Next Article in Journal
Evaluation on the Performance of Airflow Distribution Device of Pneumatic Seeder for Rapeseed through CFD Simulations
Next Article in Special Issue
Co-Design and Experimentation of a Prototype of Agroecological Micro-Farm Meeting the Objectives Set by Climate-Smart Agriculture
Previous Article in Journal
Classification of Oil Palm Fresh Fruit Bunches Based on Their Maturity Using Thermal Imaging Technique
Previous Article in Special Issue
Multidimensional Typology of Mexican Farmers in the Context of Climate Change
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential of Novel Gene Editing-Based Approaches in Forages and Rumen Archaea for Reducing Livestock Methane Emissions

1
Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, AB T1J 4B1, Canada
2
Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
3
Embrapa Dairy Cattle, Juiz de Fora 36038-330, MG, Brazil
4
Embrapa Genetic Resources and Biotechnology, Brasília 70770-917, DF, Brazil
*
Authors to whom correspondence should be addressed.
Agriculture 2022, 12(11), 1780; https://doi.org/10.3390/agriculture12111780
Submission received: 9 September 2022 / Revised: 14 October 2022 / Accepted: 18 October 2022 / Published: 26 October 2022

Abstract

:
Rising emissions of anthropogenic greenhouse gases such as carbon dioxide (CO2), nitrous oxide (N2O) and methane (CH4) are a key driver of climate change, which is predicted to have myriad detrimental consequences in coming years if not kept in check. Given the potency of CH4 in terms of trapping heat in the atmosphere in the short term, as well as the fact that ruminant production currently contributes approximately 30% of anthropogenic emissions, there is an impetus to substantially decrease the generation of ruminant-derived CH4. While various strategies are being assessed in this context, a multi-faceted approach is likely required to achieve significant reductions. Feed supplementation is one strategy that has shown promise in this field by attenuating methanogenesis in rumen archaea; however, this can be costly and sometimes impractical. In this review, we examine and discuss the prospect of directly modulating forages and/or rumen archaea themselves in a manner that would reduce methanogenesis using CRISPR/Cas-mediated gene editing platforms. Such an approach could provide a valuable alternative to supplementation and has the potential to contribute to the sustainability of agriculture, as well as the mitigation of climate change, in the future.

1. Introduction

Anthropogenic greenhouse gas (GHG) emissions, which are mainly driven by energy production, industrial processes, transport, waste management and forestry/agriculture, have been rising dramatically in recent years [1]. These gases function to trap heat energy, thereby increasing global temperatures, and are thus the main drivers of climate change. Indeed, it has been predicted that temperatures could increase by up to 5.7 °C globally by the end of the 21st century compared to temperatures in the years 1850–1900 in a scenario of very high GHG emissions [2]. While the yields of many crop species will decline dramatically with even incremental temperature increases in certain regions, further detrimental impacts will be incurred through escalations in the intensity and frequency of heat waves, droughts, flooding and soil salinization, as well as the worsening of disease and pest outbreaks [3,4,5]. Furthermore, such climate change scenarios would also lead to a series of additional devastating effects, including increased risks of wildfires, shifts in biodiversity and the extinction of species [6,7]. As a result of this, moderating anthropogenic GHG emissions is becoming a fundamental priority globally, with 131 countries intending to, or having already adopted or announced plans for net zero emission targets in the coming decades [8].
Of all GHGs emitted due to anthropogenic causes, carbon dioxide (CO2), methane (CH4) and nitrous oxide (N2O) comprise the highest proportions, making up approximately 75%, 18% and 4% of the total volume, respectively [9]. Although the amount of CH4 emitted is substantially less than CO2 and it is a relatively short-lived pollutant, it has a global warming potential that is 34-times higher than that of CO2 on a per unit mass basis over a 100-year time scale [10]. While global CH4 emissions derive from multiple sources, approximately 30% of anthropogenic CH4 arises from ruminant production [11], primarily due to inefficiencies in the enteric fermentation of insoluble carbohydrates (mainly lignin, cellulose and hemicellulose) in the rumen [12]. To limit global warming to 1.5 °C, it has been suggested that global agricultural CH4 emissions need to be reduced by 24–47% by 2050 compared to 2010 [13], which is particularly challenging given the projection that the production of ruminant-derived meat needs to increase by 88% in that same time period to meet growing demand [14].
In the rumen, insoluble carbohydrates are broken down into soluble sugars, which then undergo anaerobic fermentation to yield volatile fatty acids, including acetate, propionate and butyrate. This fermentation process also leads to the generation of CO2 and molecular hydrogen (H2), which, at increased ruminal partial pressures, can inhibit microbial enzymes and thus has a detrimental effect on the rate of carbohydrate fermentation [15]. However, H2 levels are typically kept in check by methanogens, which are a group of archaea that naturally occur in the rumen and use H2 and CO2 as substrates to produce CH4 [16]. The resulting CH4 is released into the atmosphere through eructation and breathing [17], and the amount produced is typically directly proportional to dry matter intake [16]. While this conversion of H2 to CH4 is beneficial in terms of rumen fermentation, it has a deleterious effect on the environment in the context of climate change and also represents a considerable loss for ruminant livestock producers, since approximately 2–15% of energy ingested is used for this process rather than for meat and milk production [18,19].
Over recent years, various methods have been assessed for their ability to mitigate CH4 emissions from ruminant production systems, which would in turn reduce environmental impacts and economic losses for producers. For example, the utilization of diets with higher grain contents or concentrates have been found to lead to incremental reductions in CH4 emissions from ruminants [20,21], as has the use of controlled rotational grazing [22] and animal breeding [23]. In addition, the administration of bacteriophages [24] and bacteriocins [25,26], as well as anti-methanogen vaccines [27], has also been suggested as a possible option in this context; however, their use is likely challenging due to the diversity of ruminal methanogens. In addition, their effectiveness in terms of reducing CH4 emissions is still unclear, especially long-term in vivo [24].
Furthermore, a range of different diet supplements have shown promise with regard to decreasing CH4 emissions from ruminants, including lipids, synthetic halogenated compounds, algae, short-chain nitrocompounds, nitrates, ionophores such as monensin, plant secondary compounds and essential oils [28,29,30,31,32,33,34,35,36,37,38,39,40]. For example, supplementation with red macroalgae Asparagopsis spp. has been shown to reduce CH4 emissions by upwards of 95% in vitro [41,42] and up to approximately 25–98% in ruminants in vivo [40,43,44,45,46]. This occurs due to the fact that these macroalgae accumulate low-molecular-weight halogenated compounds (predominantly the brominated halomethane, bromoform) [41] that inhibit the methyl-coenzyme M reductase (MCR) enzyme, which catalyzes the rate-limiting and final step of the methanogenesis pathway, in rumen archaea [47]. Similarly, approximately 20–60% reductions in CH4 emissions have also been reported when feed was supplemented with 3-nitrooxypropanol, which also inhibits the final step in methanogenesis [17], without interfering with milk or meat production [35,36,48,49,50,51,52,53]. Although these approaches have shown various degrees of success, the use of additives tends to be costly [54] and at times impractical, with possible safety and environmental impacts, which limit their use [46,55,56,57]. As such, the direct genetic manipulation of forages/feed and/or the methanogens themselves to elicit these same effects could provide valuable alternative approaches.
In this review paper, we comprehensively examine and discuss the potential of CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein)-based gene editing platforms in plants and microbes as novel options for reducing CH4 emissions from ruminants. The development of these, and other, strategies could conceivably contribute to a reduction in the impact of milk and meat production on CH4 emissions, and thus climate change, in the future.

2. CRISPR/Cas Gene Editing

As a means of increasing the pace and precision of genetic improvements, and with the additional advantage of potentially mitigating concerns encompassing the use of transgenic organisms, highly specific gene editing platforms that allow for the provision of mutations at pre-defined genomic loci have been rapidly gaining popularity over recent years [58]. While several gene editing systems have been developed to date [59,60,61], CRISPR/Cas has become the tool of choice as a result of its low cost, superior flexibility, ease with which one can multiplex and overall simplicity. The CRISPR/Cas gene editing platform is derived from bacterial or archaeal adaptive immune systems that protect mainly against bacteriophages through the genomic integration of small viral genetic fragments, which are then transcribed into small RNAs that serve as recognition molecules. These small RNA molecules avert subsequent bacteriophage invasion by guiding Cas nucleases to homologous viral DNA or RNA, which leads to their cleavage [58]. In its simplest form as a gene editing tool, the platform requires the introduction of a class 2 Cas nuclease, which elicits a double-stranded break (DSB) in DNA at a specific pre-defined chromosomal locus, as well as a single guide RNA (sgRNA) that directs Cas to the target site within the host genome, into host cells. These sgRNAs typically comprise a chimeric RNA consisting of an approximately 20 nt customizable CRISPR RNA (crRNA) that is complementary to the selected target loci and serves as a guide for Cas, as well as a fixed trans-activating CRISPR RNA (tracrRNA), which is necessary for Cas recruitment. In virtually all cases, crRNAs must be designed to anneal immediately upstream of a protospacer adjacent motif (PAM) sequence, which is a necessity for cleavage by the Cas nuclease. In the case of Cas9 from Streptomyces pyogenes, which is the most commonly used Cas nuclease (class 2, type II) for gene editing, the PAM consists of 5′–NGG–3′ [62], or less often, 5′–NAG–3′ [63], with the DSB generally occurring approximately 3 nt upstream [62].
The DSBs incurred by the Cas nuclease are then repaired by the host’s inherent DNA repair mechanisms, including either non-homologous end joining (NHEJ) or homology-directed repair (HDR; Figure 1). In the case of NHEJ-mediated gene editing, which is error-prone and the predominant DSB repair pathway in the somatic cells of higher plants [64], small insertions or deletions (indels) are typically incurred in the target gene that can disrupt its function. Since these mutations occur through the host’s own DSB repair mechanisms, in many cases, they can be indistinguishable from those transpiring spontaneously or through conventional breeding approaches [58]. Conversely, HDR-mediated gene editing, which is highly accurate and a major DSB repair pathway in many archaea [65], requires the presence of a DNA repair template and can be used to elicit the insertion of heterologous DNA or gene replacements [66,67].
Less frequently used CRISPR/Cas-based platforms also exist that make use of modified Cas enzymes (either catalytically inactive or with only nickase activity) fused to enzymes or domains with other functional capacities. These modified Cas enzymes are then guided to a selected genomic locus by the sgRNA without provoking a DSB [68]. Such platforms include, for example, cytosine and adenine base-editor systems, which allow for C-to-T or A-to-G substitutions [69,70], prime editing, which enables small, specific nucleotide changes to be made at a target locus [71,72], and epigenetic editing, which allows for the regulation of transcriptional activity [73,74]. While all of the aforementioned gene editing tools allow for the theoretical removal of the editing machinery once the edit has been achieved, other CRISPR/Cas platforms also exist that would necessitate the persistent presence of a Cas- and sgRNA(s)-encoding transgene in the host to maintain the desired outcome, including those resulting in transcriptional modulation through the direct activation or repression of target gene expression [75,76], or the cleavage of single-stranded RNA rather than double-stranded DNA [77,78]. While all of these platforms have value, those that require the enduring presence of a transgene would likely be more difficult to implement in an agricultural context due to regulatory constraints, as discussed below.
Many countries have recently modernized, or are in the process of modernizing, regulatory policies to incorporate gene editing technology into their frameworks. In the majority of countries, these updated processes apply across plants, microorganisms and animals, while in other countries, such as Canada, discussions are ongoing regarding regulatory procedures for microorganisms and animals. Gene editing-mediated improvements in organisms achieved using site-directed nucleases (SDN) such as Cas can typically be divided into three classes, including those involving the provocation of point mutations or indels through NHEJ (SDN-1; typically used for gene knock-out or knock-down), the introduction of short DNA insertions or base pair modifications using an external DNA repair template and HDR (SDN-2), and the insertion of longer DNA sequences such as a transgene (SDN-3). In terms of updated regulatory guidelines, organisms that have been modulated using SDN-1 gene editing approaches are not automatically subjected to regulation in countries such as the United States, Canada, Brazil, Argentina, Israel, Nigeria, Australia and Japan, for example [79]. Organisms bred using an SDN-2 approach, on the other hand, are only necessarily regulated in a small subset of these countries, such as Australia [80]. Finally, organisms in which an SDN-3 approach was used for improvement are typically considered to be ‘genetically modified’ (GM) in most countries, and are thus subject to regulatory processes [58,79]. In contrast, the European Union, New Zealand and South Africa currently consider all forms of gene-edited organisms to be GM, regardless of the approach used [79]. This global asynchrony will almost certainly be problematic in terms of trade going forward. In addition, gene editing technologies are advancing at such a rapid rate that even updated policies may not capture recent progress in the field. As such, there is a push towards the further harmonization of regulatory guidelines that are sufficiently flexible to encompass innovations in this area [58].

