veillet et al., 2019Veillet F, Perrot L, Chauvin L, Kermarrec MP, Guyon-Debast A, Chauvin JE, Nogué F and Mazier M (2019) Transgene-free genome editing in tomato and potato plants using Agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. Int J Mol Sci 20:402), and oilseed rape (Wu et al., 2020Wu J, Chen C, Xian G, Liu D, Lin L, Yin S, Sun Q, Fang Y, Zhang H and Wang Y (2020) Engineering herbicide-resistant oilseed rape by CRISPR/Cas9-mediated cytosine base-editing. Plant Biotechnol J 18:1857-1859.). Jiang et al. (2020Jiang Y-Y, Chai Y-P, Lu M-H, Han X-L, Lin Q, Zhang Y, Zhang Q, Zhou Y, Wang X-C, Gao C et al. (2020) Prime editing efficiently generates W542L and S621I double mutations in two ALS genes in maize. Genome Biol 21:257.) also generated double mutations inZmALS1andZmALS2 using prime editors in maize, with higher efficiency than previously reported. Moreover, base and prime editing can also be helpful to disrupt or even introduce regulatory sequences to alter gene expression (Molla et al., 2021Molla KA, Sretenovic S, Bansal KC and Qi Y (2021) Precise plant genome editing using base editors and prime editors. Nat Plants 7:1166-1187.). For example, in strawberry (Fragaria vesca), base editing of an upstream open reading frame (uORF) led to the development of lines showing a continuum in sugar content (Xing et al., 2020Xing S, Chen K, Zhu H, Zhang R, Zhang H, Li B and Gao C (2020) Fine-tuning sugar content in strawberry. Genome Biol 21:230.).

Although with less obvious applications to complex traits, these examples of base and prime editing highlight the importance of such technologies to develop new maize cultivars. While prime editing opens the possibility of inducing precise mutations in coding or regulatory sequences, major improvements in editing efficiency are still required before the technique can be largely applied to maize.

Limitations and strategies for the application of genome editing in maize breeding

Despite its great potential for plant breeding, CRISPR-based GE faces important obstacles that hinder its large-scale application to deploy new commercial maize lines. Given the high specificity of the CRISPR/Cas systems, sequencing of the target sites is required for each different genotype to be edited, ensuring the correct design of sgRNAs. Recently, this obstacle has been somehow alleviated with the publication of the genome of 26 maize inbred lines used as founders for the maize nested association mapping population (Hufford et al., 2021Hufford MB, Seetharam AS, Woodhouse MR, Chougule KM, Ou S, Liu J, Ricci WA, Guo T, Olson A, Qiu Y et al. (2021) De novo assembly, annotation, and comparative analysis of 26 diverse maize genomes. Science 373:655-662.). This resource may not only help sgRNA design but also hint at loci associated with relevant agronomic traits, representing potential targets for genome editing. However, it is still advisable to re-sequence target loci from the explant-donor lines, as even these genotypes can present some polymorphisms that could interfere with editing efficiency. Sequencing the target genotypes would also help in identifying potential off-targets.

One of the main bottlenecks for applying biotechnological tools in maize breeding is the recalcitrance of maize lines to Agrobacterium-mediated transformation. Although there are some maize lines amenable to this standard transformation method, such as Hi-II and B104, most of these lines are not suitable for proper field trials of GE events and/or for commercial application (Kausch et al., 2021aKausch AP, Nelson-Vasilchik K, Tilelli M and Hague JP (2021a) Maize tissue culture, transformation, and genome editing. In Vitro Cell Dev Biol Plant 57:653-671.; bKausch AP, Wang K, Kaeppler HF and Gordon-Kamm W (2021b) Maize transformation: history, progress, and perspectives. Mol Breeding 41:38.; Yassitepe et al., 2021Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.). In addition, even among these transformable maize lines, transformation efficiency with standard protocols is usually low. Taken together, the low efficiency of some GE techniques in plants (e.g., homology-directed repair and prime editing) and the recalcitrance of most maize genotypes to transformation hamper GE application for maize breeding. For instance, one of the most studied maize lines, B73, which is also the reference maize genome, is highly recalcitrant to standard transformation protocols (Andorf et al., 2019Andorf C, Beavis WD, Hufford M, Smith S, Suza WP, Wang K, Woodhouse M, Yu J and Lübberstedt T (2019) Technological advances in maize breeding: past, present and future. Theor Appl Genet 132:817-849.; Yassitepe et al., 2021Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.).

Cultivated maize plants are usually hybrids between parental elite inbred lines. These inbred lines are genetically homozygous and harbor alleles encoding important agronomic traits. Hybrids obtained by crossing distinct inbred lines perform better in the field due to a phenomenon known as heterosis (Li et al., 2021Li D, Zhou Z, Lu X, Jiang Y, Li G, Li J, Wang H, Chen S, Li X, Würschum T et al. (2021) Genetic dissection of hybrid performance and heterosis for yield-related traits in maize. Front Plant Sci 12:774478.). Thus, breeding programs routinely cross different inbred parents to produce new F1 hybrid combinations. Although very successful for classical plant breeding, cultivated hybrids represent an additional obstacle to applying GE in maize. For example, for a trait resulting from a gene knockout, both parental lines need to be edited, which is usually unfeasible owing to genotype recalcitrance to transformation. Furthermore, even though hybrids are widely used in crop breeding to capture heterosis, their phenotypic superiority obtained in F1 is lost in further crosses.

Maize transformation has been extensively reviewed from different perspectives (Kausch et al., 2021aKausch AP, Nelson-Vasilchik K, Tilelli M and Hague JP (2021a) Maize tissue culture, transformation, and genome editing. In Vitro Cell Dev Biol Plant 57:653-671.,bKausch AP, Wang K, Kaeppler HF and Gordon-Kamm W (2021b) Maize transformation: history, progress, and perspectives. Mol Breeding 41:38.; Yassitepe et al., 2021Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.). Several strategies to overcome the low transformation efficiency, genotype dependency, and time-consuming introgression of GE alleles have been reported. Two promising systems involve improved transformation protocols based on the ectopic expression of morphogenic genes, which help in the regeneration process, and methods for in trans genome editing in a transformation-free manner. In addition, breakthroughs in synthetic apomixis are promising for fixing hybrid vigor in GE lines.