3. The Genetic Modulation of Forages for Reduced CH4 Emissions

3.1. CRISPR/Cas Gene Editing in Forages

While progress has been made in terms of enhancing various traits, including biomass production, stress tolerance and nutritional quality, using conventional breeding approaches in forage crops [81], the outcrossing and polyploid nature of many of these species complicates this process. As such, biotechnology has the potential to further our ability to elicit timely improvements in such crops. Although transgenic approaches, including over-expression and RNAi-mediated down-regulation, comprise the most common biotechnological strategies used in forages to date [54,82,83,84,85], issues related to public perception, as well as the exorbitant cost and time associated with regulatory processes, impedes the implementation of such plants in the field [58]. In contrast, most countries in which regulatory policies have been modernized to take into account gene-edited varieties have opted to treat edited plants lacking foreign DNA as they would conventionally bred plants, since the mutations elicited are derived from the plant’s own DNA repair mechanisms [58]. As such, the use of CRISPR/Cas is becoming increasingly popular in many crop species, including forages [86,87,88]. However, in its simplest and most commonly used form in plants, which depends upon the generation of NHEJ-mediated indels within coding regions, it is mainly limited to reducing or knocking out gene function [58]. Although several promising CRISPR/Cas-mediated NHEJ-based systems have shown promise for the up-regulation of gene expression or translation in plants, including the disruption of repressor elements within target gene promoters [89] or upstream open reading frames within 5′ untranslated regions of a target gene [90], they are far more complicated to apply and require the presence of the necessary regulatory elements. Therefore, negative regulators of selected pathways, which are often less well-characterized than positive regulators, typically must be identified and targeted to elicit desired phenotypic outcomes using CRISPR/Cas-mediated gene editing.

3.2. The Potential of Forage Lipid Modulation as a Means of Reducing CH4 Emissions from Ruminants

Feed supplementation in the form of vegetable oils or oilseeds has shown great promise for its ability to persistently reduce enteric CH4 emissions [91,92,93,94,95]. The ability of lipids to reduce CH4 emissions has been suggested to occur, at least in part, through the hydrogenation of unsaturated fatty acids, which provides an alternative sink for H2, as well as an inhibitory effect on methanogen activity and protozoal numbers [19,91,96]. In addition, since the amount of CH4 produced correlates with the quantity a ruminant eats, increasing the proportion of lipids in feed also reduces emissions through an augmentation in caloric density and consequent decrease in dry matter intake [97,98].
Unfortunately, the vegetative tissues of forage crops contain only very small amounts of lipids (typically 1.5–3% dry weight [DW]), which varies somewhat depending on the species [99], time of season and stage of development [99,100,101,102]. In addition, the drying of herbage, which is required for the production of hay, has been found to reduce total fatty acid contents even further [99,103]. Since dietary supplementation with vegetable oils can be prohibitively costly and impractical for producers [16], and there is also a competitive need for oilseeds for human consumption [104], it has been difficult to implement the use of such supplements on a large scale. Given that a 1% increase in the lipid content of livestock feed can reduce CH4 emissions by 3.5% to 5.6% [96,105], and lipids also provide more energy per unit than the other main components of plant cells (carbohydrates and proteins), the direct manipulation of forage crops themselves to accumulate lipids would be an ideal scenario in terms of economic viability for producers.
Plants contain a wide range of lipids, the composition of which diverges extensively among tissues. Unlike oilseeds, where the vast majority of lipids are in the form of the storage molecule triacylglycerol (TAG) [106], leaf lipids primarily comprise galactolipids (mainly monogalactosyldiacylglycerol [MGDG] and digalactosyldiacylglycerol), sulfolipids (mainly sulphoquinovosyldiacylglycerol) and phospholipids (mainly phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, phosphatidylserine and phosphatidylglycerol) that largely function as membrane constituents [107]. While only a very small proportion of TAG accumulates in plant photosynthetic tissues (generally less than 1% of total lipids) [108], it has been suggested that it plays an important role in protecting cells against oxidative damage and lipotoxicity by sequestering toxic free fatty acids [109], as well as in stress responses [110] and, to a small extent, as an energy store for periods of darkness [111].
The biosynthesis of the various lipid classes in both leaves and seeds begins with a common phase termed de novo fatty acid biosynthesis, which occurs in plastids and is initiated with the conversion of acetyl-coenzyme A (CoA) into malonyl-CoA via the catalytic action of acetyl-CoA carboxylase [112]. In photosynthetic tissues, the resulting malonyl-CoA is then used for the ultimate production of 16:0-, 18:0- and 18:1-acyl carrier proteins, which are then either utilized within the chloroplast for the production of various glycerolipids in the ‘prokaryotic’ lipid biosynthetic pathway, or are converted to free fatty acids, activated to CoA esters and exported out of the plastid for use in the ‘eukaryotic’ pathway that takes place on the endoplasmic reticulum (ER), where a small amount of TAG is generated in addition to other glycerolipids (Figure 2) [106]. In all plant species, a proportion of glycerolipids produced in the eukaryotic pathway in the ER are transferred back to plastids, where they serve as precursors for galactolipid biosynthesis [113]. In the majority of plants, this pathway is used exclusively for the biosynthesis of galactolipids, and due to the substrate preferences of enzymes utilized in the eukaryotic pathway, this results in a relatively high proportion of 18-carbon unsaturated fatty acids in the galactolipid fraction (these species are often referred to as ‘18:3 plants’). In contrast, a subset of plant species also synthesizes a proportion of galactolipids through the prokaryotic pathway in plastids, which results in a higher proportion of 16-carbon unsaturated fatty acids in their galactolipids (these species are often referred to as ‘16:3 plants’) [113,114].
Unfortunately, with the exception of preliminary evidence suggesting that incremental increases in shoot lipid content of alfalfa (Medicago sativa L.) and sainfoin (Onobrychis viciifolia Scop.) may be possible through ethyl methanesulfonate (EMS) mutagenesis [115], progress has been very limited in this context using conventional breeding approaches, at least in part because little genetic variability exists in this trait [99]. In contrast, substantial gains in vegetative lipid contents have been achieved in plants using transgenic approaches, largely through the combinatorial manipulation of multiple genes within lipid biosynthetic and degradation pathways, as well genes involved in the formation of lipid droplets and carbon availability. For example, TAG levels upwards of 30% DW have been achieved in tobacco (Nicotiana tabacum) leaves, which is akin to levels typically present in oilseeds [116]. However, this level of lipid augmentation is not desirable in a forage crop, since lipids at levels higher than approximately 6–7% of dietary dry matter limit the amount of organic matter that is fermented in the rumen and/or feed intake, which can have a negative impact on livestock performance and quality [117]. Smaller increases in total fatty acids have been achieved in forage species or their close relatives, such as ryegrass (Lolium perenne), sorghum (Sorghum bicolor) and Lotus japonicus, through the over-expression of DIACYLGLYCEROL ACYLTRANSFERASE 1 (DGAT1)/CYSTEINE OLEOSIN, WRINKLED 1 (WRI1)/DGAT2a/OLEOSIN-L, and MYB73, respectively [118,119,120]. In addition, it has been shown that the aforementioned ryegrass lines, which exhibited approximately 6.0–6.4% total fatty acids on a DW basis (compared to approximately 3.7% in wild-type plants), led to 10–15% reductions in CH4 production using in vitro assessments [121], which points to the potential usefulness of such an approach. However, the resulting plants are transgenic and are thus subject to strict regulatory policies, which hinders their implementation [58].
While there is growing interest in the use of gene editing approaches for the modulation of seed oil content [122,123], as of yet, relatively little progress has been made in terms of enhancing lipid levels in the vegetative tissues of plants using such methods. However, several negative regulators of shoot lipid accumulation have now been identified in plants [124,125,126], which largely function in pathways that compete with lipid/TAG biosynthesis or facilitate TAG turnover and have the potential to act as candidate targets for such endeavors in the future. While the down-regulation/mutation of genes encoding some of these regulators, such as sucrose-H+ symporter 2 (SUC2), has been found to lead to substantial phenotypic abnormalities [127], others have been shown to enhance vegetative lipid contents without substantial defects in biomass production, which makes them ideal targets for disruption via CRISPR/Cas.
One strategy that is being explored as a means of enhancing TAG contents in vegetative tissues involves reducing the proportion of substrates used for the biosynthesis of membrane lipids so that more are available for TAG production. For example, thylakoid lipid assembly in plastids requires a substantial proportion of fatty acids synthesized in both prokaryotic and eukaryotic pathways. In chloroplasts, phosphatidic acid (PA) is dephosphorylated through the catalytic action of PA phosphatase to generate diacylglycerol (DAG), which is a precursor of the predominant thylakoid membrane lipid MGDG. PA and DAG are also produced in the eukaryotic pathway on the ER, and it has been suggested that one or both of these molecules can then be transferred to chloroplasts by a group of trigalactosyldiacylglycerol (TGD) proteins, including TGD1-TGD5 located in plastid membranes [113,128]. Therefore, it is conceivable that reducing the amount of PA directed towards chloroplasts may concomitantly allow for increases in shoot TAG content. Indeed, in Arabidopsis, tgd1 and tgd5 mutants have been found to exhibit significant increases in TAG in vegetative tissues [127,128,129]. However, due to the importance of thylakoid membranes in plants, these mutants have been found to display a moderate reduction in growth [128,130]. A similar phenomenon was also observed in tobacco and Arabidopsis whereby the MONOGALACTOSYLDIACYLGLYCEROL SYNTHASE 1 gene (MGD1), which encodes an enzyme that catalyzes the conversion of DAG to MGDG, was down-regulated or mutated [131,132]. Furthermore, the down-regulation of a TGD5 homolog in Medicago truncatula, which is a close diploid relative of alfalfa, did not significantly impact shoot TAG accumulation in transient assessments using virus-induced gene silencing (VIGS) [126]. Taken together with the fact that increasing TAG at the expense of other lipids would likely not lead to increases in total lipid content, these findings indicate that such a strategy may not be suitable for CRISPR/Cas-based applications in forages.
An alternative approach would be to enhance the availability of substrates for overall lipid biosynthesis in vegetative tissues by reducing the proportion of carbon used for the production of non-lipid-related compounds in plant cells, which would theoretically make additional carbon available for lipid biosynthesis. For instance, starch and oil biosynthesis pathways compete with one another for assimilated carbon precursors in plants, and the disruption of key enzymes involved in the starch biosynthetic pathway has been found to enhance lipid levels in vegetative tissues previously. In the starch biosynthetic pathway, the rate-limiting step comprises the conversion of glucose-1-phosphate (G1P) to adenosine diphosphate glucose (ADP-glucose), which acts as the glucosyl donor for starch synthases, via the catalytic action of ADP-glucose pyrophosphorylase (AGPase) [133]. In Arabidopsis, the mutation/down-regulation of the gene encoding the catalytic isoform of the small subunit of AGPase led to significant increases in vegetative TAG compared to the wild type [127,134], while its VIGS-mediated down-regulation in M. truncatula resulted in a significant elevation in total shoot lipid content upwards of 4.5% DW [126]. In addition, although there is some evidence that the mutation of these genes may lead to a slight reduction in growth and early flowering, at least under short days, in Arabidopsis, these alterations appear to be species and/or isoform-specific [127,135]. For example, rice, maize, barley and potato with disrupted AGPase activity have been found to display normal vegetative growth with morphological abnormalities only present in certain instances in starch-accumulating organs, such as grains or tubers [136,137,138,139,140]. Mutants deficient in AGPase also tend to exhibit substantial increases in soluble carbohydrate levels at the expense of starch [127,141,142], which may have the potential to reduce the incidence of ruminal acidosis [143] and further decrease CH4 emissions [144]. However, such an inverse correlation between starch and lipid levels has not always been observed when this gene is disrupted/down-regulated on its own [142], and in many cases, it appears that this strategy instead provides an additive contribution to enhancements in vegetative lipid content when combined with modifications in a variety of other genes [127,134,142,145]. As such, further research on forage species is required to determine the suitability of such an approach for reducing CH4 emissions and improving livestock health in the future.
Reducing the breakdown of TAG has also been demonstrated as a potential strategy for eliciting enhanced lipid accumulation in vegetative tissues. For instance, the mutation of SUGAR DEPENDENT1 (SDP1), which encodes a patatin-like acyl-hydrolase domain-containing lipase that hydrolyzes TAG into glycerol and free fatty acids [146], has been found to enhance TAG content in Arabidopsis vegetative tissues [109,125,142]. Similarly, the mutation of PEROXISOMAL ABC TRANSPORTER 1 (PXA1), which functions in the transport of fatty acids into peroxisomes for β-oxidation, as well as COMPARATIVE GENE IDENTIFICATION-58 (CGI-58), which acts cooperatively with PXA1 to facilitate lipid turnover [147], in Arabidopsis has also been found to lead to significant increases in TAG accumulation in vegetative tissues [124,147]. In line with this, the VIGS-induced down-regulation of SDP1 and PXA1 homologs in M. truncatula prompted up to 40% increases in total shoot lipid contents [126]. Furthermore, while both sdp1 and pxa1 Arabidopsis mutants displayed detriments in seed germination and early post-germinative growth in the absence of sucrose, they are otherwise similar to the wild type in their growth following the onset of photosynthesis [146,147,148], and no obvious phenotypic effects have been noted in cgi-58 mutants to date [124,147]. It is possible that the germination/post-germinative deficits apparent in Arabidopsis sdp1 and pxa1 mutants are specific to oilseed species due to the fact that these processes rely upon the breakdown of TAG as an energy source prior to the initiation of photosynthesis in young seedlings [146], and as such, these same consequences may not be apparent in other plant groups, such as legume or grass forages. Taken together, this suggests that all three genes may provide interesting candidates for CRISPR/Cas-mediated mutation in forages.
Furthermore, a combinatory approach involving the simultaneous disruption/down-regulation of genes encoding AGPase subunits and SDP1/PXA1 may provide even further increases in vegetative TAG/lipid contents as has been demonstrated in Arabidopsis and potato previously [142,149]. Similarly, the deletion of a region immediately upstream of DGAT2, including a non-essential photosynthesis-related gene and the DGAT2 promoter, using CRISPR/Cas9 with dual sgRNAs in the Arabidopsis sdp1 mutant allowed the DGAT2 coding sequence to fall under the control of the more highly expressed upstream gene. This led to the up-regulation of DGAT2 expression in leaves [150]. Since DGAT2 encodes a key enzyme of acyl-CoA-dependent TAG biosynthesis [151], this novel NHEJ-mediated gain-of-function gene editing approach allowed for the doubling of total leaf lipid contents [150], which would be ideal in a forage crop for ruminant production. However, it is possible that these combinatorial approaches could be associated with a decline in biomass production [142,150], although in the latter case, this decrease appears to be minor and further research is required to determine whether it is significant under field conditions.
While all of these strategies hold promise for enhancing lipid content in forage shoots, which could feasibly mitigate CH4 emissions from the ruminants that feed on them, there is some evidence that not only lipid content, but also lipid composition, may play a role in such an outcome. For example, medium-chain and unsaturated fatty acids appear to provide the greatest reduction in CH4 emissions, in the latter case potentially through their ability to act as electron acceptors during biohydrogenation [152,153]. Therefore, it is likely that fatty acid compositional changes in edited genotypes also need to be taken into consideration in this context.