Morphogenic regulators: groundbreaking tools toward “universal” transformation protocols

A promising strategy to overcome the recalcitrance of most maize genotypes to genetic transformation is the introduction of morphogenic regulators (MRs) in the construct designed for genetic transformation. The most frequent MRs used in maize are the transcription factors BABY BOOM (BBM) and WUSCHEL (WUS) (Lowe et al., 2016Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho M-J, Scelonge C, Lenderts B, Chamberlin M, Cushatt J et al. (2016) Morphogenic regulators BABY BOOM and WUSCHEL improve monocot transformation. Plant Cell 28:1998-2015.; Mookkan et al., 2017Mookkan M, Nelson-Vasilchik K, Hague J, Zhang ZJ and Kausch AP (2017) Selectable marker independent transformation of recalcitrant maize inbred B73 and sorghum P898012 mediated by morphogenic regulators BABY BOOM and WUSCHEL2. Plant Cell Rep 36:1477-1491.; Lowe et al., 2018Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T and Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54:240-252.; Barone et al., 2020Barone P, Wu E, Lenderts B, Anand A, Gordon-Kamm W, Svitashev S and Kumar S (2020) Efficient gene targeting in maize using inducible CRISPR-Cas9 and marker-free donor template. Mol Plant 13:1219-1227.; Aesaert et al., 2022Aesaert S, Impens L, Coussens G, van Lerberge E, Vanderhaeghen R, Desmet L, Vanhevel Y, Bossuyt S, Wambua AN, van Lijsebettens M et al. (2022) Optimized transformation and gene editing of the B104 public maize inbred by improved tissue culture and use of morphogenic regulators. Front Plant Sci 13:883847.). When co-expressed, BBM and WUS can induce somatic embryogenesis in several plant tissues, including the scutellum of immature zygotic embryos (currently the most efficient starting material for maize transformation). This method reduces the time required for tissue culture since it skips the callus-forming stage by directly inducing somatic embryogenesis (Lowe et al., 2016Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho M-J, Scelonge C, Lenderts B, Chamberlin M, Cushatt J et al. (2016) Morphogenic regulators BABY BOOM and WUSCHEL improve monocot transformation. Plant Cell 28:1998-2015.; Lowe et al., 2018Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T and Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54:240-252.). However, the most significant advantage of such a technique relies on its ability to induce somatic embryogenesis in genotypes otherwise recalcitrant to tissue regeneration (Lowe et al., 2016Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho M-J, Scelonge C, Lenderts B, Chamberlin M, Cushatt J et al. (2016) Morphogenic regulators BABY BOOM and WUSCHEL improve monocot transformation. Plant Cell 28:1998-2015., 2018Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T and Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54:240-252.; Masters et al., 2020Masters A, Kang M, McCaw M, Zobrist JD, Gordon-Kamm W, Jones T and Wang K (2020) Agrobacterium-mediated immature embryo transformation of recalcitrant maize inbred lines using morphogenic genes. J Vis Exp 2020:156.).

Because constitutive expression of MRs impairs normal plant development, MR expression must be confined to the embryogenesis induction phase. For this, MR expression cassettes are either excised from the T-DNA after somatic embryogenesis or only transiently expressed soon after transformation. Auto-excision is usually achieved by a recombination system such as the CRE/loxP (Lowe et al., 2016Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho M-J, Scelonge C, Lenderts B, Chamberlin M, Cushatt J et al. (2016) Morphogenic regulators BABY BOOM and WUSCHEL improve monocot transformation. Plant Cell 28:1998-2015.; Mookkan et al., 2017Mookkan M, Nelson-Vasilchik K, Hague J, Zhang ZJ and Kausch AP (2017) Selectable marker independent transformation of recalcitrant maize inbred B73 and sorghum P898012 mediated by morphogenic regulators BABY BOOM and WUSCHEL2. Plant Cell Rep 36:1477-1491.; Zhang et al., 2019Zhang Q, Zhang Y, Lu M-H, Chai Y-P, Jiang Y-Y, Zhou Y, Wang X-C and Chen Q-J (2019) A novel ternary vector system united with morphogenic genes enhances CRISPR/Cas delivery in maize. Plant Physiol 181:1441-1448.; Masters et al., 2020Masters A, Kang M, McCaw M, Zobrist JD, Gordon-Kamm W, Jones T and Wang K (2020) Agrobacterium-mediated immature embryo transformation of recalcitrant maize inbred lines using morphogenic genes. J Vis Exp 2020:156.; Aesaert et al., 2022Aesaert S, Impens L, Coussens G, van Lerberge E, Vanderhaeghen R, Desmet L, Vanhevel Y, Bossuyt S, Wambua AN, van Lijsebettens M et al. (2022) Optimized transformation and gene editing of the B104 public maize inbred by improved tissue culture and use of morphogenic regulators. Front Plant Sci 13:883847.), while other strategies rely on the delivery of MRs in a separate plasmid designed not to be integrated into the genome (Svitashev et al., 2016Svitashev S, Schwartz C, Lenderts B, Young JK and Mark Cigan A (2016) Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat Commun 7:13274.; Hoerster et al., 2020Hoerster G, Wang N, Ryan L, Wu E, Anand A, McBride K, Lowe K, Jones T and Gordon-Kamm B (2020) Use of non-integrating Zm-Wus2 vectors to enhance maize transformation. In Vitro Cell Dev Biol Plant 56:265-279.). Choosing appropriate promoters for MRs also improved the quality of recovered transformed events. It can even eliminate the need to remove the MR cassette from the T-DNA (Lowe et al., 2018Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T and Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54:240-252.).

In their seminal work, Lowe et al. (2016Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho M-J, Scelonge C, Lenderts B, Chamberlin M, Cushatt J et al. (2016) Morphogenic regulators BABY BOOM and WUSCHEL improve monocot transformation. Plant Cell 28:1998-2015.) recovered transgenic quality events from four recalcitrant maize lines at a frequency of up to 13.7%. Similar results were obtained for the reference genotype B73, with a transformation frequency of approximately 15% (Mookkan et al., 2017Mookkan M, Nelson-Vasilchik K, Hague J, Zhang ZJ and Kausch AP (2017) Selectable marker independent transformation of recalcitrant maize inbred B73 and sorghum P898012 mediated by morphogenic regulators BABY BOOM and WUSCHEL2. Plant Cell Rep 36:1477-1491.). Moreover, at least 22 DuPont Pioneer’s inbred lines were responsive to the MR-based transformation protocol, indicating that it may indeed be genotype-independent (Lowe et al., 2018Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T and Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54:240-252.). More recently, the technique was applied to B104, a line commonly used for transformation in academic settings. By fine-tuning the media culture and including an MR expression cassette in the T-DNA, the transformation efficiency of B104 increased from 1% to 5% (Aesaert et al., 2022Aesaert S, Impens L, Coussens G, van Lerberge E, Vanderhaeghen R, Desmet L, Vanhevel Y, Bossuyt S, Wambua AN, van Lijsebettens M et al. (2022) Optimized transformation and gene editing of the B104 public maize inbred by improved tissue culture and use of morphogenic regulators. Front Plant Sci 13:883847.).