3.3. The Potential of Other Forage Traits to Reduce CH4 Emissions from Ruminants

In addition to the modulation of forage shoot lipid content/quality, there is evidence that several other forage traits may also have potential in terms of mitigating CH4 emissions from ruminants. For example, forages containing condensed tannins (CTs), which are polyphenolic secondary metabolites that bind proteins and thus slow down their degradation in the rumen and also function in plant defense and stress response mechanisms [154], have been shown to reduce CH4 emissions in ruminants in vivo [155,156]. Although the mechanisms driving CH4 mitigation in this case are not well-understood, they may derive, at least in part, from a decline in fiber digestibility, an ability to act as a H2 sink and/or the direct inhibition of methanogens [157,158]. However, results have been highly inconsistent among studies [154]; it is a phenomenon that has been suggested to be attributable to differences in CT composition occurring within and among species, as well as environmental effects [159,160,161] and the co-presence of other secondary plant metabolites [162]. Unfortunately, relatively little is currently known regarding such effects, and further research is necessary to advance our understanding of CTs in forage crops. Similarly, increasing the digestibility of forages has also been suggested as a possible means of mitigating CH4 emissions from ruminants [163]. As is the case with CTs, however, the effect has been rather inconsistent among studies [164], and since enhanced digestibility tends to increase dry matter intake, CH4 emissions have been found to increase in certain cases [163].

4. The Genetic Modulation of Rumen Methanogens for Reduced CH4 Emissions

4.1. CRISPR/Cas Gene Editing in Archaea

The S. pyogenes-derived CRISPR/Cas9 platform has been shown to lead to the successful introduction of insertions (knock-in) and deletions (knock-out) through HDR with high efficiency in the methanogen, Methanosarcina acetivorans, without any evidence of off-target mutations [165]. Furthermore, as is the case in plants, once editing had been achieved in the target gene, plasmids bearing the gene editing machinery could successfully be removed from edited strains [165]. While this means that the resulting knock-out strains lacked foreign DNA, the same was not the case for knock-in strains, as a DNA fragment was inserted into the genome in this instance. The fact that transformation efficiencies were thousands of times higher when a DNA repair template was used corroborates that HDR is the preferred DNA repair mechanism in archaea, with NHEJ-mediated repair being extremely rare [65]. However, the co-introduction of NHEJ machinery from the closely related Methanocella paludicola, which is the only archaeal species in which a complete NHEJ complex has been detected thus far [166], also allowed for the efficient provision of targeted deletions without any need for a repair template [165].
Since most archaea encode adaptive CRISPR/Cas-based immune systems, another strategy for achieving genome editing is to make use of their endogenous apparatus. This means that only sgRNA(s) and DNA templates would need to be introduced to elicit HDR [167]. In line with this, endogenous class 1, type I and III CRISPR/Cas systems have been successfully used to introduce deletions, insertions and point mutations in the genome of the thermophilic archaeon, Sulfolobus islandicus [168], and it is feasible that a similar strategy would apply across all archaea that encode active CRISPR/Cas systems. To date, native CRISPR/Cas systems have been reported in multiple Methanobrevibacter strains, including M. arboriphilicus ANOR1 and M. oralis JMR01 [169,170], which suggests that such an approach would be valuable for the modulation of rumen methanogens.

4.2. Potential CRISPR/Cas-Based Approaches in Rumen Methanogens to Mitigate CH4 Emissions

The microbial community in the rumen is one of the most diverse gut ecosystems in the animal kingdom, including archaea, bacteria, protozoa, fungi and a largely unknown virome [171]. Although gaps remain in our understanding of these communities at present, the recent introduction and integration of metagenomic, transcriptomic, proteomic and metabolomic techniques provides the possibility of acquiring a systems-level understanding of microorganism species composition in the rumen and the role of such associations on host metabolism [172,173,174,175].
Archaea are the second most abundant microbial community in the rumen, of which approximately 77% are hydrogenotrophic methanogens, which reduce CO2 to CH4 with H2 as an electron donor, and 22% are methylotrophic methanogens, which instead make use of small methylated carbon compounds [176,177]. While the two most prominent groups of rumen methanogens are Methanobrevibacter gottschalkii and Methanobrevibacter ruminantium, with other genera such as Methanomicrobium contributing smaller proportions, an animal’s diet can influence their abundance in the rumen [176,178]. Interestingly, the methanogens Methanobrevibacter and Methanomicrobium can also form a symbiotic relationship with protozoa [179], allowing for the H2 generated by fermenters to be utilized by methanogens, which in turn keeps H2 concentrations low and favors the metabolism of the fermenters [180].
Recent studies have also revealed that rumen microbial population structure and function are influenced not only by nutritional factors, but also by host genetics [174,181]. For example, an examination of the association of host genetics with the phylogenetic and functional composition of the rumen microbiome led to the identification of 22 operational taxonomic units (OTUs) whose abundances were associated with rumen metabolic traits and host physiological traits with measurable heritability [182]. Similarly, genome-wide association analysis of a common set of single-nucleotide polymorphisms found in cattle genomes and corresponding rumen bacterial community composition led to the identification of OTUs, families and phyla with high heritability [183]. Although our understanding of how a stable rumen microbial community assembles itself and what factors affect microbial composition/function is still limited, these findings indicate that certain rumen microbial features are heritable.
The availability of the M. ruminantium genome sequence [184] and the recent prediction of its operome functional properties [185], along with the fact that the biochemistry of methanogenic pathways is well-known [186,187], are opening up new possibilities of using genetic strategies to modulate CH4 production in the rumen. This highlights the possibility of further improving animal production efficiency and mitigating the emission of GHGs from ruminants using gene editing strategies on rumen archaea in vitro, and subsequently re-introducing them into the rumen (Figure 3). While such an approach holds promise, further research is essential to gain a better understanding of the key microorganisms involved in rumen methanogenesis, as well as their activities, to successfully manipulate rumen processes in this manner.
As is the case with plants, one of the biggest challenges of using gene editing in rumen methanogens to reduce CH4 emissions is the identification of suitable target genes, which can be difficult due to the intricacy of the methanogenesis process. Indeed, targeting an inappropriate gene would not only be ineffective, but could also lead to the generation of less competitive methanogens in the rumen microbiome or even be lethal. Intriguingly, it has been shown that the transcript abundance of genes encoding enzymes involved in the hydrogenotrophic methanogenesis pathway is significantly lower in the rumen of sheep producing low CH4 emissions than in those of sheep with high CH4 emissions [188]. The largest transcriptional differences were apparent in genes that encode the key MCR enzyme [188], which is found in all methanogens and is essential for their growth [189]. Although such differences in the transcriptomes of rumen methanogens between ruminants with low and high CH4 emissions could arise due to a variety of reasons, one can speculate that the down-regulation of particular genes involved in hydrogenotrophic methanogenesis could be a useful strategy to reduce CH4 emissions. Unfortunately, deletions introduced through HDR-mediated CRISPR/Cas in genes encoding an MCR and a heterodisulfide reductase (hdrED), which is another essential component of methanogenesis in methylotrophic pathways, in M. acetivorans yielded very few transformants, indicating the lethality of these genetic modifications [165]. In contrast, deletions in genes encoding monomethylamina-specific methyltransferases (mtmCB1 and mtmCB2), which enable methylamine utilization in the methylotrophic pathway, led to the production of thousands of transformants [165], which suggests that the knockout of methyltransferase genes may be a superior target choice in the context of reducing CH4 production in the rumen, at least in the case of ruminant methanogens that utilize the methylotrophic pathway.
In addition to knock-outs achieved through gene interruption, reductions in gene expression can also be achieved using a CRISPR interference system (CRISPRi), which is achieved without eliciting any break in DNA strands and may provide benefits when targeting essential genes such as MCR. For example, the expression of a target gene can be repressed using an inactive Cas9 (with no endonucleolytic activity; dCas9), which together with an sgRNA binds to the target DNA sequence of a protein-coding region to block RNA polymerase and transcript elongation [190,191]. In M. acetivorans, such a CRISPRi system has been stably integrated into the genome or converted into a replicating plasmid, resulting in up to a 90% reduction in target gene transcript abundance [192]. A similar, but alternative, approach to the use of dCas9 for CRISPRi is to make use of an endogenous Cas protein that targets RNAs rather than double-stranded DNA for cleavage, such as in the class 1 type III-B CRISPR/Cas system [193] that can be found in at least certain archaea [194]. This approach has been successfully applied in Sulfolobus solfataricus to down-regulate specific RNAs by taking advantage of an endogenous endonuclease along with an introduced guide crRNA and could theoretically also be applicable in other archaea [195,196]. However, with all types of CRISPRi, the editing machinery and/or gRNA are required long-term in the archaeal strain to maintain the knock-down effect, and the transgenic nature of the resulting strains in the rumen of live cattle could potentially hinder implementation, at least in the short term.