Other recent strategies use a different set of MRs. GROWTH REGULATING FACTOR 5 (GRF5) and a GRF4/GRF-INTERACTING FACTOR 1 (GIF1) fusion has been reported to improve regeneration efficiency in monocot and dicot plants (Debernardi et al., 2020Debernardi JM, Tricoli DM, Ercoli MF, Hayta S, Ronald P, Palatnik JF and Dubcovsky J (2020) A GRF-GIF chimeric protein improves the regeneration efficiency of transgenic plants. Nat Biotechnol 38:1274-1279.; Kong et al., 2020Kong J, Martin-Ortigosa S, Finer J, Orchard N, Gunadi A, Batts LA, Thakare D, Rush B, Schmitz O, Stuiver M et al. (2020) Overexpression of the transcription factor GROWTH-REGULATING factor5 improves transformation of dicot and monocot species. Front Plant Sci 11:572319.). These morphogenic genes are involved in organ development, which may represent an advantage over BBM/WUS constructs since their continued expression does not culminate in developmental penalties for the transformed plant. Overexpression of putative GRF5 maize orthologs increased the transformation efficiency of the A188 inbred line by approximately 3-fold (Kong et al., 2020Kong J, Martin-Ortigosa S, Finer J, Orchard N, Gunadi A, Batts LA, Thakare D, Rush B, Schmitz O, Stuiver M et al. (2020) Overexpression of the transcription factor GROWTH-REGULATING factor5 improves transformation of dicot and monocot species. Front Plant Sci 11:572319.), while the GRF4-GIF1 fusion not only increased the number of regenerated plants in rice, triticale, and wheat but also increased the number of wheat genotypes amenable to transformation (Debernardi et al., 2020Debernardi JM, Tricoli DM, Ercoli MF, Hayta S, Ronald P, Palatnik JF and Dubcovsky J (2020) A GRF-GIF chimeric protein improves the regeneration efficiency of transgenic plants. Nat Biotechnol 38:1274-1279.).

“Transformation-free” methods for genome editing

In the case of in trans genome editing, methods including the desired-target mutator (DTM) (Li et al., 2017Li C, Liu C, Qi X, Wu Y, Fei X, Mao L, Cheng B, Li X and Xie C (2017) RNA-guided Cas9 as an in vivo desired-target mutator in maize. Plant Biotechnol J 15:1566-1576.), haploid induction (HI) editing technology (HI-Edit) (Kelliher et al., 2019Kelliher T, Starr D, Su X, Tang G, Chen Z, Carter J, Wittich PE, Dong S, Green J, Burch E et al. (2019) One-step genome editing of elite crop germplasm during haploid induction. Nat Biotechnol 37:287-292.), and haploid-inducer mediated genome editing system (IMGE) (Wang B et al., 2019Wang C, Liu Q, Shen Y, Hua Y, Wang J, Lin J, Wu M, Sun T, Cheng Z, Mercier R et al. (2019) Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat Biotechnol 37:283-286.) may accelerate the development of edited maize varieties. These technologies allow delivering the CRISPR/Cas machinery directly to transformation-recalcitrant lines by crossing elite inbred lines with a stably transformed line harboring a CRISPR/Cas construct, allowing the continued activity of CRISPR/Cas to generate new mutated alleles. Thus, although not completely avoiding genetic transformation, DTM and HI-Edit/IMGE limits the transformation step to amenable genotypes (recently reviewed by Gao 2021Gao C (2021) Genome engineering for crop improvement and future agriculture. Cell 184:1621-1635.; Gu et al., 2021Gu X, Liu L and Zhang H (2021) Transgene-free genome editing in plants. Front Genome Ed 3:805317.; Yassitepe et al., 2021Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.; Impens et al., 2022Impens L, Jacobs TB, Nelissen H, Inzé D and Pauwels L (2022) Mini-Review: transgenerational CRISPR/Cas9 gene editing in plants. Front Genome Ed 4:825042.).

DTM was first used to generate newly edited alleles of the LIGULELESS1 (LG1) gene in maize (Li et al., 2017Li C, Liu C, Qi X, Wu Y, Fei X, Mao L, Cheng B, Li X and Xie C (2017) RNA-guided Cas9 as an in vivo desired-target mutator in maize. Plant Biotechnol J 15:1566-1576.). Crossing T1 plants harboring CRISPR machinery with six different maize lines resulted in over 20% mutated alleles in the progeny. As a breeding scheme, this method requires a series of backcrosses with marker-assisted selection to integrate the new alleles into an elite line for hybrid production. However, this approach avoids the linkage drag effect and uses a smaller population size to recover the recurrent genome, increasing the precision and time to introduce a new allele into elite lines (Li et al., 2017Li C, Liu C, Qi X, Wu Y, Fei X, Mao L, Cheng B, Li X and Xie C (2017) RNA-guided Cas9 as an in vivo desired-target mutator in maize. Plant Biotechnol J 15:1566-1576.). DTM was also applied to induce knockout mutations ofZmWX, resulting in hybrids with high endosperm amylopectin content (Qi et al., 2020Qi X, Wu H, Jiang H, Zhu J, Huang C, Zhang X, Liu C and Cheng B (2020) Conversion of a normal maize hybrid into a waxy version using in vivo CRISPR/Cas9 targeted mutation activity. Crop J 8:440-448.). A series of mutated alleles were generated in both hybrid parent lines. After two backcrosses, lines from both parents showed more than 87% of the genome recovered and ~ 20% more amylopectin content than their wild-type counterparts (Qi et al., 2020Qi X, Wu H, Jiang H, Zhu J, Huang C, Zhang X, Liu C and Cheng B (2020) Conversion of a normal maize hybrid into a waxy version using in vivo CRISPR/Cas9 targeted mutation activity. Crop J 8:440-448.).