5. Conclusions

Given the considerable role of rumen methanogens in the emission of GHGs, it will be of paramount importance in coming years to target this process for improvement as a contribution towards mitigating the worst climate change scenarios. While this will likely entail a multi-pronged approach involving livestock genetic factors and possible supplementation, the direct CRISPR/Cas-based manipulation of forages and rumen methanogens could conceivably provide two of a combination of strategies with additive effects to achieve such an outcome. As such, these approaches warrant further study to determine their efficacy in this context and also to strike a balance among effectiveness, methanogen or plant competitiveness, animal performance, consumer acceptance and ease of implementation. Although regulatory processes could conceivably impede the global adoption of gene-edited archaea and crops due to discrepancies among nations, it is likely that those generated using SDN-1 and/or SDN-2 approaches will be treated as non-GM in the vast majority of countries. Considering the opportunities that gene editing can offer to mitigate GHG emissions in an agricultural context, a regulatory alignment among countries will be of utmost importance to safely stimulate innovation and avoid further trade barriers in the future.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful for the support provided by Agriculture and Agri-Food Canada (S.D.S., H.P., S.A.), the Beef Cattle Research Council (S.D.S., H.P.), the Saskatchewan Agriculture Development Fund (S.D.S., H.P.), Alberta Milk (S.D.S.), the Saskatchewan Cattlemen’s Association (S.D.S.), the Canada Research Chairs Program (G.C.), the Natural Sciences and Engineering Research Council of Canada (G.C.), and the Brazilian Agricultural Research Corporation (Embrapa, Brazil) (L.S.A.C., D.M.C.B.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Minx, J.C.; Lamb, W.F.; Andrew, R.M.; Canadell, J.G.; Crippa, M.; Döbbeling, N.; Forster, P.M.; Guizzardi, D.; Olivier, J.; Peters, G.P.; et al. A comprehensive and synthetic dataset for global, regional, and national greenhouse gas emissions by sector 1970–2018 with an extension to 2019. Earth Syst. Sci. Data 2021, 13, 5213–5252. [Google Scholar] [CrossRef]
  2. Intergovernmental Panel on Climate Change (IPCC). Climate Change 2021: The Physical Science Basis. In Contribution of Working Group I to the Sixth Assessment Report of the Intergovernmental Panel on Climate Change, Technical Summary; Masson-Delmotte, V.P., Zhai, P., Pirani, A., Connors, S.L., Péan, C., Chen, Y., Goldfarb, L., Gomi, M.I., Matthews, J.B.R., Berger, S., et al., Eds.; Cambridge University Press: Cambridge, UK, 2021; Available online: https://www.ipcc.ch/report/ar6/wg1/downloads/report/IPCC_AR6_WGI_TS.pdf (accessed on 28 July 2022).
  3. Tito, R.; Vasconcelos, H.L.; Feeley, K.J. Global climate change increases risk of crop yield losses and food insecurity in the tropical Andes. Glob. Chang. Biol. 2017, 24, e592–e602. [Google Scholar] [CrossRef] [PubMed]
  4. Arora, N.K. Impact of climate change on agriculture production and its sustainable solutions. Environ. Sustain. 2019, 2, 95–96. [Google Scholar] [CrossRef] [Green Version]
  5. Cammarano, D.; Ceccarelli, S.; Grando, S.; Romagosa, I.; Benbelkacem, A.; Akar, T.; Al-Yassin, A.; Pecchioni, N.; Francia, E.; Ronga, D. The impact of climate change on barley yield in the Mediterranean basin. Eur. J. Agron. 2019, 106, 1–11. [Google Scholar] [CrossRef]
  6. Malhi, Y.; Franklin, J.; Seddon, N.; Solan, M.; Turner, M.G.; Field, C.B.; Knowlton, N. Climate change and ecosystems: Threats, opportunities and solutions. Phil. Trans. R. Soc. B Biol. Sci. 2020, 375, 20190104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Román-Palacios, C.; Wiens, J.J. Recent responses to climate change reveal the drivers of species extinction and survival. Proc. Natl. Acad. Sci. USA 2020, 117, 4211–4217. [Google Scholar] [CrossRef] [PubMed]
  8. Höhne, N.; Gidden, M.J.; den Elzen, M.; Hans, F.; Fyson, C.; Geiges, A.; Jeffery, M.L.; Gonzales-Zuñiga, S.; Mooldijk, S.; Hare, W.; et al. Wave of net zero emission targets opens window to meeting the Paris Agreement. Nat. Clim. Chang. 2021, 11, 820–822. [Google Scholar] [CrossRef]
  9. IPCC. Climate Change 2022: Mitigation of Climate Change. In Working Group III Contribution to the IPCC Sixth Assessment Report (AR6); IPCC: Geneva, Switzerland, 2022; Available online: https://report.ipcc.ch/ar6wg3/pdf/IPCC_AR6_WGIII_FinalDraft_FullReport.pdf (accessed on 28 July 2022).
  10. IPCC. Climate Change 2014: Mitigation of Climate Change. In Contribution of Working Group III to the Fifth Assessment Report of the Intergovernmental Panel on Climate Change; Edenhofer, O., Pichs-Madruga, R., Sokona, Y., Farahani, E., Kadner, S., Seyboth, K., Adler, A., Baum, I., Brunner, S., Eickemeier, P., et al., Eds.; Cambridge University Press: Cambridge, UK, 2014; Available online: https://www.ipcc.ch/site/assets/uploads/2018/02/ipcc_wg3_ar5_full.pdf (accessed on 28 July 2022).
  11. Jackson, R.B.; Saunois, M.; Bousquet, P.; Canadell, J.G.; Poulter, B.; Stavert, A.R.; Bergamaschi, P.; Niwa, Y.; Segers, A.; Tsuruta, A. Increasing anthropogenic methane emissions arise equally from agricultural and fossil fuel sources. Environ. Res. Lett. 2020, 15, 071002. [Google Scholar] [CrossRef]
  12. Gerber, P.J.; Steinfeld, H.; Henderson, B.; Motter, A.; Opio, C.; Dijkman, J.; Falcucci, A.; Tempio, G. Tackling Climate Change through Livestock—A Global Assessment of Emissions and Mitigation Opportunities; Food and Agriculture Organization of the United Nations (FAO): Rome, Italy, 2013; Available online: https://www.fao.org/3/i3437e/i3437e.pdf (accessed on 28 July 2022).
  13. Reisinger, A.; Clark, H.; Cowie, A.L.; Emmet-Booth, J.; Gonzalez Fischer, C.; Herrero, M.; Howden, M.; Leahy, S. How necessary and feasible are reductions of methane emissions from livestock to support stringent temperature goals? Phil. Trans. R. Soc. A 2021, 379, 20200452. [Google Scholar] [CrossRef] [PubMed]
  14. Searchinger, T.; Waite, R.; Hanson, C.; Ranganathan, J. Creating a Sustainable Food Future; World Resources Institute: Washington, DC, USA, 2019; Available online: https://files.wri.org/d8/s3fs-public/wrr-food-full-report.pdf (accessed on 30 August 2022).
  15. Leng, R.A. Interactions between microbial consortia in biofilms: A paradigm shift in rumen microbial ecology and enteric methane mitigation. Anim. Prod. Sci. 2014, 54, 519. [Google Scholar] [CrossRef]
  16. Beauchemin, K.A.; Ungerfield, E.M.; Eckard, R.J.; Wang, M. Fifty years of research on rumen methanogenesis: Lessons learned and future challenges for mitigation. Animal 2020, 14, s2–s16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Duin, E.C.; Wagner, T.; Shima, S.; Prakash, D.; Cronin, B.; Yáñez-Ruiz, D.R.; Duval, S.; Rümbeli, R.; Stemmler, R.T.; Thauer, R.K.; et al. Mode of action uncovered for the specific reduction of methane emissions from ruminants by the small molecule 3-nitrooxypropanol. Proc. Natl. Acad. Sci. USA 2016, 113, 6172–6177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Van Nevel, C.J.; Demeyer, D.I. Control of rumen methanogenesis. Environ. Monit. Assess. 1996, 42, 73–97. [Google Scholar] [CrossRef]
  19. Johnson, K.A.; Johnson, D.E. Methane emissions from cattle. J. Anim. Sci. 1995, 73, 2483–2492. [Google Scholar] [CrossRef] [PubMed]
  20. McGeough, E.J.; O’Kiely, P.; Hart, K.J.; Moloney, A.P.; Boland, T.M.; Kenny, D.A. Methane emissions, feed intake, performance, digestibility, and rumen fermentation of finishing beef cattle offered whole-crop wheat silages differing in grain content. J. Anim. Sci. 2010, 88, 2703–2716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Van Wyngaard, J.D.V.; Meeske, R.; Erasmus, L.J. Effect of concentrate feeding level on methane emissions, production performance and rumen fermentation of Jersey cows grazing ryegrass pasture during spring. Anim. Feed. Sci. Technol. 2018, 241, 121–132. [Google Scholar] [CrossRef] [Green Version]
  22. DeRamus, H.A.; Clement, T.C.; Giampola, D.D.; Dickison, P.C. Methane emissions of beef cattle on forages. J. Environ. Qual. 2003, 32, 269–277. [Google Scholar] [CrossRef] [PubMed]
  23. Pickering, N.K.; Oddy, V.H.; Basarab, J.; Cammack, K.; Hayes, B.; Hegarty, R.S.; Lassen, J.; McEwan, J.C.; Miller, S.; Pinares-Patiño, C.S.; et al. Genetic possibilities to reduce enteric methane emissions from ruminants. Animal 2015, 9, 1431–1440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. McAllister, T.A.; Newbold, C.J. Redirecting rumen fermentation to reduce methanogenesis. Anim. Prod. Sci. 2008, 48, 7–13. [Google Scholar] [CrossRef]
  25. Lee, S.S.; Hsu, J.T.; Mantovani, H.C.; Russell, J.B. The effect of bovicin HC5, a bacteriocin from Streptococcus bovis HC5, on ruminal methane production in vitro. FEMS Microbiol. Lett. 2002, 217, 51–55. [Google Scholar] [CrossRef]
  26. Sar, C.; Mwenya, B.; Pen, B.; Morikawa, R.; Takaura, K.; Kobayashi, T.; Takahashi, J. Effect of nisin on ruminal methane production and nitrate/nitrite reduction in vitro. Aust. J. Agric. Res. 2005, 56, 803–810. [Google Scholar] [CrossRef]
  27. Baca-González, V.; Asensio-Calavia, P.; González-Acosta, S.; de la Lastra, J.M.P.; de la Nuez, A.M. Are vaccines the solution for methane emissions from ruminants? A systematic review. Vaccines 2020, 8, 460. [Google Scholar] [CrossRef] [PubMed]
  28. Beauchemin, K.A.; McGinn, S.M.; Petit, H.V. Methane abatement strategies for cattle: Lipid supplementation of diets. Can. J. Anim. Sci. 2007, 87, 431–440. [Google Scholar] [CrossRef]
  29. Animut, G.; Puchala, R.; Goetsch, A.L.; Patra, A.K.; Sahlu, T.; Varel, V.H.; Wells, J. Methane emission by goats consuming diets with different levels of condensed tannins from lespedeza. Anim. Feed Sci. Technol. 2008, 144, 212–227. [Google Scholar] [CrossRef]
  30. Beauchemin, K.A.; McAllister, T.A.; McGinn, S.M. Dietary mitigation of enteric methane from cattle. Anim. Nutr. 2020, 6, 231–246. [Google Scholar] [CrossRef]
  31. Tomkins, N.W.; Colegate, S.M.; Hunter, R.A. A bromochloromethane formulation reduces enteric methanogenesis in cattle fed grain-based diets. Anim. Prod. Sci. 2009, 49, 1053–1058. [Google Scholar] [CrossRef]
  32. Appuhamy, J.R.N.; Strathe, A.B.; Jayasundara, S.; Wagner-Riddle, C.; Dijkstra, J.; France, J.; Kebreab, E. Anti-methanogenic effects of monensin in dairy and beef cattle: A meta-analysis. J. Dairy Sci. 2013, 96, 5161–5173. [Google Scholar] [CrossRef] [Green Version]
  33. Hristov, A.N.; Lee, C.; Cassidy, T.T.; Heyler, K.; Tekippe, J.A.; Varga, G.A.; Corl, B.; Brandt, R.C. Effect of Origanum vulgare L. leaves on rumen fermentation, production, and milk fatty acid composition in lactating dairy cows. J. Dairy Sci. 2013, 96, 1189–1202. [Google Scholar] [CrossRef]
  34. Moate, P.J.; Williams, S.R.O.; Torok, V.A.; Hannah, M.C.; Ribaux, B.E.; Tavendale, M.H.; Eckard, R.J.; Jacobs, J.L.; Auldist, M.J.; Wales, W.J. Grape marc reduces methane emissions when fed to dairy cows. J. Dairy Sci. 2014, 97, 5073–5087. [Google Scholar] [CrossRef] [Green Version]
  35. Dijkstra, J.; Bannink, A.; France, J.; Kebreab, E.; van Gastelen, S. Antimethanogenic effects of 3-nitrooxypropanol depend on supplementation dose, dietary fiber content, and cattle type. J. Dairy Sci. 2018, 101, 9041–9047. [Google Scholar] [CrossRef]
  36. Martinez-Fernandez, G.; Duval, S.; Kindermann, M.; Schirra, H.J.; Denman, S.E.; McSweeney, C.S. 3-NOP vs. halogenated compound: Methane production, ruminal fermentation and microbial community response in forage fed cattle. Front. Microbiol. 2018, 9, 1582. [Google Scholar] [CrossRef] [PubMed]