Although the DTM strategy was an advance on genome editing approaches for maize breeding, especially by avoiding the linkage drag effect, it still requires a series of backcrosses to integrate the newly edited allele on an elite line. An advance in the in trans genome editing strategy was reported a couple of years later by two research groups, integrating genome editing with haploid induction (Hi-Edit) (Kelliher et al., 2019Kelliher T, Starr D, Su X, Tang G, Chen Z, Carter J, Wittich PE, Dong S, Green J, Burch E et al. (2019) One-step genome editing of elite crop germplasm during haploid induction. Nat Biotechnol 37:287-292.) and the haploid-inducer mediated genome editing system (IMGE) (Wang B et al., 2019Wang C, Liu Q, Shen Y, Hua Y, Wang J, Lin J, Wu M, Sun T, Cheng Z, Mercier R et al. (2019) Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat Biotechnol 37:283-286.). The use of double haploid (DH) in maize breeding is a common practice in several private and public breeding programs by eliminating generations of self-pollination for inbred production (Chaikam et al., 2019Chaikam V, Molenaar W, Melchinger AE and Boddupalli PM (2019) Doubled haploid technology for line development in maize: technical advances and prospects. Theor Appl Genet 132:3227-3243.). In HI-Edit/IMGE methods, the CRISPR/Cas machinery is introduced into a haploid inducer line that is used to pollinate maternal elite lines. In the haploid progeny, the paternal genome carrying the CRISPR/Cas9 transgene is eliminated. However, in a fraction of these haploids, the maternal genome is edited in trans. The next step is to treat the haploid progeny with a chromosome-doubling agent to produce DH lines. Then, these lines are screened for the presence of new edits, which are immediately monoallelic (homozygous) (Figure 3). For instance, genome-edited knockouts for the ZmLG1 and UB2 genes were generated and crossed with a haploid inductor line, resulting in edited haploid progenies at 3-4% efficiency (Wang B et al., 2019Wang B, Zhu L, Zhao B, Zhao Y, Xie Y, Zheng Z, Li Y, Sun J and Wang H (2019) Development of a haploid-inducer mediated genome editing system for accelerating maize breeding. Mol Plant 12:597-602.). In another example, a haploid inductor edited line for the MTL pollen-specific phospholipase gene was generated and used to pollinate different maize lines, successfully generating GE double haploids (Kelliher et al., 2019Kelliher T, Starr D, Su X, Tang G, Chen Z, Carter J, Wittich PE, Dong S, Green J, Burch E et al. (2019) One-step genome editing of elite crop germplasm during haploid induction. Nat Biotechnol 37:287-292.).

Figure 3 -
In trans genome editing in maize. First, a haploid-inducer (HI) line (amenable to transformation) is equipped with the CRISPR/Cas machinery targeting a specific locus (A). Next, HI pollen is used to pollinate plants from a non-transformable genotype (B). After fertilization, the CRISPR/Cas machinery encoded by the male parental genome edits the female genome (C). The male genome is degraded, resulting in a haploid embryo containing only the female genome (D). Chromosome doubling is achieved by applying chemical agents, resulting in a non-transgenic double haploid plant harboring the edited female genome (E).

In addition to being a transformation-free method, the Hi-Edit/IMGE approach generates transgene-free edited plants, an important aspect for GM-unfriendly markets. Further developments in the Hi-Edit/IMGE methods have been published, mainly to improve the haploid induction rate (Kelliher et al., 2017Kelliher T, Starr D, Richbourg L, Chintamanani S, Delzer B, Nuccio ML, Green J, Chen Z, McCuiston J, Wang W et al. (2017) MATRILINEAL, a sperm-specific phospholipase, triggers maize haploid induction. Nature 542:105-109., 2019Kelliher T, Starr D, Su X, Tang G, Chen Z, Carter J, Wittich PE, Dong S, Green J, Burch E et al. (2019) One-step genome editing of elite crop germplasm during haploid induction. Nat Biotechnol 37:287-292.; Zhong et al., 2019Zhong Y, Liu C, Qi X, Jiao Y, Wang D, Wang Y, Liu Z, Chen C, Chen B, Tian X et al. (2019) Mutation of ZmDMP enhances haploid induction in maize. Nat Plants 5:575-580.), identify seeds with haploid embryos by modifying visual traits such as anthocyanin biosynthesis (Chaikam et al., 2019Chaikam V, Molenaar W, Melchinger AE and Boddupalli PM (2019) Doubled haploid technology for line development in maize: technical advances and prospects. Theor Appl Genet 132:3227-3243.) or integrate visible transgenic markers into the inducer lines (Yu and Birchler 2016Yu W and Birchler JA (2016) A green fluorescent protein-engineered haploid inducer line facilitates haploid mutant screens and doubled haploid breeding in maize. Mol Breed 36:1-12.; Yan et al., 2021Yan Y, Zhu J, Qi X, Cheng B, Liu C and Xie C (2021) Establishment of an efficient seed fluorescence reporter‐assisted CRISPR/Cas9 gene editing in maize. J Integr Plant Biol jipb.13086.; Xu et al., 2021Xu J, Yin Y, Jian L, Han B, Chen Z, Yan J and Liu X (2021) Seeing is believing: a visualization toolbox to enhance selection efficiency in maize genome editing. Plant Biotechnol J 19:872-874.). Thus, in trans genome editing methods can precisely introduce desired traits into the genome of elite lines, overcoming the time-consuming traditional introgression efforts. As a result, they can also increase allele variability, which is potentially beneficial for future breeding demands.

Synthetic apomixis and its potential role for maize GE

Clonally propagating maize hybrid F1 seeds could be used to maintain the heterotic progeny resulting from inbred crosses, facilitating hybrid seed production. This purpose can be achieved by engineering apomixis, an asexual reproductive pathway that gives rise to offspring identical to the maternal plant (Wang C et al., 2019Wang C, Liu Q, Shen Y, Hua Y, Wang J, Lin J, Wu M, Sun T, Cheng Z, Mercier R et al. (2019) Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat Biotechnol 37:283-286.). Apomixis-based systems can be developed to induce the formation of unreduced gametes in the ovule and a two-step approach based on converting meiosis into mitosis, followed by eliminating the paternal genome has been tested in some plant species. For instance, a Mitosis instead of Meiosis (MiMe) technology was developed in rice by stacking mutations in the meiotic genes REC8, PAIR1, and OSD1 (Mieulet et al., 2016Mieulet D, Jolivet S, Rivard M, Cromer L, Vernet A, Mayonove P, Pereira L, Droc G, Courtois B, Guiderdoni E et al. (2016) Turning rice meiosis into mitosis. Cell Res 26:1242-1254.). More recently, F1 heterozygosity was fixed in rice by recreating the MiMe genotype via CRISPR/Cas9. Such genotype produces diploid gametes and tetraploid seeds (Wang C et al., 2019Wang C, Liu Q, Shen Y, Hua Y, Wang J, Lin J, Wu M, Sun T, Cheng Z, Mercier R et al. (2019) Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat Biotechnol 37:283-286.). Further mutation of the haploid induction MATRILINEAL (MTL) gene led to paternal genome elimination. The MiMe rice plants without the paternal genome produce self-fertilized F1 hybrids and clonal seeds with the same ploidy and heterozygous genotype. A similar result was obtained by combining the MiMe triple mutant with ectopic expression of the BBM1 transcription factor in the egg cell, which can trigger parthenogenesis. This approach resulted in clonal progeny that retained the asexual-propagation capability for multiple generations (Khanday et al., 2019Khanday I, Skinner D, Yang B, Mercier R and Sundaresan V (2019) A male-expressed rice embryogenic trigger redirected for asexual propagation through seeds. Nature 565:91-95.).