  37. Hart, K.J.; Jones, H.G.; Waddams, K.E.; Worgan, H.J.; Zweifel, B.; Newbold, C.J. An essential oil blend decreases methane emissions and increases milk yield in dairy cows. Open J. Anim. Sci. 2019, 9, 259. [Google Scholar] [CrossRef] [Green Version]
  38. Roque, B.M.; Van Lingen, H.J.; Vrancken, H.; Kebreab, E. Effect of Mootral: A garlic- and citrus-extract-based feed additive: On enteric methane emissions in feedlot cattle. Transl. Anim. Sci. 2019, 3, 1383–1388. [Google Scholar] [CrossRef]
  39. Feng, X.Y.; Dijkstra, J.; Bannink, A.; van Gastelen, S.; France, J.; Kebreab, E. Anti-methanogenic effects of nitrate supplementation in cattle: A meta-analysis. J. Dairy Sci. 2020, 103, 11375–11385. [Google Scholar] [CrossRef] [PubMed]
  40. Roque, B.M.; Venegas, M.; Kinley, R.D.; de Nys, R.; Duarte, T.L.; Yang, X.; Kebreab, E. Red seaweed (Asparagopsis taxiformis) supplementation reduces enteric methane by over 80 percent in beef steers. PLoS ONE 2021, 16, e0247820. [Google Scholar] [CrossRef] [PubMed]
  41. Machado, L.; Magnusson, M.; Paul, N.A.; Kinley, R.; de Nys, R.; Tomkins, N. Identification of bioactives from the red seaweed Asparagopsis taxiformis that promote antimethanogenic activity in vitro. J. Appl. Phycol. 2016, 28, 3117–3126. [Google Scholar] [CrossRef]
  42. Roque, B.M.; Brooke, C.G.; Ladau, J.; Polley, T.; Marsh, L.J.; Najafi, N.; Pandey, P.; Singh, L.; Kinley, R.; Salwen, J.K.; et al. Effect of the macroalgae Asparagopsis taxiformis on methane production and rumen microbiome assemblage. Anim. Microbiome 2019, 1, 3. [Google Scholar] [CrossRef] [Green Version]
  43. Li, X.; Norman, H.C.; Kinley, R.D.; Laurence, M.; Wilmot, M.; Bender, H.; de Nys, R.; Tomkins, N. Asparagopsis taxiformis decreases enteric methane production from sheep. Anim. Prod. Sci. 2018, 58, 681–688. [Google Scholar] [CrossRef]
  44. Roque, B.M.; Salwen, J.K.; Kinley, R.; Kebreab, E. Inclusion of Asparagopsis armata in lactating dairy cows’ diet reduces enteric methane emission by over 50%. J. Clean Prod. 2019, 234, 132–138. [Google Scholar] [CrossRef]
  45. Kinley, R.D.; Martinez-Fernandez, G.; Matthews, M.K.; de Nys, R.; Magnusson, M.; Tomkins, N.W. Mitigating the carbon footprint and improving productivity of ruminant livestock agriculture using a red seaweed. J. Clean Prod. 2020, 59, 120836. [Google Scholar] [CrossRef]
  46. Stefenoni, H.A.; Räisänen, S.E.; Cueva, S.F.; Wasson, D.E.; Lage, C.F.A.; Melgar, A.; Fetter, M.E.; Smith, P.; Hennessy, M.; Vecchiarelli, B.; et al. Effects of the macroalga Asparagopsis taxiformis and oregano leaves on methane emission, rumen fermentation, and lactational performance of dairy cows. J. Dairy Sci. 2021, 104, 4157–4173. [Google Scholar] [CrossRef] [PubMed]
  47. Min, B.R.; Parker, D.; Brauer, D.; Waldrip, H.; Lockard, C.; Hales, K.; Akbay, A.; Augyte, S. The role of seaweed as a potential dietary supplementation for enteric methane mitigation in ruminants: Challenges and opportunities. Anim. Nut. 2021, 7, 1371–1387. [Google Scholar] [CrossRef] [PubMed]
  48. Haisan, J.; Sun, Y.; Guan, L.L.; Beauchemin, K.A.; Iwaasa, A.; Duval, S.; Barreda, D.R.; Oba, M. The effects of feeding 3-nitrooxypropanol on methane emissions and productivity of Holstein cows in mid lactation. J. Dairy Sci. 2014, 97, 3110–3119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hristov, A.N.; Oh, J.; Giallongo, F.; Frederick, T.W.; Harper, M.T.; Weeks, H.L.; Branco, A.F.; Moate, P.J.; Deighton, M.H.; Williams, S.R.O.; et al. An inhibitor persistently decreased enteric methane emission from dairy cows with no negative effect on milk production. Proc. Natl. Acad. Sci. USA 2015, 112, 10663–10668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Lopes, J.C.; de Matos, L.F.; Harper, M.T.; Giallongo, F.; Oh, J.; Gruen, D.; Ono, S.; Kindermann, M.; Duval, S.; Hristov, A.N. Effect of 3-nitrooxypropanol on methane and hydrogen emissions, methane isotopic signature, and ruminal fermentation in dairy cows. J. Dairy Sci. 2016, 99, 5335–5344. [Google Scholar] [CrossRef] [Green Version]
  51. Vyas, D.; McGinn, S.M.; Duval, S.M.; Kindermann, M.; Beauchemin, K.A. Effects of sustained reduction of enteric methane emissions with dietary supplementation of 3-nitrooxypropanol on growth performance of growing and finishing beef cattle. J. Anim. Sci. 2016, 94, 2024–2034. [Google Scholar] [CrossRef] [Green Version]
  52. Vyas, D.; Alemu, A.W.; McGinn, S.M.; Duval, S.M.; Kindermann, M.; Beauchemin, K.A. The combined effects of supplementing monensin and 3-nitrooxypropanol on methane emissions, growth rate, and feed conversion efficiency in beef cattle fed high-forage and high-grain diets. J. Anim. Sci. 2018, 96, 2923–2938. [Google Scholar] [CrossRef]
  53. Melgar, A.; Welter, K.C.; Nedelkov, K.; Martins, C.M.M.R.; Harper, M.T.; Oh, J.; Räisänen, S.E.; Chen, X.; Cueva, S.F.; Duval, S.; et al. Dose-response effect of 3-nitrooxypropanol on enteric methane emissions in dairy cows. J. Dairy Sci. 2020, 103, 6145–6156. [Google Scholar] [CrossRef]
  54. Singer, S.D.; Hannoufa, A.; Acharya, S. Molecular improvement of alfalfa for enhanced productivity and adaptability in a changing environment. Plant Cell Environ. 2018, 41, 1955–1971. [Google Scholar] [CrossRef]
  55. Hünerberg, M.; Little, S.M.; Beauchemin, K.A.; McGinn, S.M.; O’Connor, D.; Okine, E.K.; Harstad, O.M.; Kröbel, R.; McAllister, T. Feeding high concentrations of corn dried distillers’ grains decreases methane, but increases nitrous oxide emissions from beef cattle production. Agric. Syst. 2014, 127, 19–27. [Google Scholar] [CrossRef]
  56. Lee, C.; Beauchemin, K.A. A review of feeding supplementary nitrate to ruminant animals: Nitrate toxicity, methane emissions, and production performance. Can. J. Anim. Sci. 2014, 94, 557–570. [Google Scholar] [CrossRef]
  57. Honan, M.; Feng, X.; Tricarico, J.M.; Kebreab, E. Feed additives as a strategic approach to reduce enteric methane production in cattle: Modes of action, effectiveness and safety. Anim. Prod. Sci. 2022, 62, 1303–1317. [Google Scholar] [CrossRef]
  58. Singer, S.D.; Laurie, J.; Bilichak, A.; Kumar, S.; Singh, J. Genetic variation and unintended risk in the context of old and new breeding techniques. Crit. Rev. Plant Sci. 2021, 40, 68–108. [Google Scholar] [CrossRef]
  59. Paques, F.; Duchateau, P. Meganucleases and DNA double-strand break-induced recombination: Perspectives for gene therapy. Curr. Gene Ther. 2007, 7, 49–66. [Google Scholar] [CrossRef] [PubMed]
  60. Papaioannou, I.; Simons, J.P.; Owen, J.S. Oligonucleotide-directed gene-editing technology: Mechanisms and future prospects. Exp. Opin. Biol. Ther. 2012, 12, 329–342. [Google Scholar] [CrossRef]
  61. Gaj, T.; Gersbach, C.A.; Barbas, C.F., III. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013, 31, 397–405. [Google Scholar] [CrossRef] [Green Version]
  62. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  63. Meng, X.; Hu, X.; Liu, Q.; Song, X.; Gao, C.; Li, J.; Wang, K. Robust genome editing of CRISPR-Cas9 at NAG PAMs in rice. Sci. China Life Sci. 2018, 61, 122–125. [Google Scholar] [CrossRef]
  64. Puchta, H.; Fauser, F. Double-strand break repair and its application to genome engineering in plants. In Advances in New Technology for Targeted Modification of Plant Genomes; Zhang, F., Ed.; Springer: New York, NY, USA, 2015; pp. 1–20. [Google Scholar]
  65. White, M.F.; Allers, T. DNA repair in the archaea—An emerging picture. FEMS Microbiol. Rev. 2018, 42, 514–526. [Google Scholar] [CrossRef] [Green Version]
  66. Zhao, Y.; Zhang, C.; Liu, W.; Gao, W.; Liu, C.; Song, G.; Li, W.-X.; Mao, L.; Chen, B.; Xu, Y.; et al. An alternative strategy for targeted gene replacement in plants using a dual-sgRNA/Cas9 design. Sci. Rep. 2016, 6, 23890. [Google Scholar] [CrossRef]
  67. Lu, Y.; Tian, Y.; Shen, R.; Yao, Q.; Wang, M.; Chen, M.; Dong, J.; Zhang, T.; Li, F.; Lei, M.; et al. Targeted, efficient sequence insertion and replacement in rice. Nat. Biotechnol. 2020, 38, 1402–1407. [Google Scholar] [CrossRef] [PubMed]
  68. Hu, Y.; Li, W. Development and application of CRISPR-Cas based tools. Front. Cell Dev. Biol. 2022, 10, 834646. [Google Scholar] [CrossRef] [PubMed]
  69. Zong, Y.; Wang, Y.; Li, C.; Zhang, R.; Chen, K.; Ran, Y.; Qiu, J.-L.; Wang, D.; Gao, C. Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 438–440. [Google Scholar] [CrossRef]
  70. Huang, T.P.; Newby, G.A.; Liu, D.R. Precision genome editing using cytosine and adenine base editors in mammalian cells. Nat. Prot. 2021, 16, 1089–1128. [Google Scholar] [CrossRef]
  71. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157. [Google Scholar] [CrossRef]
  72. Lin, Q.; Zong, Y.; Wue, C.; Wang, S.; Jin, S.; Zhu, Z.; Wang, Y.; Anzalone, A.V.; Raguram, A.; Doman, J.L.; et al. Prime genome editing in rice and wheat. Nat. Biotechnol. 2020, 38, 582–585. [Google Scholar] [CrossRef] [PubMed]
  73. Hilton, I.B.; D’Ippolito, A.M.; Vockley, C.M.; Thakore, P.I.; Crawford, G.E.; Reddy, T.E.; Gersbach, C.A. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 2015, 33, 510–517. [Google Scholar] [CrossRef] [Green Version]
  74. Devesa-Guerra, I.; Morales-Ruiz, T.; Pérez-Roldán, J.; Parrilla-Doblas, J.T.; Dorado-León, M.; García-Ortiz, M.V.; Ariza, R.R.; Roldán-Arjona, T. DNA methylation editing by CRISPR-guided excision of 5-methylcytosine. J. Mol. Biol. 2020, 432, 2204–2216. [Google Scholar] [CrossRef] [PubMed]
  75. Li, Z.; Wang, F.; Li, J.F. Targeted transcriptional activation in plants using a potent dead Cas9-derived synthetic gene activator. Curr. Prot. 2019, 127, e89. [Google Scholar] [CrossRef]
  76. Zheng, Y.; Su, T.; Qi, Q. Microbial CRISPRi and CRISPRa systems for metabolic engineering. Biotechnol. Bioprocess Eng. 2019, 24, 579–591. [Google Scholar] [CrossRef]
  77. Zhan, X.; Zhang, F.; Zhong, Z.; Chen, R.; Wang, Y.; Chang, L.; Bock, R.; Nie, B.; Zhang, J. Generation of virus-resistant potato plants by RNA genome targeting. Plant Biotechnol. J. 2019, 17, 1814–1822. [Google Scholar] [CrossRef] [Green Version]
  78. Lin, W.; Liu, Y.; Chemparathy, A.; Pande, T.; La Russa, M.; Qi, L.S. A comprehensive analysis and resource to use CRISPR-Cas13 for broad-spectrum targeting of RNA viruses. Cell Rep. Med. 2021, 2, 100245. [Google Scholar] [CrossRef] [PubMed]
  79. Sprink, T.; Wilhelm, R.; Hartung, F. Genome editing around the globe: An update on policies and perceptions. Plant Physiol. 2022, 190, 1579–1587. [Google Scholar] [CrossRef] [PubMed]
  80. Zhang, Y.; Restall, J.; Crisp, P.; Godwin, I.; Liu, G. Current status and prospects of plant genome editing in Australia. Vitro Cell. Dev. Biol. Plant 2021, 57, 574–583. [Google Scholar] [CrossRef] [PubMed]
  81. Casler, M.D.; van Santen, E. Breeding objectives in forages. In Fodder Crops and Amenity Grasses. Handbook of Plant Breeding; Boller, B., Ed.; Springer: New York, NY, USA, 2010; Volume 5, pp. 115–136. [Google Scholar] [CrossRef]
  82. Chen, L.; Auh, C.-K.; Dowling, P.; Bell, J.; Chen, F.; Hopkins, A.; Dixon, R.A.; Wang, Z.-Y. Improved forage digestibility of tall fescue (Festuca arundinacea) by transgenic down-regulation of cinnamyl alcohol dehydrogenase. Plant Biotechnol. J. 2003, 1, 437–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tang, L.; Cai, H.; Zhai, H.; Luo, X.; Wang, Z.; Cui, L.; Bai, X. Overexpression of Glycine soja WRKY20 enhances both drought and salt tolerance in transgenic alfalfa (Medicago sativa L.). Plant Cell Tiss. Org. Cult. 2014, 118, 77–86. [Google Scholar] [CrossRef]
  84. Arshad, M.; Feyissa, B.A.; Amyot, L.; Aung, B.; Hannoufa, A. MicroRNA156 improves drought stress tolerance in alfalfa (Medicago sativa) by silencing SPL13. Plant Sci. 2017, 258, 122–136. [Google Scholar] [CrossRef] [PubMed]
  85. Singer, S.D.; Weselake, R.J.; Acharya, S. Molecular enhancement of alfalfa: Improving quality traits for superior livestock performance and reduced environmental impact. Crop Sci. 2018, 58, 55–71. [Google Scholar] [CrossRef]
  86. Zhang, Y.; Ran, Y.; Nagy, I.; Lenk, I.; Qiu, J.-L.; Asp, T.; Jensen, C.S.; Gao, C. Targeted mutagenesis in ryegrass (Lolium spp.) using the CRISPR/Cas9 system. Plant Biotechnol. J. 2020, 18, 1854–1856. [Google Scholar] [CrossRef]
  87. Dinkins, R.D.; Hancock, J.; Coe, B.L.; May, J.B.; Goodman, J.P.; Bass, W.T.; Liu, J.; Fan, Y.; Zheng, Q.; Zhu, H. Isoflavone levels, nodulation and gene expression profiles of a CRISPR/Cas9 deletion mutant in the isoflavone synthase gene of red clover. Plant Cell Rep. 2021, 40, 517–528. [Google Scholar] [CrossRef]
  88. Singer, S.D.; Burton Hughes, K.; Subedi, U.; Dhariwal, G.K.; Kader, K.; Acharya, S.; Chen, G.; Hannoufa, A. The CRISPR/Cas9-mediated modulation of SQUAMOSA PROMOTER-BINDING PROTEIN-LIKE 8 in alfalfa leads to distinct phenotypic outcomes. Front. Plant Sci. 2022, 12, 774146. [Google Scholar] [CrossRef]
  89. Rodríguez-Leal, D.; Lemmon, Z.H.; Man, J.; Bartlett, M.E.; Lippman, Z.B. Engineering quantitative trait variation for crop improvement by genome editing. Cell 2017, 171, 470–480. [Google Scholar] [CrossRef] [Green Version]
  90. Zhang, H.; Si, X.; Ji, X.; Fan, R.; Liu, J.; Chen, K.; Wang, D.; Gao, C. Genome editing of upstream open reading frames enables translational control in plants. Nat. Biotechnol. 2018, 36, 894–898. [Google Scholar] [CrossRef]
  91. Beauchemin, K.A.; McGinn, S.M.; Benchaar, C.; Holtshausen, L. Crushed sunflower, flax, or canola seeds in lactating dairy cow diets: Effects on methane production, rumen fermentation, and milk production. J. Dairy Sci. 2009, 92, 2118–2127. [Google Scholar] [CrossRef]
  92. Grainger, C.; Beauchemin, K.A. Can enteric methane emissions from ruminants be lowered without lowering their production? Anim. Feed Sci. Technol. 2011, 166–167, 308–320. [Google Scholar] [CrossRef]
  93. Nogueira, R.G.S.; Perna, F.; Pereira, A.S.C.; Cassiano, E.C.O.; Carvalho, R.F.; Rodrigues, P.H.M. Methane mitigation and ruminal fermentation changes in cows fed cottonseed and vitamin E. Sci. Agrícola 2020, 77, e20180247. [Google Scholar] [CrossRef]
  94. Vargas, J.E.; Andrés, S.; López-Ferreras, L.; Snelling, T.J.; Yáñez-Ruíz, D.R.; García-Estrada, C.; López, S. Dietary supplemental plant oils reduce methanogenesis from anaerobic microbial fermentation in the rumen. Sci. Rep. 2020, 10, 1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Muñoz, C.; Villalobos, R.; Peralta, A.M.T.; Morales, R.; Urrutia, N.L.; Ungerfield, E.M. Long-term and carryover effects of supplementation with whole oilseeds on methane emission, milk production and milk fatty acid profile of grazing dairy cows. Animals 2021, 11, 2978. [Google Scholar] [CrossRef]
  96. Beauchemin, K.A.; Kreuzer, M.; O’mara, F.; McAllister, T.A. Nutritional management for enteric methane abatement: A review. Aust. J. Exp. Agric. 2008, 48, 21–27. [Google Scholar] [CrossRef]
  97. Eugène, M.; Massé, D.; Chiquette, J.; Benchaar, C. Meta-analysis on the effects of lipid supplementation on methane production in lactating dairy cows. Can. J. Anim. Sci. 2008, 88, 331–337. [Google Scholar] [CrossRef]
  98. Bayat, A.R.; Tapio, I.; Vilkki, J.; Shingfield, K.J.; Leskinen, H. Plant oil supplements reduce methane emissions and improve milk fatty acid composition in dairy cows fed grass silage-based diets without affecting milk yield. J. Dairy Sci. 2018, 101, 1136–1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Boufaïed, H.; Chouinard, P.Y.; Tremblay, G.F.; Petit, H.V.; Michaud, R.; Bélanger, G. Fatty acids in forages. I. Factors affecting concentrations. Can. J. Anim. Sci. 2003, 83, 501–511. [Google Scholar] [CrossRef]
  100. Dewhurst, R.J.; Scollan, N.D.; Lee, M.R.F.; Ougham, H.J.; Humphreys, M.O. Forage breeding and management to increase the beneficial fatty acid content of ruminant products. Proc. Nutr. Soc. 2003, 62, 329–336. [Google Scholar] [CrossRef]
  101. Clapham, W.M.; Foster, J.G.; Neel, J.P.S.; Fedders, J.M. Fatty acid composition of traditional and novel forages. J. Agric. Food Chem. 2005, 53, 10068–10073. [Google Scholar] [CrossRef] [PubMed]
  102. Mel’uchová, B.; Blaško, J.; Kubinec, R.; Górová, R.; Dubravská, J.; Margetín, M.; Soják, L. Seasonal variations in fatty acid composition of pasture forage plants and CLA content in ewe milk fat. Small Rumin. Res. 2008, 78, 56–65. [Google Scholar] [CrossRef]
  103. Glasser, F.; Doreau, M.; Maxin, G.; Baumont, R. Fat and fatty acid content and composition of forages: A meta-analysis. Anim. Feed Sci. Technol. 2013, 185, 19–34. [Google Scholar] [CrossRef]
  104. Henderson, B.; Falcucci, A.; Mottet, A.; Early, L.; Werner, B.; Steinfeld, H.; Gerber, P. Marginal costs of abating greenhouse gases in the global ruminant livestock sector. Mitig. Adapt. Strateg. Glob. Chang. 2017, 22, 199–224. [Google Scholar] [CrossRef]
  105. Moate, P.J.; Williams, S.R.O.; Grainger, C.; Hannah, M.C.; Ponnampalam, E.N.; Eckard, R.J. Influence of cold-pressed canola, brewers grains and hominy meal as dietary supplements suitable for reducing enteric methane emissions from lactating dairy cows. Anim. Feed. Sci. Technol. 2011, 166–167, 254–264. [Google Scholar] [CrossRef]
  106. Li-Beisson, Y.; Shorrosh, B.; Beisson, F.; Andersson, M.X.; Arondel, V.; Bates, P.D.; Baud, S.; Bird, D.; DeBono, A.; Durrett, T.P.; et al. Acyl-lipid metabolism. Arab. Book 2013, 11, e0161. [Google Scholar] [CrossRef]
  107. Reszczyńska, E.; Hanaka, A. Lipids composition in plant membranes. Cell Biochem. Biophys. 2020, 78, 401–414. [Google Scholar] [CrossRef]
  108. Yang, Z.; Ohlrogge, J.B. Turnover of fatty acids during natural leaf senescence of Arabidopsis, Brachypodium, and switchgrass and in Arabidopsis β-oxidation mutants. Plant Physiol. 2009, 150, 1981–1989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Fan, J.; Yu, L.; Xu, C. A central role for triacylglycerol in membrane lipid breakdown, fatty Acid β-oxidation, and plant survival under extended darkness. Plant Physiol. 2017, 174, 1517–1530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Lu, J.; Xu, Y.; Wang, J.; Singer, S.D.; Chen, G. The role of triacylglycerol in plant stress response. Plants 2020, 9, 472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Lin, W.; Oliver, D.J. Role of triacylglycerol in leaves. Plant Sci. 2008, 175, 233–237. [Google Scholar] [CrossRef]
  112. Ohlrogge, J.B.; Jaworski, J.G.; Post-Beittenmiller, D. De novo fatty acid biosynthesis. In Lipid Metabolism in Plants; Moore, T.S., Ed.; CRC Press: Boca Raton, FL, USA, 1993; pp. 4–32. [Google Scholar]
  113. He, M.; Qin, C.-W.; Wang, X.; Ding, N.-Z. Plant unsaturated fatty acids: Biosynthesis and regulation. Front. Plant Sci. 2020, 11, 390. [Google Scholar] [CrossRef] [Green Version]
  114. Jayawardhane, K.N.; Singer, S.D.; Weselake, R.J.; Chen, G. Plant sn-glycerol-3-phosphate acyltransferases: Biocatalysts involved in the biosynthesis of intracellular and extracellular lipids. Lipids 2018, 53, 469–480. [Google Scholar] [CrossRef]
  115. Wijekoon, C.P.; Singer, S.D.; Weselake, R.J.; Petrie, J.R.; Singh, S.; Jayawardhane, K.N.; Shah, S.; Chen, G.; Eastmond, P.J.; Acharya, S.N. Enhancement of total lipid production in vegetative tissues of alfalfa and sainfoin using chemical mutagenesis. Crop Sci. 2020, 60, 2990–3003. [Google Scholar] [CrossRef]
  116. Vanhercke, T.; Divi, U.K.; El Tahchy, A.; Liu, Q.; Mitchell, M.; Taylor, M.C. Step changes in leaf oil accumulation via iterative metabolic engineering. Metabol. Eng. 2017, 39, 237–246. [Google Scholar] [CrossRef]
  117. National Research Council (NRC). Nutrient Requirements of Dairy Cattle, 7th Revised ed.; National Academy Press: Washington, DC, USA, 2001. [Google Scholar]
  118. Liu, Y.-F.; Li, Z.-T.; Lu, X.; Song, Q.-X.; Lam, S.-M.; Zhang, W.-K.; Ma, B.; Lin, Q.; Man, W.-Q.; Du, W.-G.; et al. Soybean GmMYB73 promotes lipid accumulation in transgenic plants. BMC Plant Biol. 2014, 14, 73. [Google Scholar] [CrossRef]
  119. Beechey-Gradwell, Z.D.; Winichayakul, S.; Roberts, N.J. High lipid perennial ryegrass growth under variable nitrogen, water and carbon dioxide supply. J. N. Zeal. Grass. 2018, 80, 219–224. [Google Scholar] [CrossRef]
  120. Vanhercke, T.; Belide, S.; Taylor, M.C.; El Tahchy, A.; Okada, S.; Rolland, V.; Liu, Q.; Mitchell, M.; Shrestha, P.; Venables, I.; et al. Up-regulation of lipid biosynthesis increases the oil content in leaves of Sorghum bicolor. Plant Biotechnol. J. 2019, 17, 220–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Winichayakul, S.; Beechey-Gradwell, Z.; Muetzel, S.; Molano, G.; Crowther, T.; Lewis, S.; Xue, H.; Burke, J.; Bryan, G.; Roberts, N.J. In vitro gas production and rumen fermentation profile of fresh and ensiled genetically modified high-metabolizable energy ryegrass. J. Dairy Sci. 2020, 103, 2405–2418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Subedi, U.; Jayawardhane, K.N.; Pan, X.; Ozga, J.; Chen, G.; Foroud, N.A.; Singer, S.D. The potential of genome editing for improving seed oil content and fatty acid composition in oilseed crops. Lipids 2020, 55, 495–512. [Google Scholar] [CrossRef] [PubMed]
  123. Tian, Y.; Liu, X.; Fan, C.; Li, T.; Qin, H.; Li, X.; Chen, K.; Zheng, Y.; Cheng, F.; Xu, Y. Enhancement of tobacco (Nicotiana tabacum L.) seed lipid content for biodiesel production by CRISPR-Cas9-mediated knockout of NtAn1. Front. Plant Sci. 2021, 11, 599474. [Google Scholar] [CrossRef]
  124. James, C.N.; Horn, P.J.; Case, C.R.; Gidda, S.K.; Zhang, D.; Mullen, R.T.; Dyer, J.M.; Anderson, R.G.W.; Chapman, K.D. Disruption of the Arabidopsis CGI-58 homologue produces Chanarin—Dorfman-like lipid droplet accumulation in plants. Proc. Natl. Acad. Sci. USA 2010, 107, 17833–17838. [Google Scholar] [CrossRef] [Green Version]
  125. Kelly, A.A.; van Erp, H.; Quettier, A.L.; Shaw, E.; Menard, G.; Kurup, S.; Eastmond, P.J. The SUGAR-DEPENDENT1 lipase limits triacylglycerol accumulation in vegetative tissues of Arabidopsis. Plant Physiol. 2013, 162, 1282–1289. [Google Scholar] [CrossRef] [Green Version]
  126. Wijekoon, C.; Singer, S.D.; Weselake, R.J.; Petrie, J.R.; Chen, G.; Singh, S.; Eastmond, P.J.; Acharya, S.N. Down-regulation of key genes involved in carbon metabolism in Medicago truncatula results in increased lipid accumulation in vegetative tissue. Crop Sci. 2020, 60, 1798–1808. [Google Scholar] [CrossRef]
  127. Zhai, Z.; Liu, H.; Xu, C.; Shanklin, J. Sugar potentiation of fatty acid and triacylglycerol accumulation. Plant Physiol. 2017, 175, 696–707. [Google Scholar] [CrossRef] [Green Version]
  128. Fan, J.; Zhai, Z.; Yan, C.; Xu, C. Arabidopsis TRIGALACTOSYLDIACYLGLYCEROL5 interacts with TGD1, TGD2, and TGD4 to facilitate lipid transfer from the endoplasmic reticulum to plastids. Plant Cell 2015, 27, 2941–2955. [Google Scholar] [CrossRef]
  129. Fan, J.; Yan, C.; Xu, C. Phospholipid: Diacylglycerol acyltransferase-mediated triacylglycerol biosynthesis is crucial for protection against fatty acid-induced cell death in growing tissues of Arabidopsis. Plant J. 2013, 76, 930–942. [Google Scholar] [CrossRef]
  130. Xu, C.; Fan, J.; Froehlich, J.E.; Awai, K.; Benning, C. Mutation of the TGD1 chloroplast envelope protein affects phosphatidate metabolism in Arabidopsis. Plant Cell 2005, 17, 3094–3110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Jarvis, P.; Dörmann, P.; Peto, C.A.; Lutes, J.; Benning, C.; Chory, J. Galactolipid deficiency and abnormal chloroplast development in the Arabidopsis MGD synthase 1 mutant. Proc. Natl. Acad. Sci. USA 2000, 97, 8175–8179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wu, H.Y.; Liu, C.; Li, M.C.; Zhao, M.M.; Gu, D.; Xu, Y.N. Effects of monogalactoglycerolipid deficiency and diacylglycerol acyltransferase overexpression on oil accumulation in transgenic tobacco. Plant Mol. Biol. Rep. 2013, 31, 1077–1088. [Google Scholar] [CrossRef]