Despite the recent advances in synthetic apomixis in monocots such as rice, the process is still poorly explored in maize, in which efforts are more focused in the development of haploid induction. Apomixis-based genome editing strategies can be exploited to directly edit hybrids, fix hybrid vigor and facilitate clonal propagation of GE lines, although they need to be further optimized to be vastly applied in hybrid seed production in various crops (recently reviewed by Ozias-Akins and Conner 2020Ozias-Akins P and Conner JA (2020) Clonal reproduction through seeds in sight for crops. Trends Genet 36:215-226.; Chen et al., 2021Chen G, Zhou Y, Kishchenko O, Stepanenko A, Jatayev S, Zhang D and Borisjuk N (2021) Gene editing to facilitate hybrid crop production. Biotechnol Adv 46:107676.; Yin et al., 2022Yin PP, Tang LP, Zhang XS and Su YH (2022) Options for engineering apomixis in plants. Front Plant Sci 13:864987.).

Concluding Remarks

The development of new maize cultivars incorporating complex quantitative traits modified by biotechnological approaches can be a reality in the near future, considering the recent advances in genome editing. Although biotech maize resistant to insects and herbicides has dramatically impacted maize production worldwide, the improvement of complex quantitative traits such as drought and heat tolerance, nutrient acquisition and use efficiency, and yield are imperative to meet the ever-growing demand for maize-derived products. Currently, maize breeding programs have a wide range of unprecedented available tools to help speed up and accurately develop new cultivars. These tools include high-throughput phenotyping, advanced statistics and computational methods, crop models, new genome sequencing and predictions, and rising genome editing approaches. Here, we presented and discussed some basic concepts and emerging genome editing tools for maize and other crops. There are still critical challenges and questions to be addressed before the massive application of genome editing tools to maize breeding is attained. However, because genome-edited cultivars can be already accepted as non-GM in countries that produce nearly 80% of the global crops and acceptance has been growing in many other countries (Jenkins et al., 2021Jenkins D, Dobert R, Atanassova A and Pavely C (2021) Impacts of the regulatory environment for gene editing on delivering beneficial products. In Vitro Cell Dev Biol Plant 57:609-626.), it is expected that this technology will be a focus of intense development efforts and thus help democratize agricultural biotechnology in the benefit of sustainable food production for the global society.

Acknowledgements

We are grateful to the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) for supporting this research under the project “The Genomics for Climate Change Research Center (GCCRC)”, grant 2016/23218-0. This study was partly funded by Empresa Brasileira de Pesquisa Agropecuária (EMBRAPA) and Universidade Estadual de Campinas (UNICAMP). AK and VCHS received FAPESP postdoctoral fellowships (2021/08486-6 and 2018/06442-9, respectively). JHL received a postdoctoral fellowship from EMBRAPA and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) (381669/2019-0). IRG is recipient of CNPq productivity fellowship (308896/2020-3).

References

  • Aesaert S, Impens L, Coussens G, van Lerberge E, Vanderhaeghen R, Desmet L, Vanhevel Y, Bossuyt S, Wambua AN, van Lijsebettens M et al (2022) Optimized transformation and gene editing of the B104 public maize inbred by improved tissue culture and use of morphogenic regulators. Front Plant Sci 13:883847.
  • Andorf C, Beavis WD, Hufford M, Smith S, Suza WP, Wang K, Woodhouse M, Yu J and Lübberstedt T (2019) Technological advances in maize breeding: past, present and future. Theor Appl Genet 132:817-849.
  • Anzalone AV, Koblan LW and Liu DR (2020) Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 38:824-844.
  • Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A et al (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149-157.
  • Barone P, Wu E, Lenderts B, Anand A, Gordon-Kamm W, Svitashev S and Kumar S (2020) Efficient gene targeting in maize using inducible CRISPR-Cas9 and marker-free donor template. Mol Plant 13:1219-1227.
  • Boyer JS, Schlegel A, Delmer D, Porter D, Warner D, Gruis F, Gaffney J, Habben J, Schussler J, Shanahan J et al (2013) The U.S. drought of 2012 in perspective: a call to action. Glob Food Sec 2:139-143.
  • Brás TA, Seixas J, Carvalhais N and Jagermeyr J (2021) Severity of drought and heatwave crop losses tripled over the last five decades in Europe. Environ Res Lett 16:065012.
  • Chaikam V, Molenaar W, Melchinger AE and Boddupalli PM (2019) Doubled haploid technology for line development in maize: technical advances and prospects. Theor Appl Genet 132:3227-3243.
  • Chen G, Zhou Y, Kishchenko O, Stepanenko A, Jatayev S, Zhang D and Borisjuk N (2021) Gene editing to facilitate hybrid crop production. Biotechnol Adv 46:107676.
  • Chen K, Wang Y, Zhang R, Zhang H and Gao C (2019) CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu Rev Plant Biol 70:667-697.
  • Daryanto S, Wang L and Jacinthe PA (2016) Global synthesis of drought effects on maize and wheat production. PLoS One 11:e0156362.
  • Debernardi JM, Tricoli DM, Ercoli MF, Hayta S, Ronald P, Palatnik JF and Dubcovsky J (2020) A GRF-GIF chimeric protein improves the regeneration efficiency of transgenic plants. Nat Biotechnol 38:1274-1279.
  • Ganguly DR, Hickey LT and Crisp PA (2022) Harnessing genetic variation at regulatory regions to fine-tune traits for climate-resilient crops. Mol Plant 15:222-224.
  • Gao C (2021) Genome engineering for crop improvement and future agriculture. Cell 184:1621-1635.
  • Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI and Liu DR (2017) Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551:464-471.
  • Gong C, Huang S, Song R and Qi W (2021) Comparative study between the CRISPR/Cpf1 (Cas12a) and CRISPR/Cas9 systems for multiplex gene editing in maize. Agriculture 11:429.
  • Gornall J, Betts R, Burke E, Clark R, Camp J, Willett K and Wiltshire A (2010) Implications of climate change for agricultural productivity in the early twenty-first century. Philos Trans R Soc Lond B Biol Sci 365:2973-2989.
  • Gu X, Liu L and Zhang H (2021) Transgene-free genome editing in plants. Front Genome Ed 3:805317.
  • Hoerster G, Wang N, Ryan L, Wu E, Anand A, McBride K, Lowe K, Jones T and Gordon-Kamm B (2020) Use of non-integrating Zm-Wus2 vectors to enhance maize transformation. In Vitro Cell Dev Biol Plant 56:265-279.
  • Hufford MB, Seetharam AS, Woodhouse MR, Chougule KM, Ou S, Liu J, Ricci WA, Guo T, Olson A, Qiu Y et al (2021) De novo assembly, annotation, and comparative analysis of 26 diverse maize genomes. Science 373:655-662.
  • Huot B, Yao J, Montgomery BL and He SY (2014) Growth-defense tradeoffs in plants: a balancing act to optimize fitness. Mol Plant 7:1267-1287.
  • Hütsch BW and Schubert S (2018) Maize harvest index and water use efficiency can be improved by inhibition of gibberellin biosynthesis. J Agron Crop Sci 204:209-218.
  • Impens L, Jacobs TB, Nelissen H, Inzé D and Pauwels L (2022) Mini-Review: transgenerational CRISPR/Cas9 gene editing in plants. Front Genome Ed 4:825042.
  • Jenkins D, Dobert R, Atanassova A and Pavely C (2021) Impacts of the regulatory environment for gene editing on delivering beneficial products. In Vitro Cell Dev Biol Plant 57:609-626.
  • Jiang Y-Y, Chai Y-P, Lu M-H, Han X-L, Lin Q, Zhang Y, Zhang Q, Zhou Y, Wang X-C, Gao C et al (2020) Prime editing efficiently generates W542L and S621I double mutations in two ALS genes in maize. Genome Biol 21:257.
  • Kausch AP, Nelson-Vasilchik K, Tilelli M and Hague JP (2021a) Maize tissue culture, transformation, and genome editing. In Vitro Cell Dev Biol Plant 57:653-671.
  • Kausch AP, Wang K, Kaeppler HF and Gordon-Kamm W (2021b) Maize transformation: history, progress, and perspectives. Mol Breeding 41:38.
  • Kelliher T, Starr D, Richbourg L, Chintamanani S, Delzer B, Nuccio ML, Green J, Chen Z, McCuiston J, Wang W et al (2017) MATRILINEAL, a sperm-specific phospholipase, triggers maize haploid induction. Nature 542:105-109.
  • Kelliher T, Starr D, Su X, Tang G, Chen Z, Carter J, Wittich PE, Dong S, Green J, Burch E et al (2019) One-step genome editing of elite crop germplasm during haploid induction. Nat Biotechnol 37:287-292.
  • Khanday I, Skinner D, Yang B, Mercier R and Sundaresan V (2019) A male-expressed rice embryogenic trigger redirected for asexual propagation through seeds. Nature 565:91-95.
  • Komor AC, Kim YB, Packer MS, Zuris JA and Liu DR (2016) Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420-424.
  • Kong J, Martin-Ortigosa S, Finer J, Orchard N, Gunadi A, Batts LA, Thakare D, Rush B, Schmitz O, Stuiver M et al (2020) Overexpression of the transcription factor GROWTH-REGULATING factor5 improves transformation of dicot and monocot species. Front Plant Sci 11:572319.
  • Lassoued R, Macall DM, Hesseln H, Phillips PWB and Smyth SJ (2019) Benefits of genome-edited crops: expert opinion. Transgenic Res 28:247-256.
  • Lemmon ZH, Bukowski R, Sun Q and Doebley JF (2014) The role of cis regulatory evolution in maize domestication. PLoS Genet 10:e1004745.
  • Li C, Liu C, Qi X, Wu Y, Fei X, Mao L, Cheng B, Li X and Xie C (2017) RNA-guided Cas9 as an in vivo desired-target mutator in maize. Plant Biotechnol J 15:1566-1576.
  • Li C, Zong Y, Wang Y, Jin S, Zhang D, Song Q, Zhang R and Gao C (2018) Expanded base editing in rice and wheat using a Cas9-adenosine deaminase fusion. Genome Biol 19:59.
  • Li D, Zhou Z, Lu X, Jiang Y, Li G, Li J, Wang H, Chen S, Li X, Würschum T et al (2021) Genetic dissection of hybrid performance and heterosis for yield-related traits in maize. Front Plant Sci 12:774478.
  • Li Y, Zhu J, Wu H, Liu C, Huang C, Lan J, Zhao Y and Xie C (2020) Precise base editing of non-allelic acetolactate synthase genes confers sulfonylurea herbicide resistance in maize. Crop J 8:449-456.
  • Liang Z, Chen K, Li T, Zhang Y, Wang Y, Zhao Q, Liu J, Zhang H, Liu C, Ran Y et al (2017) Efficient DNA-free genome editing of bread wheat using CRISPR/Cas9 ribonucleoprotein complexes. Nat Commun 8:14261.
  • Lin Q, Jin S, Zong Y, Yu H, Zhu Z, Liu G, Kou L, Wang Y, Qiu J-L, Li J et al (2021) High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat Biotechnol 39:923-927.
  • Lin Q, Zong Y, Xue C, Wang S, Jin S, Zhu Z, Wang Y, Anzalone A V., Raguram A, Doman JL et al (2020) Prime genome editing in rice and wheat. Nat Biotechnol 38:582-585.
  • Liu L, Gallagher J, Arevalo ED, Chen R, Skopelitis T, Wu Q, Bartlett M and Jackson D (2021) Enhancing grain-yield-related traits by CRISPR-Cas9 promoter editing of maize CLE genes. Nat Plants 7:287-294.
  • Liu X, Zhang S, Jiang Y, Yan T, Fang C, Hou Q, Wu S, Xie K, An X and Wan X (2022) Use of CRISPR/Cas9-based gene editing to simultaneously mutate multiple homologous genes required for pollen development and male fertility in maize. Cells 11:439.
  • Lobell DB, Schlenker W and Costa-Roberts J (2011) Climate trends and global crop production since 1980. Science 333:616-620.
  • Lorenzo CD, Debray K, Herwegh D, Develtere W, Impens L, Schaumont D, Vandeputte W, Aesaert S, Coussens G, Boe Y de et al (2022) BREEDIT: A novel multiplex genome editing strategy to improve complex quantitative traits in maize. Plant Cell 35:218-238.
  • Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T and Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54:240-252.
  • Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho M-J, Scelonge C, Lenderts B, Chamberlin M, Cushatt J et al (2016) Morphogenic regulators BABY BOOM and WUSCHEL improve monocot transformation. Plant Cell 28:1998-2015.
  • Marzec M, Brąszewska-Zalewska A and Hensel G (2020) Prime Editing: a new way for genome editing. Trends Cell Biol 30:257-259.
  • Masters A, Kang M, McCaw M, Zobrist JD, Gordon-Kamm W, Jones T and Wang K (2020) Agrobacterium-mediated immature embryo transformation of recalcitrant maize inbred lines using morphogenic genes. J Vis Exp 2020:156.
  • Mieulet D, Jolivet S, Rivard M, Cromer L, Vernet A, Mayonove P, Pereira L, Droc G, Courtois B, Guiderdoni E et al (2016) Turning rice meiosis into mitosis. Cell Res 26:1242-1254.
  • Minkenberg B, Wheatley M and Yang Y (2017) CRISPR/Cas9-enabled multiplex genome editing and its application. Prog Mol Biol Transl Sci 149:111-132.
  • Mishra R, Joshi RK and Zhao K (2020) Base editing in crops: current advances, limitations and future implications. Plant Biotechnol J 18:20-31.
  • Molla KA, Sretenovic S, Bansal KC and Qi Y (2021) Precise plant genome editing using base editors and prime editors. Nat Plants 7:1166-1187.
  • Mookkan M, Nelson-Vasilchik K, Hague J, Zhang ZJ and Kausch AP (2017) Selectable marker independent transformation of recalcitrant maize inbred B73 and sorghum P898012 mediated by morphogenic regulators BABY BOOM and WUSCHEL2. Plant Cell Rep 36:1477-1491.
  • Najera VA, Twyman RM, Christou P and Zhu C (2019) Applications of multiplex genome editing in higher plants. Curr Opin Biotechnol 59:93-102.
  • Ning Q, Jian Y, Du Y, Li Y, Shen X, Jia H, Zhao R, Zhan J, Yang F, Jackson D et al (2021) An ethylene biosynthesis enzyme controls quantitative variation in maize ear length and kernel yield. Nat Commun 12:5832.
  • Ozias-Akins P and Conner JA (2020) Clonal reproduction through seeds in sight for crops. Trends Genet 36:215-226.
  • Peng J, Richards DE, Hartley NM, Murphy GP, Devos KM, Flintham JE, Beales J, Fish LJ, Worland AJ, Pelica F et al (1999) “Green revolution” genes encode mutant gibberellin response modulators. Nature 400:256-261.
  • Qi W, Zhu T, Tian Z, Li C, Zhang W and Song R (2016) High-efficiency CRISPR/Cas9 multiplex gene editing using the glycine tRNA-processing system-based strategy in maize. BMC Biotechnol 16:58.
  • Qi X, Wu H, Jiang H, Zhu J, Huang C, Zhang X, Liu C and Cheng B (2020) Conversion of a normal maize hybrid into a waxy version using in vivo CRISPR/Cas9 targeted mutation activity. Crop J 8:440-448.
  • Qin L, Li J, Wang Q, Xu Z, Sun L, Alariqi M, Manghwar H, Wang G, Li B, Ding X et al (2020) High‐efficient and precise base editing of C•G to T•A in the allotetraploid cotton (Gossypium hirsutum) genome using a modified CRISPR /Cas9 system. Plant Biotechnol J 18:45-56.
  • Ray DK, Gerber JS, MacDonald GK and West PC (2015) Climate variation explains a third of global crop yield variability. Nat Commun 6:5989.
  • Rodgers-Melnick E, Vera DL, Bass HW and Buckler ES (2016) Open chromatin reveals the functional maize genome. Proc Natl Acad Sci U S A 113:3177-3184.
  • Rodríguez-Leal D, Lemmon ZH, Man J, Bartlett ME and Lippman ZB (2017) Engineering quantitative trait variation for crop improvement by genome editing. Cell 171:470-480.e8.
  • Schmidt SM, Belisle M and Frommer WB (2020) The evolving landscape around genome editing in agriculture. EMBO Rep 21:e50680.
  • Schmitz RJ, Grotewold E and Stam M (2022) Cis-regulatory sequences in plants: Their importance, discovery, and future challenges. Plant Cell 34:718-741.
  • Shi J, Drummond BJ, Wang H, Archibald RL and Habben JE (2016) Maize and Arabidopsis ARGOS proteins interact with ethylene receptor signaling complex, supporting a regulatory role for ARGOS in ethylene signal transduction. Plant Physiol 171:2783-2797.
  • Shi J, Gao H, Wang H, Lafitte HR, Archibald RL, Yang M, Hakimi SM, Mo H and Habben JE (2017) ARGOS8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnol J 15:207-216.
  • Shi J, Habben JE, Archibald RL, Drummond BJ, Chamberlin MA, Williams RW, Renee Lafitte H, Weers BP, Lafitte HR and Weers BP (2015) Overexpression of ARGOS genes modifies plant sensitivity to ethylene, leading to improved drought tolerance in both arabidopsis and maize. Plant Physiol 169:266-282.
  • Simmons CR, Lafitte HR, Reimann KS, Brugière N, Roesler K, Albertsen MC, Greene TW and Habben JE (2021) Successes and insights of an industry biotech program to enhance maize agronomic traits. Plant Sci 307:110899.
  • Sretenovic S, Liu S, Li G, Cheng Y, Fan T, Xu Y, Zhou J, Zheng X, Coleman G, Zhang Y et al (2021) Exploring C-to-G base editing in rice, tomato, and poplar. Front Genome Ed 3:756766.
  • Svitashev S, Schwartz C, Lenderts B, Young JK and Mark Cigan A (2016) Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat Commun 7:13274.
  • Swinnen G, Goossens A and Pauwels L (2016) Lessons from domestication: targeting cis-regulatory elements for crop improvement. Trends Plant Sci 21:506-515.
  • Tang X, Sretenovic S, Ren Q, Jia X, Li M, Fan T, Yin D, Xiang S, Guo Y, Liu L et al (2020) Plant prime editors enable precise gene editing in rice cells. Mol Plant 13:667-670.
  • Teixeira JEC, Weldekidan T, de Leon N, Flint-Garcia S, Holland JB, Lauter N, Murray SC, Xu W, Hessel DA, Kleintop AE et al (2015) Hallauer’s Tusón: A decade of selection for tropical-to-temperate phenological adaptation in maize. Heredity (Edinb) 114:229-240.
  • Veillet F, Perrot L, Chauvin L, Kermarrec MP, Guyon-Debast A, Chauvin JE, Nogué F and Mazier M (2019) Transgene-free genome editing in tomato and potato plants using Agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. Int J Mol Sci 20:402
  • Wang B, Zhu L, Zhao B, Zhao Y, Xie Y, Zheng Z, Li Y, Sun J and Wang H (2019) Development of a haploid-inducer mediated genome editing system for accelerating maize breeding. Mol Plant 12:597-602.
  • Wang C, Liu Q, Shen Y, Hua Y, Wang J, Lin J, Wu M, Sun T, Cheng Z, Mercier R et al (2019) Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat Biotechnol 37:283-286.
  • Whelan AI, Gutti P and Lema MA (2020) Gene editing regulation and innovation economics. Front Bioeng Biotechnol 8:303.
  • White RJ (2011) Transcription by RNA polymerase III: More complex than we thought. Nat Rev Genet 12:459-463.
  • Wittkopp PJ and Kalay G (2012) Cis-regulatory elements: Molecular mechanisms and evolutionary processes underlying divergence. Nat Rev Genet 13:59-69.
  • Woo JWW, Kim JJSJJ-S, Kwon SI Il, Corvalán C, Cho SWW, Kim H, Kim S-TS-GTG, Kim S-TS-GTG, Choe S and Kim JJSJJ-S (2015) DNA-free genome editing in plants with preassembled CRISPR-Cas9 ribonucleoproteins. Nat Biotechnol 33:1162-1164.
  • Woźniak E, Tyczewska A and Twardowski T (2021) A shift towards biotechnology: Social opinion in the EU. Trends Biotechnol 39:214-218.
  • Wu J, Chen C, Xian G, Liu D, Lin L, Yin S, Sun Q, Fang Y, Zhang H and Wang Y (2020) Engineering herbicide-resistant oilseed rape by CRISPR/Cas9-mediated cytosine base-editing. Plant Biotechnol J 18:1857-1859.
  • Xie K, Minkenberg B and Yang Y (2015) Boosting CRISPR/Cas9 multiplex editing capability with the endogenous tRNA-processing system. Proc Natl Acad Sci 112:3570-3575.
  • Xing S, Chen K, Zhu H, Zhang R, Zhang H, Li B and Gao C (2020) Fine-tuning sugar content in strawberry. Genome Biol 21:230.
  • Xu J, Yin Y, Jian L, Han B, Chen Z, Yan J and Liu X (2021) Seeing is believing: a visualization toolbox to enhance selection efficiency in maize genome editing. Plant Biotechnol J 19:872-874.
  • Xu R, Li J, Liu X, Shan T, Qin R and Wei P (2020) Development of plant prime-editing systems for precise genome editing. Plant Commun 1:100043.
  • Xu R, Yang Y, Qin R, Li H, Qiu C, Li L, Wei P and Yang J (2016) Rapid improvement of grain weight via highly efficient CRISPR/Cas9-mediated multiplex genome editing in rice. J Genet Genomics 43:529-532.
  • Xu W, Yang Y, Yang B, Krueger CJ, Xiao Q, Zhao S, Zhang L, Kang G, Wang F, Yi H et al (2022) A design optimized prime editor with expanded scope and capability in plants. Nat Plants 8:45-52.
  • Xu W, Zhang C, Yang Y, Zhao S, Kang G, He X, Song J and Yang J (2020) Versatile nucleotides substitution in plant using an improved prime editing system. Mol Plant 13:675-678.
  • Yan Y, Zhu J, Qi X, Cheng B, Liu C and Xie C (2021) Establishment of an efficient seed fluorescence reporter‐assisted CRISPR/Cas9 gene editing in maize. J Integr Plant Biol jipb.13086.
  • Yassitepe JE de CT, da Silva VCH, Hernandes-Lopes J, Dante RA, Gerhardt IR, Fernandes FR, da Silva PA, Vieira LR, Bonatti V and Arruda P (2021) Maize transformation: From plant material to the release of genetically modified and edited varieties. Front Plant Sci 12:766702.
  • Yin PP, Tang LP, Zhang XS and Su YH (2022) Options for engineering apomixis in plants. Front Plant Sci 13:864987.
  • Yu W and Birchler JA (2016) A green fluorescent protein-engineered haploid inducer line facilitates haploid mutant screens and doubled haploid breeding in maize. Mol Breed 36:1-12.
  • Zhang J, Zhang X, Chen R, Yang L, Fan K, Liu Y, Wang G, Ren Z and Liu Y (2020) Generation of transgene-free semidwarf maize plants by gene editing of GIBBERELLIN-OXIDASE20-3 using CRISPR/Cas9. Front Plant Sci 11:1048.
  • Zhang Q, Zhang Y, Lu M-H, Chai Y-P, Jiang Y-Y, Zhou Y, Wang X-C and Chen Q-J (2019) A novel ternary vector system united with morphogenic genes enhances CRISPR/Cas delivery in maize. Plant Physiol 181:1441-1448.
  • Zhang Y, Liang Z, Zong Y, Wang Y, Liu J, Chen K, Qiu JL and Gao C (2016) Efficient and transgene-free genome editing in wheat through transient expression of CRISPR/Cas9 DNA or RNA. Nat Commun 7:1-8.
  • Zhong Y, Liu C, Qi X, Jiao Y, Wang D, Wang Y, Liu Z, Chen C, Chen B, Tian X et al (2019) Mutation of ZmDMP enhances haploid induction in maize. Nat Plants 5:575-580.
  • Zong Y, Liu Y, Xue C, Li B, Li X, Wang Y, Li J, Liu G, Huang X, Cao X et al (2022) An engineered prime editor with enhanced editing efficiency in plants. Nat Biotechnol 40:1394-1402.
  • Zong Y, Wang Y, Li C, Zhang R, Chen K, Ran Y, Qiu JL, Wang D and Gao C (2017) Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat Biotechnol 35:438-440.

Internet Resources

  • CEPEA-ESALQ - Centro de Estudos Avançados em Economia Aplicada ESALQ (2021) Indicador do Milho Esalq/BM&FBovespa, CEPEA-ESALQ - Centro de Estudos Avançados em Economia Aplicada ESALQ (2021) Indicador do Milho Esalq/BM&FBovespa, https://www.cepea.esalq.usp.br/br/indicador/milho.aspx (accessed 12 Oct 2021).
    » https://www.cepea.esalq.usp.br/br/indicador/milho.aspx
  • CONAB - Companhia Nacional de Abastecimento (2021) Acompanhamento de safra brasileira, CONAB - Companhia Nacional de Abastecimento (2021) Acompanhamento de safra brasileira, https://www.conab.gov.br/info-agro/safras/graos (accessed 12 Oct 2021).
    » https://www.conab.gov.br/info-agro/safras/graos
  • INMET - Instituto Nacional de Meteorologia (2021) SPI - Índice de Precipitação Padronizada, INMET - Instituto Nacional de Meteorologia (2021) SPI - Índice de Precipitação Padronizada, https://portal.inmet.gov.br/servicos/spi-índice-de-precipitação-padronizada (accessed 12 Oct 2021).
    » https://portal.inmet.gov.br/servicos/spi-índice-de-precipitação-padronizada
  • IPCC - The Intergovernmental Panel on Climate Change (2022) Climate Change 2022: Impacts, adaptation, and vulnerability. Sixth Assessment Report, Sixth Assessment Report, https://www.ipcc.ch/report/ar6/wg2/ (accessed 12 Oct 2021).
    » https://www.ipcc.ch/report/ar6/wg2/
  • 2
    License information: This is an open-access article distributed under the terms of the Creative Commons Attribution License (type CC-BY), which permits unrestricted use, distribution and reproduction in any medium, provided the original article is properly cited.

Associate Editor:

Rogerio Margis

Publication Dates

  • Publication in this collection
    03 Mar 2023
  • Date of issue
    2023

History

  • Received
    10 July 2022
  • Accepted
    23 Dec 2022
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br