  133. Yu, T.-S.; Lue, W.-L.; Wang, S.-M.; Chen, J. Mutation of Arabidopsis plastid phosphoglucose isomerase affects leaf starch synthesis and floral initiation. Plant Physiol. 2000, 123, 319–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Sanjaya; Durrett, T.P.; Weise, S.E.; Benning, C. Increasing the energy density of vegetative tissues by diverting carbon from starch to oil biosynthesis in transgenic Arabidopsis. Plant Biotechnol. J. 2011, 9, 874–883. [Google Scholar] [CrossRef]
  135. Lin, T.-P.; Caspar, T.; Somerville, C.; Preiss, J. Isolation and characterization of a starchless mutant of Arabidopsis thaliana (L.) Heynh lacking ADPglucose pyrophosphorylase activity. Plant Physiol. 1988, 86, 1131–1135. [Google Scholar] [CrossRef] [Green Version]
  136. Müller-Röber, B.; Sonnewald, U.; Willmitzer, L. Inhibition of the ADP-glucose pyrophosphorylase in transgenic potatoes leads to sugar-storing tubers and influences tuber formation and expression of tuber storage protein genes. EMBO J. 1992, 11, 1229–1238. [Google Scholar] [CrossRef]
  137. Rösti, S.; Rudi, H.; Rudi, K.; Opsahl-Sorteberg, H.-G.; Fahy, B.; Denyer, K. The gene encoding the cytosolic small subunit of ADP-glucose pyrophosphorylase in barley endosperm also encodes the major plastidial small subunit in the leaves. J. Exp. Bot. 2006, 57, 3619–3626. [Google Scholar] [CrossRef] [Green Version]
  138. Lee, S.-K.; Hwang, S.-K.; Han, M.; Eom, J.-S.; Kang, H.-G.; Han, Y.; Choi, S.-B.; Cho, M.-H.; Bhoo, S.H.; An, G.; et al. Identification of the ADP-glucose pyrophosphorylase isoforms essential for starch synthesis in the leaf and seed endosperm of rice (Oryza sativa L.). Plant Mol. Biol. 2007, 65, 531–546. [Google Scholar] [CrossRef]
  139. Rösti, S.; Fahy, B.; Denyer, K. A mutant of rice lacking the leaf large subunit of ADP-glucose pyrophosphorylase has drastically reduced leaf starch content but grows normally. Funct. Plant Biol. 2007, 34, 480–489. [Google Scholar] [CrossRef]
  140. Cossegal, M.; Chambrier, P.; Mbelo, S.; Balzergue, S.; Martin-Magniette, M.-L.; Moing, A.; Deborde, C.; Guyon, V.; Perez, P.; Rogowsky, P. Transcriptional and metabolic adjustments in ADP-glucuse pyrophosphorylase-deficient bt2 maize kernels. Plant Physiol. 2008, 146, 1553–1570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Slewinski, T.L.; Ma, Y.; Baker, F.; Huang, M.; Meeley, R.; Braun, D.M. Determining the role of Tie-dyed1 in starch metabolism: Epistasis analysis with a maize ADP-glucose pyrophosphorylase mutant lacking leaf starch. J. Hered. 2008, 99, 661–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Yu, L.; Fan, J.; Yan, C.; Xu, C. Starch deficiency enhances lipid biosynthesis and turnover in leaves. Plant Physiol. 2018, 178, 118–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Khorrami, B.; Khiaosa-ard, R.; Zebeli, Q. Models to predict the risk of subacute ruminal acidosis in dairy cows based on dietary and cow factors: A meta-analysis. J. Dairy Sci. 2021, 104, 7761–7780. [Google Scholar] [CrossRef] [PubMed]
  144. Rivero, M.J.; Keim, J.P.; Balocchi, O.A.; Lee, M.R.F. In vitro fermentation patterns and methane output of perennial ryegrass differing in water-soluble carbohydrate and nitrogen concentrations. Animals 2020, 10, 1076. [Google Scholar] [CrossRef] [PubMed]
  145. Zale, J.; Jung, J.H.; Kim, J.Y.; Pathak, B.; Karan, R.; Liu, H.; Chen, X.; Wu, H.; Candreva, J.; Zhai, Z.; et al. Metabolic engineering of sugarcane to accumulate energy-dense triacylglycerols in vegetative biomass. Plant Biotechnol. J. 2016, 14, 661–669. [Google Scholar] [CrossRef] [PubMed]
  146. Eastmond, P.J. SUGAR-DEPENDENT1 encodes a patatin domain triacylglycerol lipase that initiates storage oil breakdown in germinating Arabidopsis seeds. Plant Cell 2006, 18, 665–675. [Google Scholar] [CrossRef] [Green Version]
  147. Park, S.; Gidda, S.K.; James, C.N.; Horn, P.J.; Khuu, N.; Seay, D.C.; Keereetaweep, J.; Chapman, K.D.; Mullen, R.T.; Dyer, J.M. The α/β hydrolase CGI-58 and peroxisomal transport protein PXA1 coregulate lipid homeostasis and signaling in Arabidopsis. Plant Cell 2013, 25, 1726–1739. [Google Scholar] [CrossRef] [Green Version]
  148. Kelly, A.A.; Quettier, A.-L.; Shaw, E.; Eastmond, P.J. Seed storage oil mobilization is important but not essential for germination or seedling establishment in Arabidopsis. Plant Physiol. 2011, 157, 866–875. [Google Scholar] [CrossRef]
  149. Xu, X.; Vanhercke, T.; Shrestha, P.; Luo, J.; Akbar, S.; Konik-Rose, C.; Venugoban, L.; Hussain, D.; Tian, L.; Singh, S.; et al. Upregulated lipid biosynthesis at the expense of starch production in potato (Solanum tuberosum) vegetative tissues via simultaneous downregulation of ADP-glucose pyrophosphorylase and sugar dependent1 expressions. Front. Plant Sci. 2019, 10, 1444. [Google Scholar] [CrossRef] [Green Version]
  150. Bhunia, R.K.; Medard, G.N.; Eastmond, P.J. A native promoter-gene fusion created by CRISPR/Cas9-mediated genomic deletion offers a transgene-free method to drive oil accumulation in leaves. FEBS Lett. 2022, 596, 1865–1870. [Google Scholar] [CrossRef] [PubMed]
  151. Chen, G.; Harwood, J.L.; Lemieux, M.J.; Stone, S.J.; Weselake, R.J. Acyl-CoA: Diacylglycerol acyltransferase: Properties, physiological roles, metabolic engineering and intentional control. Prog. Lipid Res. 2022, 88, 101181. [Google Scholar] [CrossRef] [PubMed]
  152. Boadi, D.; Benchaar, C.; Chiquett, J.; Massé, D. Mitigation strategies to reduce enteric methane emissions from dairy cows: Update review. Can. J. Anim. Sci. 2004, 84, 319–335. [Google Scholar] [CrossRef] [Green Version]
  153. Haque, M.N. Dietary manipulation: A sustainable way to mitigate methane emissions from ruminants. J. Anim. Sci. Technol. 2018, 60, 15. [Google Scholar] [CrossRef] [Green Version]
  154. Verma, S.; Taube, F.; Malisch, C.S. Examining the variables leading to apparent incongruity between antimethanogenic potential of tannins and their observed effects in ruminants—A review. Sustainability 2021, 13, 2743. [Google Scholar] [CrossRef]
  155. Chung, Y.H.; McGeough, E.J.; Acharya, S.; McAllister, T.A.; McGinn, S.M.; Harstad, O.M.; Beauchemin, K.A. Enteric methane emission, diet digestibility, and nitrogen excretion from beef heifers fed sainfoin or alfalfa. J. Anim. Sci. 2013, 91, 4861–4874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Huyen, N.T.; Desrues, O.; Alferink, S.J.J.; Zandstra, T.; Verstegen, M.W.A.; Hendriks, W.H.; Pellikaan, W.F. Inclusion of sainfoin (Onobrychis viciifolia) silage in dairy cow rations affects nutrient digestibility, nitrogen utilization, energy balance, and methane emissions. J. Dairy Sci. 2016, 99, 3566–3577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Tavendale, M.H.; Meagher, L.P.; Pacheco, D.; Walker, N.; Attwood, G.T.; Sivakumaran, S. Methane production from in vitro rumen incubations with Lotus pedunculatus and Medicago sativa, and effects of extractable condensed tannin fractions on methanogenesis. Anim. Feed Sci. Technol. 2005, 123–124, 403–419. [Google Scholar] [CrossRef]
  158. Naumann, H.D.; Tedeschi, L.O.; Zeller, W.E.; Huntley, N.F. The role of condensed tannins in ruminant animal production: Advances, limitations and future directions. R. Bras. Zootec. 2017, 46, 929–949. [Google Scholar] [CrossRef]
  159. Hatew, B.; Carbonero, C.H.; Stringano, E.; Sales, L.F.; Smith, L.M.J.; Mueller-Harvey, I.; Hendriks, W.H.; Pellikaan, W.F. Diversity of condensed tannin structures affects rumen in vitro methane production in sainfoin (Onobrychis viciifolia) accessions. Grass Forage Sci. 2015, 70, 474–490. [Google Scholar] [CrossRef]
  160. Naumann, H.; Sepela, R.; Rezaire, A.; Masih, S.E.; Zeller, W.E.; Reinhardt, L.A.; Robe, J.T.; Sullivan, M.L.; Hagerman, A.E. Relationships between structures of condensed tannins from Texas legumes and methane production during in vitro rumen digestion. Molecules 2018, 23, 2123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Aboagye, A.; Beauchemin, K.A. Potential of molecular weight and structure of tannins to reduce methane emissions from ruminants: A review. Animals 2019, 9, 856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Jayanegara, A.; Marquardt, S.; Wina, E.; Kreuzer, M.; Leiber, F. In vitro indications for favourable non-additive effects on ruminal methane mitigation between high-phenolic and high-quality forages. Brit. J. Nutr. 2013, 109, 615–622. [Google Scholar] [CrossRef] [Green Version]
  163. Eugène, M.; Klumpp, K.; Sauvant, D. Methane mitigating options with forages fed to ruminants. Grass Forage Sci. 2021, 76, 196–204. [Google Scholar] [CrossRef]
  164. Van Gastelen, S.; Dijkstra, J.; Bannink, A. Are dietary strategies to mitigate enteric methane emission equally effective across dairy cattle, beef cattle, and sheep? J. Dairy Sci. 2019, 102, 6109–6130. [Google Scholar] [CrossRef] [Green Version]
  165. Nayak, D.D.; Metcalf, W.W. Cas9-mediated genome editing in the methanogenic archaeon Methanosarcina acetivorans. Proc. Natl. Acad. Sci. USA 2017, 114, 2976–2981. [Google Scholar] [CrossRef] [Green Version]
  166. Bartlett, E.J.; Brissett, N.C.; Doherty, A.J. Ribonucleolytic resection is required for repair of strand displaced nonhomologous end-joining intermediates. Proc. Natl. Acad. Sci. USA 2013, 110, E1984–E1991. [Google Scholar] [CrossRef] [Green Version]
  167. Li, Y.; Peng, N. Endogenous CRISPR-Cas system-based genome editing and antimicrobials: Review and prospects. Front. Microbiol. 2019, 10, 2471. [Google Scholar] [CrossRef] [Green Version]
  168. Li, Y.; Pan, S.; Zhang, Y.; Ren, M.; Feng, M.; Peng, N.; Chen, L.; Liang, Y.X.; She, Q. Harnessing type I and type III CRISPR-Cas systems for genome editing. Nucleic Acids Res. 2016, 44, e34. [Google Scholar] [CrossRef] [PubMed]
  169. Khelaifia, S.; Garibal, M.; Robert, C.; Raoult, D.; Drancourt, M. Draft genome sequence of a human-associated isolate of Methanobrevibacter arboriphilicus, the lowest-G + C-content Archaeon. Genome Announc. 2014, 2, e01181-13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Khelaifia, S.; Garibal, M.; Robert, C.; Raoult, D.; Drancourt, M. Draft genome sequencing of Methanobrevibacter oralis strain JMR01, isolated from the human intestinal microbiota. Genome Announc. 2014, 2, e00073-14. [Google Scholar] [CrossRef] [PubMed]
  171. Weimer, P.J. Redundancy, resilience, and host specificity of the ruminal microbiota: Implications for engineering improved ruminal fermentations. Front. Microbiol. 2015, 6, 296. [Google Scholar] [CrossRef] [Green Version]
  172. Kamke, J.; Kittelmann, S.; Soni, P.; Li, Y.; Tavendale, M.; Ganesh, S.; Janssen, P.H.; Shi, W.; Froula, J.; Rubin, E.M.; et al. Rumen metagenome and metatranscriptome analyses of low methane yield sheep reveals a Sharpea-enriched microbiome characterised by lactic acid formation and utilisation. Microbiome 2016, 4, 56. [Google Scholar] [CrossRef] [Green Version]
  173. Auffret, M.D.; Stewart, R.D.; Dewhurst, R.J.; Duthie, C.-A.; Watson, M.; Roehe, R. Identification of microbial genetic capacities and potential mechanisms within the rumen microbiome explaining differences in beef cattle feed efficiency. Front. Microbiol. 2020, 11, 1229. [Google Scholar] [CrossRef] [PubMed]
  174. Newbold, D.J.; Ramos-Morales, E. Ruminal microbiome and microbial metabolome: Effects of diet and ruminant host. Animal 2020, 14, s78–s86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Sun, H.-Z.; Peng, K.-L.; Xue, M.-Y.; Liu, J.-X. Metagenomics analysis revealed the distinctive ruminal microbiome and resistive profiles in dairy buffaloes. Anim. Microbiome 2021, 3, 44. [Google Scholar] [CrossRef]
  176. Henderson, G.; Cox, F.; Ganesh, S.; Jonker, A.; Young, W.; Global Rumen Census Collaborators; Janssen, P.H. Rumen microbial community composition varies with diet and host, but a core microbiome is found across a wide geographical range. Sci. Rep. 2015, 5, 14567. [Google Scholar] [CrossRef] [Green Version]
  177. Islam, M.; Lee, S.S. Advanced estimation and mitigation strategies: A cumulative approach to enteric methane abatement from ruminants. J. Anim. Sci. Technol. 2019, 61, 122–137. [Google Scholar] [CrossRef] [Green Version]
  178. Janssen, P.H.; Kirs, M. Structure of the archaeal community of the rumen. Appl. Environ. Microbiol. 2008, 74, 3619–3625. [Google Scholar] [CrossRef] [PubMed]
  179. Patra, A.; Park, T.; Kim, M.; Yu, Z. Rumen methanogens and mitigation of methane emission by anti-methanogenic compounds and substances. J. Anim. Sci. Biotechnol. 2017, 8, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Stams, A.J.; Plugge, C.M. Electron transfer in syntrophic communities of anaerobic bacteria and archaea. Nat. Rev. Microbiol. 2009, 7, 568–577. [Google Scholar] [CrossRef] [PubMed]
  181. Difford, G.F.; Plichta, D.R.; Løvendahl, P.; Lassen, J.; Noel, S.J.; Højberg, O.; Wright, A.G.; Zhu, Z.; Kristensen, L.; Nielsen, H.B.; et al. Host genetics and the rumen microbiome jointly associate with methane emissions in dairy cows. PLoS Genet. 2018, 14, e1007580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Sasson, G.; Kruger Ben-Shabat, S.; Seroussi, E.; Doron-Faigenboim, A.; Shterzer, N.; Yaacoby, S.; Berg Miller, M.E.; White, B.A.; Halperin, E.; Mizrahi, I. Heritable bovine rumen bacteria are phylogenetically related and correlated with the cow’s capacity to harvest energy from its feed. mBio 2017, 8, e00703–e00717. [Google Scholar] [CrossRef] [Green Version]
  183. Abbas, W.; Howard, J.T.; Paz, H.A.; Hales, K.E.; Wells, J.E.; Kuehn, L.A.; Erickson, G.E.; Spangler, M.L.; Fernando, S.C. Influence of host genetics in shaping the rumen bacterial community in beef cattle. Sci. Rep. 2020, 10, 15101. [Google Scholar] [CrossRef]
  184. Leahy, S.C.; Kelly, W.J.; Altermann, E.; Ronimus, R.S.; Yeoman, C.J.; Pacheco, D.M.; Li, D.; Kong, Z.; McTavish, S.; Sang, C.; et al. The genome sequence of the rumen methanogen Methanobrevibacter ruminantium reveals new possibilities for controlling ruminant methane emissions. PLoS ONE 2010, 5, e8926. [Google Scholar] [CrossRef] [Green Version]
  185. Bharathi, M.; Senthil Kumar, N.; Chellapandi, P. Functional prediction and assignment of Methanobrevibacter ruminantium m1 operome using a combined bioinformatics approach. Front. Genet. 2020, 11, 593990. [Google Scholar] [CrossRef]
  186. Deppenmeier, U. The unique biochemistry of methanogenesis. Prog. Nucleic Acid Res. Mol. Biol. 2002, 71, 223–283. [Google Scholar] [CrossRef]
  187. Ferry, J.G. Fundamentals of methanogenic pathways that are key to the biomethanation of complex biomass. Curr. Opin. Biotechnol. 2011, 22, 351–357. [Google Scholar] [CrossRef] [Green Version]
  188. Shi, W.; Moon, C.D.; Leahy, S.C.; Kang, D.; Froula, J.; Kittelmann, S.; Fan, C.; Deutsch, S.; Gagic, D.; Seedorf, H.; et al. Methane yield phenotypes linked to differential gene expression in the sheep rumen microbiome. Genome Res. 2014, 24, 1517–1525. [Google Scholar] [CrossRef]
  189. Rother, M.; Boccazzi, P.; Bose, A.; Pritchett, M.A.; Metcalf, W.W. Methanol-dependent gene expression demonstrates that methyl-coenzyme M reductase is essential in Methanosarcina acetivorans C2A and allows isolation of mutants with defects in regulation of the methanol utilization pathway. J. Bacteriol. 2005, 187, 5552–5559. [Google Scholar] [CrossRef] [Green Version]
  190. Qi, L.S.; Larson, M.H.; Gilbert, L.A.; Doudna, J.A.; Weissman, J.S.; Arkin, A.P.; Lim, W.A. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013, 152, 1173–1183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Mandegar, M.A.; Huebsch, N.; Frolov, E.B.; Shin, E.; Truong, A.; Olvera, M.P.; Chan, A.H.; Miyaoka, Y.; Holmes, K.; Spencer, C.I.; et al. CRISPR interference efficiently induces specific and reversible gene silencing in human iPSCs. Cell Stem Cell 2016, 8, 541–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Dhamad, A.E.; Lessner, D.J. A CRISPRi-dCas9 system for archaea and its use to examine gene function during nitrogen fixation by Methanosarcina acetivorans. Appl. Environ. Microbiol. 2020, 86, e01402–e01420. [Google Scholar] [CrossRef] [PubMed]
  193. Hale, C.R.; Cocozaki, A.; Li, H.; Terns, R.M.; Terns, M.P. Target RNA capture and cleavage by the Cmr type III-B CRISPR-Cas effector complex. Genes Dev. 2014, 28, 2432–2443. [Google Scholar] [CrossRef] [Green Version]
  194. Zink, I.A.; Wimmer, E.; Schleper, C. Heavily armed ancestors: CRISPR immunity and applications in archaea with a comparative analysis of CRISPR types in Sulfolobales. Biomolecules 2020, 10, 1523. [Google Scholar] [CrossRef] [PubMed]
  195. Zebec, Z.; Manica, A.; Zhang, J.; White, M.F.; Schleper, C. CRISPR-mediated targeted mRNA degradation in the archaeon Sulfolobus solfataricus. Nucleic Acids Res. 2014, 42, 5280–5288. [Google Scholar] [CrossRef] [Green Version]
  196. Zink, I.A.; Fouqueau, T.; Tarrason Risa, G.; Werner, F.; Baum, B.; Bläsi, U.; Schleper, C. Comparative CRISPR type III-based knockdown of essential genes in hyperthermophilic Sulfolobales and the evasion of lethal gene silencing. RNA Biol. 2021, 18, 421–434. [Google Scholar] [CrossRef]
Figure 1. CRISPR/Cas-mediated gene editing platforms achieved through the targeted elicitation of double-stranded DNA breaks and NHEJ or HDR repair. HDR, homology-directed repair; NHEJ, non-homologous end joining; PAM, protospacer adjacent motif; sgRNA, single guide RNA.
Figure 1. CRISPR/Cas-mediated gene editing platforms achieved through the targeted elicitation of double-stranded DNA breaks and NHEJ or HDR repair. HDR, homology-directed repair; NHEJ, non-homologous end joining; PAM, protospacer adjacent motif; sgRNA, single guide RNA.
Agriculture 12 01780 g001
Figure 2. Simplified diagrammatic representation of lipid biosynthesis in the photosynthetic tissues of plants. Prokaryotic and eukaryotic pathways for the synthesis of lipids in photosynthetic tissues are indicated within the plastid and endoplasmic reticulum, respectively. Enzymes that have previously been reported to enhance foliar lipid content when their corresponding genes are knocked-down or knocked-out are indicated on a red background. Known reactions are denoted by solid gray lines, whereas putative events are denoted with dashed lines. Abbreviations: 3PG, 3-phosphoglycerate; ACCase, acetylCoA carboxylase; Acyl-ACP, acyl carrier protein; ADP-glucose, adenosine diphosphate glucose; AGPase, adenosine diphosphate glucose pyrophosphorylase; CGI-58, comparative gene identification-58; CO2, carbon dioxide; DAG, diacylglycerol; DGAT, diacylglycerol acyltransferase; DGDG, digalactosyldiacylglycerol; DHAP, dihydroxyacetone phosphate; F6P, fructose-6-phosphate; FAs, fatty acids; G1P, glucose-1-phosphate; G3P, glyceraldehyde 3-phosphate; G6P, glucose-6-phosphate; GPAT, glycerol-3-phosphate acyltransferase; LPA, lysophosphatidic acid; LPAT, lysophosphatidic acid acyltransferase; LPC, lysophosphatidylcholine; Malonyl-ACP, malonyl acyl carrier protein; MGD1, monogalactosyldiacylglycerol synthase 1; MGDG, monogalactosyldiacylglycerol; PA, phosphatidic acid; PAP, phosphatidic acid phosphatase; PC, phosphatidylcholine; PDAT, phospholipid:diacylglycerol acyltransferase; PG, phosphatidylglycerol; PXA1, peroxisomal ABC-transporter; SDP1, sugar-dependent 1; SQDG, sulfoguinovosyldiacylglycerol; TAG, triacylglycerol; TGD1/5, trigalactosyldiacylglycerol 1/5.
Figure 2. Simplified diagrammatic representation of lipid biosynthesis in the photosynthetic tissues of plants. Prokaryotic and eukaryotic pathways for the synthesis of lipids in photosynthetic tissues are indicated within the plastid and endoplasmic reticulum, respectively. Enzymes that have previously been reported to enhance foliar lipid content when their corresponding genes are knocked-down or knocked-out are indicated on a red background. Known reactions are denoted by solid gray lines, whereas putative events are denoted with dashed lines. Abbreviations: 3PG, 3-phosphoglycerate; ACCase, acetylCoA carboxylase; Acyl-ACP, acyl carrier protein; ADP-glucose, adenosine diphosphate glucose; AGPase, adenosine diphosphate glucose pyrophosphorylase; CGI-58, comparative gene identification-58; CO2, carbon dioxide; DAG, diacylglycerol; DGAT, diacylglycerol acyltransferase; DGDG, digalactosyldiacylglycerol; DHAP, dihydroxyacetone phosphate; F6P, fructose-6-phosphate; FAs, fatty acids; G1P, glucose-1-phosphate; G3P, glyceraldehyde 3-phosphate; G6P, glucose-6-phosphate; GPAT, glycerol-3-phosphate acyltransferase; LPA, lysophosphatidic acid; LPAT, lysophosphatidic acid acyltransferase; LPC, lysophosphatidylcholine; Malonyl-ACP, malonyl acyl carrier protein; MGD1, monogalactosyldiacylglycerol synthase 1; MGDG, monogalactosyldiacylglycerol; PA, phosphatidic acid; PAP, phosphatidic acid phosphatase; PC, phosphatidylcholine; PDAT, phospholipid:diacylglycerol acyltransferase; PG, phosphatidylglycerol; PXA1, peroxisomal ABC-transporter; SDP1, sugar-dependent 1; SQDG, sulfoguinovosyldiacylglycerol; TAG, triacylglycerol; TGD1/5, trigalactosyldiacylglycerol 1/5.
Agriculture 12 01780 g002
Figure 3. Possible in vitro approach for the gene editing of rumen archaea. Methanogenic archaea would be collected from the rumen and cultured in vitro, at which point genes involved in methanogenesis could be knocked down or knocked out using genome editing technologies such as CRISPR/Cas9. The gene-edited archaea would then be replicated in vitro and reintroduced into the rumen for repopulation. As long as the edited archaea were competitive within the rumen population, the expected result would be a reduction in the production of CH4 from the ruminant.
Figure 3. Possible in vitro approach for the gene editing of rumen archaea. Methanogenic archaea would be collected from the rumen and cultured in vitro, at which point genes involved in methanogenesis could be knocked down or knocked out using genome editing technologies such as CRISPR/Cas9. The gene-edited archaea would then be replicated in vitro and reintroduced into the rumen for repopulation. As long as the edited archaea were competitive within the rumen population, the expected result would be a reduction in the production of CH4 from the ruminant.
Agriculture 12 01780 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Subedi, U.; Kader, K.; Jayawardhane, K.N.; Poudel, H.; Chen, G.; Acharya, S.; Camargo, L.S.A.; Bittencourt, D.M.d.C.; Singer, S.D. The Potential of Novel Gene Editing-Based Approaches in Forages and Rumen Archaea for Reducing Livestock Methane Emissions. Agriculture 2022, 12, 1780. https://doi.org/10.3390/agriculture12111780

AMA Style

Subedi U, Kader K, Jayawardhane KN, Poudel H, Chen G, Acharya S, Camargo LSA, Bittencourt DMdC, Singer SD. The Potential of Novel Gene Editing-Based Approaches in Forages and Rumen Archaea for Reducing Livestock Methane Emissions. Agriculture. 2022; 12(11):1780. https://doi.org/10.3390/agriculture12111780

Chicago/Turabian Style

Subedi, Udaya, Kazi Kader, Kethmi N. Jayawardhane, Hari Poudel, Guanqun Chen, Surya Acharya, Luiz S. A. Camargo, Daniela Matias de C. Bittencourt, and Stacy D. Singer. 2022. "The Potential of Novel Gene Editing-Based Approaches in Forages and Rumen Archaea for Reducing Livestock Methane Emissions" Agriculture 12, no. 11: 1780. https://doi.org/10.3390/agriculture12111780

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop