Skip to main content
Horticulture Research logoLink to Horticulture Research
. 2022 Sep 28;9:uhac223. doi: 10.1093/hr/uhac223

Natural products of medicinal plants: biosynthesis and bioengineering in post-genomic era

Li Guo 1,#,, Hui Yao 2,#, Weikai Chen 3,#, Xumei Wang 4, Peng Ye 5, Zhichao Xu 6, Sisheng Zhang 7, Hong Wu 8,
PMCID: PMC9720450  PMID: 36479585

Abstract

Globally, medicinal plant natural products (PNPs) are a major source of substances used in traditional and modern medicine. As we human race face the tremendous public health challenge posed by emerging infectious diseases, antibiotic resistance and surging drug prices etc., harnessing the healing power of medicinal plants gifted from mother nature is more urgent than ever in helping us survive future challenge in a sustainable way. PNP research efforts in the pre-genomic era focus on discovering bioactive molecules with pharmaceutical activities, and identifying individual genes responsible for biosynthesis. Critically, systemic biological, multi- and inter-disciplinary approaches integrating and interrogating all accessible data from genomics, metabolomics, structural biology, and chemical informatics are necessary to accelerate the full characterization of biosynthetic and regulatory circuitry for producing PNPs in medicinal plants. In this review, we attempt to provide a brief update on the current research of PNPs in medicinal plants by focusing on how different state-of-the-art biotechnologies facilitate their discovery, the molecular basis of their biosynthesis, as well as synthetic biology. Finally, we humbly provide a foresight of the research trend for understanding the biology of medicinal plants in the coming decades.

Natural products in medicinal plants: hidden treasures with healing power

Plants have existed on Earth for hundreds of millions of years and evolved ingenious chemical factories to survive exogenous and endogenous stresses [1]. These chemicals known as secondary metabolites or natural products are synthesized by plants to accommodate environmental changes without disrupting much of their cellular and developmental physiological processes [2]. To date more than 100 000 natural products are present in the Kingdom of Plants, primarily involved in plant defense against biotic and abiotic stresses [3]. These powerful substances are also important chemical signals mediating plant communication with symbiotic microorganisms, and attracting pollinators and seed dispersal. Derived from primary metabolites, secondary metabolites accumulate at cellular, tissue and organ levels through diverse biosynthetic pathways [4]. Plant natural products (PNPs) are generally divided into three classes: phenolics, terpenoids, and alkaloids [5] and broadly used as pharmaceuticals, nutraceuticals, cosmetics, and fine chemicals. Phenolics are synthesized from the shikimic acid biosynthetic pathway where the final products are formed after phenylalanine and aromatic amino acids undergo deaminization, hydroxylation, and coupling reactions [7] (Fig. 1A). Ingeneral, phenolics consist of monophenols such as benzenoids,and polyphenols such as flavonoids, stilbenoids, and coumuminoids [6]. Terpenoids are biosynthesized through mevalonic acid and methylerythritol phosphate pathways from isopentenyl diphosphate (IPP, C5), the precursor and fundamental structural unit of all terpenoids including monoterpenoids (C10), sesquiterpenoids (C15), diterpenoids (C20), and triterpenoids (C30) [8] (Fig. 1B). Alkaloids are a large group of plant nitrogen-containing compounds with a broad range of pharmaceutical activities such as painkilling (e.g. morphine), cough-suppressing (e.g. noscapine), anti-inflammation (e.g. sanguinarine, berberine), and anti-cancer (e.g. vinblastine, noscapine) (Fig. 2). Originated from the amino acid and isoamylene biosynthetic pathway, alkaloids are generally classified into sparteine, quinine, mescaline, coniine, and aconitine (Fig. 2). The biosynthetic pathways of different alkaloids are diversified and often independent. Recently, Lichman [9] has summarized alkaloid pathways as four general steps: (i) accumulation of an amine precursor; (ii) accumulation of an aldehyde precursor; (iii) formation of an iminium cation; and (iv) a mannich-like reaction.

Figure 1.

Figure 1

Proposed general biosynthetic pathway of plant phenolics (A) and terpenoids (B). DMAPP: dimethylallyl pyrophosphate; IPP: isopentenyl pyrophosphate; MEP: methylerythritol phosphate; MVA: mevalonic acid.

Figure 2.

Figure 2

Common alkaloid types (A) and examples of pharmaceutical alkaloids (B) in medicinal plants.

PNPs are bioactive substances dispensable to normal plant cellular functions yet vital to biodefense and environmental adaptation of plants with a sessile lifestyle. As a major source of traditional and modern medicine, PNPs have had broad implications for human health as a herb remedy for thousands of years, and changed the course of human civilization and history [9]. The healing power of medicinal herbs has long been recognized and harnessed by our human ancestors and forefathers, who learned to use plant-based folk medicine to cure ailments such as headache, fever, and pains. For example, archaeologists found in a grave from Shanidar – an archaeological site in Iraq – that a Neanderthal man may have used plant-based medicine at around 60 000 bce, the earliest record of human use of herb medicine [10]. In addition, ancient Europeans had started to cultivate and use different varieties of opium poppy plants at least 5000 years ago [11]. Another example is tobacco, which was historically used to treat various ailments including yaws, syphilis, and black death given its strong antimicrobial activities [12,13]. Much of early human knowledge about medical herbs is documented in ancient scriptures and literature such as Treatise on Cold Pathogenic and Miscellaneous Diseases by Zhang Zhongjing (Eastern Han dynasty) and Compendium of Materia Medica composed by Li Shizhen (Ming dynasty). However, the use of medicinal herbs in traditional medicine has been largely considered as empiricism with little knowledge of the chemical properties of the effective PNPs. The first isolated PNP was morphine, a benzylisoquiloine alkaloid in opium poppy (Papaver somniferum) plants, by Germany pharmacist Friedrich Sertürner at around 1817, marking the birth of modern chemistry of natural products [14]. Later on, another alkaloid quinine isolated from the bark of cinchona tree (Cinchona officinalis) became the first effective medicine against malaria, caused by mosquito-transmitted Plasmodium species [15]. Salicin, the original source of aspirin and identified in the bark of willow tree (Salix babylonica), is another significantly used PNP, used prominently as a pain reliever [16]. To date, hundreds of plant-based bioactive compounds have been identified and many are used as an effective treatment of human diseases such as ginsenoside (anti-tumor), paclitaxel/taxol (anti-tumor), and artemisinin (anti-malaria) etc. The sesquiterpene endoperoxide artemisinin from Artemisia annua is recommended by the WHO as the most effective drug against malaria [17]. The paclitaxel (taxol) isolated from the tree barks of Taxus genus has been approved for the treatment of ovarian, breast, and lung cancer, as well as Kaposi’s sarcoma [18]. These examples have demonstrated that medicinal plants and their powerful natural products have great healing power and have shaped the development of human history.

Throughout history, the human race has battled with many infectious diseases such as tuberculosis, cholera, malaria, black death, influenza, smallpox, etc. The most recent public health challenge comes from the coronavirus pandemic initiated in 2019 (COVID-19) which continuously threatens the world with the non-stop emergence of new variants. These pandemics each and collectively have led to significant progress in human knowledge of medical sciences and the generation of new public health solutions in which medicinal plants played important roles. Because antibiotics that target bacterial and fungal pathogens are futile against viral infection, the current solution against viral infections such as influenza and coronaviruses is primarily through vaccination, whereas effective virus-killing drugs are highly desired but scarce. Whereas western medicine working against COVID remains under development and clinical trials such as sabizabulin [19], remdesivir [20], hydroxychloroquine [21], and PNPs with anti-viral activities have been reported as effective to contain viral replication and alleviate patient symptoms. For example, flavonoids such as neo-hesperidin, hesperidin, baicalin, kaempferol 3-O-rutinoside, and rutin from different sources, and a series of xanthones from Swertia plants could effectively interact with SARS-CoV-2 targets [22]. In addition, molecular docking was recently performed using flavonoids from fruit peel of Citrus reticulata ‘Chachi’ to target the spike proteins, 3CLpro, PLpro, and RdRp of SARS-CoV-2, suggesting that many flavonoids have stronger affinity with the targets than do positive control drugs [23]. Recently, in a multicenter, prospective and randomized controlled clinical trial, Lianhuaqingwen capsule, a manufactured product of the traditional Chinese medicine (TCM) formula, could significantly inhibit SARS-CoV-2 replication [24]. Despite the promising therapeutic effect of medicinal herbs against COVID, it remains unknown what is the causing substance, and mechanisms of action against the virus. Furthermore, because biosynthetic pathways of most PNPs are elusive, the working molecules cannot be obtained in sufficient quantities to satisfy the need for drugs to control global pandemics. For decades, the biochemical pathways of PNP biosynthesis, genome architecture, and regulation of PNP production, and most importantly bioengineering to massively produce bioactive PNPs are all extensively researched areas in medicinal plant biology and chemistry. Here, we attempt to review the current research of PNPs by focusing on how different state-of-the-art multi-discipline technologies facilitate their discovery, the molecular basis of biosynthesis, as well as bioengineering via breeding and synthetic biology. Finally, we highlight a few research trends regarding the exploitation of medicinal plants in the next decades [25].

Decoding biosynthetic pathways of PNPs is key to their applications

The challenge of acquiring PNPs en masse stems from the fact that medicinal plants have a tight regulation of producing these chemicals, mostly localized in specific cells and tissues under certain conditions [26]. The biosynthesis of PNPs typically goes through a cascade of enzymatic reactions converting primary metabolites into various structurally diverse secondary metabolites. Although some reactions can occur spontaneously in nature, most steps require a catalysis by enzymes such as cytochrome P450, methyltransferases, O-methyltransferases, deaminases, UDP-glucuronosyltransferases, etc. A major challenge in exploitation of PNPs is to understand their biosynthetic pathways so that the bioengineering approach can be applied to produce them on a massive scale through plant breeding or synthetic biology. Fully resolving the biosynthetic pathways of any PNP is a daunting task, because plants have evolved a complex cellular network of metabolic pathways [27], formed by various enzymes catalyzing a myriad of biochemical reactions, and regulatory proteins fine-tuning the spatial and temporal accumulation of PNPs. Currently, the full biochemical pathways remain unknown for the majority of medicinal PNPs after years of research efforts, highlighting the difficulty of decoding PNP biosynthetic pathways.

Before the genomic era, plant biosynthetic pathway characterization was laborious and time-consuming, either relying on approaches such as isotope labeling and forward genetics, such as by creating random mutants followed by analysing their metabolic profiles, or involving sequence-homology based gene cloning to identify individual biosynthetic enzymes [28]. These studies typically identify genes encoding parts of the pathways through guilt-by-association, as loss- or gain-of-function mutations of true biosynthetic genes can alter metabolic profiles. However, they are usually inept to resolve the complete components and reconstruct the pathway owing to the pleiotropic or promiscuous nature of the biosynthetic enzymes. The majority of our knowledge about PNP biosynthetic pathways so far is derived from such homology-based or transcriptome-based gene mining [29, 30]. With growing volumes of genomic data available for medicinal plants, it is now common to exploit high-throughput data mining combined with experimental validations to untangle the complex biosynthetic pathways and networks underlying PNP accumulation in medicinal herbs.

PNP biosynthetic genes are usually co-expressed and co-regulated in specific tissues and growth stages. Therefore, in the post-genomic era, gene expression profiling techniques such as whole transcriptome sequencing (RNA-seq) enable researchers to quickly narrow down the co-expressed candidate genes encoding specific biosynthetic pathways, typically via comparative transcriptiome analysis of plant samples with contrasting levels of metabolic production. Transcriptomic analysis combined with pathway inference based on chemistry logic, and experimental validations in heterologous hosts are routinely used to identify biosynthetic genes for PNPs such as thebaine and noscapine of opium poppy (P. somniferum) [31], sanguinarine and chelerythrine of Macleaya cordata [32], vinblastine of Madagascar periwinkle [33], colchicine of Colchicum autumnale [34] and strychnine of Strychnos nux-vomica [35]. Alternatively, proteomic profiling is also used to identify proteins corresponding to specific biosynthetic pathways, capturing translational and post-translational modifications unseen in transcriptomic data. Candidate genes identified by omic profiling analysis are then validated experimentally to ascertain their biochemical functions such as catalyzing specific reactions, metabolite transport or transcriptional regulation. It typically involves heterologously expressing the candidate gene(s) in microbial cells including bacteria (Escherichia coli [36], Corynebacterium glutamicum [37]), yeasts (Saccharomyces cerevisiae [38], Pichia pastoris [39]), or plant chassis such as tobacco (Nicotiana benthamiana [40]) and algae (Chlamydomonas reinhardtii [41], Phaeodactylum tricornutum [42]), followed by detection of the target metabolites using untargeted metabolomics. Metabolomics study the full complement of plant metabolites through high performance liquid chromatography (HPLC)- or gas chromatography (GC) coupled with mass spectrometry (MS). Integrated mining of multi-dimensional data such as transcriptomic or proteomic with metabolomic profiling in different tissues and growth stages of medicinal plants enables gene-metabolite association. Network mining using these big data can reconstruct the gene co-expression networks correlated with accumulation of PNPs, thus generating hypothesis for downstream experimental validations [43]. This approach still suffers from the fact that many biosynthetic genes may be cryptic or lowly expressed, leaving them undiscovered by the expression-based methods. Thus, systematic decoding of PNP biosynthetic pathways will require a framework of high-throughput analysis of omic data from plants growing under multiple conditions or developmental stages. For example, two recent studies acquired transcriptomic and metabolomic data of tomato and rice plants across the full spectrum of growing stages, and by integrative data analysis revealed gene modules associated with natural products [44, 45]. It is expected that similar analysis from full growth stages of medicinal plants will facilitate discovery of biosynthetic genes and yield critical insight into the regulatory networks underlying PNP biosynthesis.

Medicinal plant genomes yield insights into composition and evolution of biosynthetic pathways

Genome sequence dictates the foundation of biological functions of all life forms. Despite the progress made by the homology- and transcriptome-based approach, the elucidation of full biosynthetic pathways is often hampered by a lack of reference genome sequences for medicinal plants. A reference genome sets the foundation to identify all protein-coding genes, regulatory DNA elements and importantly their precise genomic locations. Next-generation sequencing (NGS) and third-generation sequencing (TGS) technologies have revolutionized biological sciences by changing the way genomes are decoded. The first human and plant (Arabidopsis thaliana) genome assembly were initially achieved using Sanger sequencing. Despite the high accuracy, Sanger sequencing is expensive and time-consuming to generate sequencing data for assembly of eukaryotic genomes of mid to large sizes. Since around 2005, genome assembly projects started to adopt high-throughput sequencing platforms like Solexa, Ion Torrent and later Illumina, giving rise to the first reference genomes for many organisms. However, contig-level assemblies using short reads have low contiguity (low N50) with numerous assembly errors due to high repeat content of plant genomes, even when a high coverage and long insert such as mate-pair libraries or linked-reads are used. Scaffold level assemblies are often improved using genetic map data such as GBS (genotyping by sequencing), or optical maps to anchor the contigs to linkage groups. However, a high-resolution genetic map is essential to reducing contig misplacement but often unavailable for non-model plants. TGS technologies developed by Pacific Biosciences (PB) and Oxford Nanopore technology (ONT) produce single-molecule DNA sequencing reads of 20 kb or longer, albeit error-prone (up to 15%). TGS became a game changer for genome assembly because long reads can often span most repetitive regions. Besides, technologies such as chromatin conformation capture sequencing (e.g. Hi-C) and Bionano are now routinely used to anchor contigs to chromosomes and correct misassemblies present in NGS and TGS draft assemblies. As a result, for model organisms and many agricultural organisms, genome assembly quality and contiguity have leaped to much higher levels with a combination of long-read and short-read sequencing data.

Medicinal plant genomes have a wide genome size range, high heterozygosity rates and repeat contents, making them difficult to assemble correctly [46]. Leveraging these different technologies combined with improving bioinformatic algorithms has yielded a growing number of high-quality medicinal plant genome assemblies and annotations. Nearly 100 species of medicinal plants (Table 1) have at least one version of reference genome available [47139], although the quality of current genome assemblies varies depending on the genome complexity of medicinal plants and choice of sequencing technologies as well as computational tools used in assembly. The herbgenomics initiative, firstly proposed in 2010, has greatly promoted the elucidation of biosynthetic pathways for many medicinal bioactive ingredients [48]. Under this initiative and many independent genome projects, several medicinal plants have had reference genomes assembled even at chromosome level, such as P. somniferum [49], Camptotheca acuminata [50], Scutellaria baicalensis [51], Panax notoginseng [52], Tripterygium wilfordii [53], Salvia miltiorrhiza [54], Taxus wallichiana [55], and Erigeron breviscapus [56]. Lately, a Chinese consortium of the 1 K Herb Genomes Project has been officially launched to produce high-quality genome sequences for 1000 high-value TCM in order to promote the study and exploitation of their PNPs.

Table 1.

Sequenced genomes of medicinal plants and their primary medicinal ingredients.

Species Genome size Contig N50 No. of annotated genes AssemblyLevel a Release time Primary medicinal ingredients Reference
Apium graveolens 3.33 Gb 790.6 kb 31 326 C 2020 Apigenin and luteolin [60]
Aralia elata 1.05 Gb 1.20 Mb 35 042 C 2022 Oleanane-type triterpenoids [61]
Coriandrum sativum 2.12 Gb 604.1 kb 40 747 C 2020 Mannitol, furfural and linalool [62]
Panax ginseng 3.41 Gb 19.75 Mb 65 913 C 2022 Dammarane-type saponins [63]
Panax japonicus 2.09 Gb 1.22 Mb 74 307 C 2022 Dammarane-type saponins [63]
Panax notoginseng 2.66 Gb 1.12 Mb 37 606 C 2020 Dammarane-type ginsenoside [52]
Panax quinquefolius 3.60 Gb 0.87 Mb 64 247 C 2022 Dammarane-type saponins [63]
Panax stipuleanatus 2.15 Gb 2.88 Mb 41 224 C 2022 Dammarane-type saponins [63]
Allium sativum 16.24 Gb 194 kb 57 561 C 2020 Allicin [64]
Dendrobium huoshanense 1.28 Gb 598 kb 21 070 C 2020 Polysaccharides and alkaloids [65]
Dendrobium officinale 1.23 Gb 1.44 Mb 27 631 C 2021 Polysaccharides, alkaloids and flavonoids [66]
Gastrodia elata 1.04 Gb 9.18 Mb 18 844 C 2022 Gastrodin, phydroxybenzyl alcohol and parishin [67]
Arctium lappa 1.73Gb 74.69 Mb 47 055 C 2022 Arctigenin and arctiin [68]
Artemisia annua 1.55 Gb 262.1 kb 54 347 C, H 2022 Artemisinin [69]
Artemisia argyi 8.03 Gb 6.25 Mb 279 294 C, H 2022 Hispidulin, jaceosidin, and eupatilin [70]
Carthamus tinctorius 1.06 Gb 21.23 Mb 33 343 C 2021 Linoleic acid and hydroxysafflor yellow A [71]
Chrysanthemum nankingense 2.53 Gb 130.7 kb 56 870 S 2018 Flavonoids and terpenes [72]
Erigeron breviscapus 1.43 Gb 140.95 kb 43 514 C 2020 Scutellarin [56]
Smallanthus sonchifolius 2.72 Gb 87.39 Mb 89 960 C 2022 Saccharides (inulin) [68]
Platycodon grandiflorus 622.86 Mb 29.34 Mb 22 358 C 2022 Triterpenoid saponins [73]
Isatis indigotica 293.88 Mb 1.18 Mb 30 323 C 2020 Indole alkaloids, β-sitosterol and daucosterol [74]
Lepidium meyenii 743 Mb 81.78 kb 96 417 S 2016 Macaridine, macamides and macaene [75]
Polygonum cuspidatum 2.56 Gb 2.77 kb 55 075 S 2019 Stilbenes and quinones [76]
Tripterygium wilfordii 348.38 Mb 4.36 Mb 28 321 C 2020 Triptolide [53]
Ceratophyllum demersum 860.5 Mb 2.57 Mb 30 138 C 2020 Plastocyanin and ferredoxin [77]
Camptotheca acuminata 414.95 Mb 1.47 Mb 27 940 C 2021 Camptothecin [50]
Benincasa hispida 912.95 Mb 145 kb 27 467 C 2019 Vitamins and flavonoids [78]
Gynostemma pentaphyllum 518.46 Mb 6.67 Mb 25 285 C 2021 Gypenosides [79]
Luffa cylindrica 656.19 Mb 8.80 Mb 25 508 C 2020 Sapogenins [80]
Momordica charantia 293.6 Mb 3.3 Mb 26 427 C 2020 Cucurbitacins and cucurbitane glycosides [81]
Siraitia grosvenorii 467.07 Mb 433.68 kb 30 565 S 2018 Mogrosides [82]
Lonicera japonica 843.2 Mb 2.1 Mb 33 939 C 2020 Luteolin and chlorogenicacid [83]
Diospyros lotus 907 Mb 1.06 Mb 40 532 C 2020 Tannin [84]
Camellia sinensis 3.02 Gb 19.96 kb 36 951 C 2017 Caffeine and theanine [85]
Glycyrrhiza uralensis 378.86 Mb 7.32 kb 38 135 S 2017 Glycyrrhizin and liquiritin [86]
Senna tora 526.36 Mb 4.03 Mb 45 268 C 2020 Anthraquinones [87]
Spatholobus suberectus 798.47 Mb 2.05 Mb 31 634 C 2019 Eriodictyol and dihydroquercetin [88]
Eucommia ulmoides 947.84 Mb 13.16 Mb 26 001 C 2020 Pinoresinol diglucoside and aucubin [89]
Calotropis gigantea 157.28 Mb 48.58 kb 18 197 S 2018 Cardenolides [90]
Catharanthus roseus 523 Mb 26.2 kb 33 829 S 2015 Vinblastine and vincristine [33]
Gelsemium elegans 335.13 Mb 10.23 Mb 26 768 C 2020 Gelsemine and koumine alkaloids [91]
Gardenia jasminoides 535.68 Mb 1.03 Mb 35 967 C 2020 Genipin and crocins [92]
Morinda officinalis 484.85 Mb 4.21 Mb 27 698 C 2021 Anthraquinones and monotropein [93]
Ophiorrhiza pumila 439.90 Mb 18.49 Mb 32 389 C 2021 Camptothecin [94]
Andrographis paniculata 284.32 Mb 5.15 Mb 24 015 C 2020 Neoandrographolide [95]
Mentha longifolia 353 Mb 4.47 kb 35 597 S 2017 Menthone and menthol [96]
Ocimum basilicum 2.06 Gb 48.30 Kb 78 990 S 2020 Linalool and cineole [97]
Origanum vulgare 630.04 Mb 26.28 kb 32 623 S 2020 Carvacrol, thymol and ocimene [97]
Perilla frutescens 1.24 Gb 3.21 Mb 93 008 C 2021 Perillaldehyde, perillaalcohol and limonene [98]
Pogostemon cablin 1.94 Gb 7.97 Mb 109 696 C, H 2022 Patchouli alcohol [99]
Rosmarinus officinalis 1.01 Gb 21.82 kb 51 389 S 2020 Rosmarinic acid, camphor and carnosol [97]
Salvia bowleyana 462.44 Mb 1.18 Mb 44 044 C 2021 Tanshinone and salvianolic acid [100]
Salvia miltiorrhiza 594.75 Mb 2.7 Mb 32 483 S 2020 Phenolic acids and tanshinones [54]
Salvia splendens 809.16 Mb 3.77 Mb 88 489 C 2021 Anticoagulant [101]
Scutellaria baicalensis 377.0 Mb 2.10 Mb 33 414 C 2020 Baicalein, scutellarein, norwogonin, wogonin [102]
Scutellaria barbata 353.0 Mb 2.50 Mb 41 697 C 2020 Baicalein, scutellarein, norwogonin, wogonin [102]
Forsythia suspensa 737.47 Mb 7.33 Mb 33 062 S 2020 Pinoresinol and phillyrin [103]
Antirrhinum majus 520 Mb 730 kb 37 714 C 2019 Antirrhinoside and linolenic acid [104]
Chimonanthus praecox 695.36 Mb 2.19 Mb 23 591 C 2020 Essential oil [105]
Cinnamomum kanehirae 730.42 Mb 498.9 kb 27 899 C 2019 Camphor [106]
Paris polyphylla 70.18 Gb 1.81 kb 34 257 S 2020 Polyphyllins [107]
Ricinus communis 325.5 Mb 21.1 kb 31 237 S 2010 Ricin [108]
Hypericum perforatum 373.65 Mb 1.41 Mb 29 150 S 2021 Hypericin, hyperforin, and melatonin [109]
Linum usitatissimum 318.25 Mb 20.1 kb 43.484 S 2012 Unsaturated fatty acids and secoisolariciresinol diglucoside [110]
Passiflora edulis 1.34 Gb 6.40 Mb 23 171 C 2021 C-glycosyl flavonoids [111]
Aquilaria sinensis 783.8 Mb 60.21 Kb 35 965 C 2020 Sesquiterpenes and flindersiachromone [112]
Punica granatum 328.38 Mb 66.97 kb 29 229 C 2017 Punicalagin [113]
Euryale ferox 725.2 Mb 4.75 Mb 40 297 C 2020 Sterols [77]
Nymphaea colorata 409 Mb 2.1 Mb 31 580 C 2020 Alkaloids [114]
Aristolochia contorta 210.53 Mb 2.63 Mb 18 311 C 2022 Benzylisoquinoline alkaloids and aristolochic acids [115]
Piper nigrum 761.22 Mb NA 63 466 C 2019 Piperine [116]
Coix lacryma-jobi 1.73 Gb 3.19 Mb 44 485 C 2020 Coixol [117]
Nelumbo nucifera 821.29 Mb 484.3 kb 32 124 C 2020 Neferine, liensinine, isoliensinine, and nuciferine [118]
Macleaya cordata 378 Mb 25 kb 22 328 S 2017 Sanguinarine and chelerythrine [119]
Papaver somniferum 2.72 Gb 1.77 Mb 51 213 C 2018 Morphine and codeine [49]
Coptis chinensis 936.60 Mb 806.6 kb 41 004 C 2021 Protoberberine-type alkaloids [120]
Cannabis sativa 876.15 Mb 1.96 Mb 33 674 C 2018 Delta-9-tetrahydrocannabinolic acid and cannabidiolic acid [121]
Broussonetia papyrifera 386.83 Mb 171.2 kb 30 512 C 2019 Saponin, vitamin B and oil [122]
Morus notabilis 335.39 Mb 5.72 Mb 26 010 C 2020 Phenolic acids, flavonoids and alkaloids [123]
Ziziphus jujuba 437.65 Mb 34.0 kb 32 808 C 2014 Ziziphus saponin [124]
Eriobotrya japonica 760.10 Mb 5.02 Mb 45 743 C 2020 Volatile oil, amygdalin and ursolic acid [125]
Rosa chinensis 513.85 Mb 22.2 Mb 36 377 C 2018 Gallic acid [126]
Boehmeria nivea 344.62 Mb 24.5 kb 30 237 S 2018 Ramie acid [127]
Pistacia vera 671.28 Mb 714.2 kb 31 784 S 2019 Phenolic compounds and vitamins [128]
Acer truncatum 633.28 Mb 773.17 kb 28 438 S 2020 Unsaturated fatty acids (Nervonic acid) [129]
Xanthoceras sorbifolium 439.97 Mb 645.45 kb 21 059 C 2019 Unsaturated fatty acids (Nervonic acid) [130]
Rhodiola crenulata 344.5 Mb 25.4 kb 31 517 S 2017 Salidroside and tyrosol [131]
Ipomoea nil 734.8 Mb 1.87 Mb 42 783 C 2016 Diterpenoids [132]
Datura stramonium 1.29 Gb 58.2 kb 30 934 S 2021 Scopolamine and atropine [133]
Wurfbainia villosa 2.80 Gb 9.13 Mb 42 588 C 2022 Bornyl acetate, borneol, and camphor [134]
Zingiber officinale 1.53 Gb 4.68 Mb 39 217 C, H 2021 Gingerols, gingerdiols, zingerone, paradols, and shogaol [135]
Selaginella tamariscina 300.73 Mb 2.14 kb 27 761 S 2018 Selaginellins and amentoflavone [136]
Ginkgo biloba 9.87 Gb 1.58 Mb 27 832 C 2021 Ginkgolide and bilobalide [137]
Gnetum montanum 4.07 Gb 475.2 kb 27 491 S 2018 Stilbenoids [138]
Taxus chinensis 10.23 Gb 2.44 Mb 42 746 C 2021 Paclitaxel [139]
Taxus wallichiana 10.9 Gb 8.6 Mb 44 008 C 2021 Paclitaxel [55]
a

Genome assembly level.

C, chromosome level; S, scaffold level; H, haplotype-resolved genome assembly; NA: not reported

The chromosome-level genome assemblies are instrumental to highly robust downstream genomic analyses such as comparative genome analysis, chromosome evolution analysis, and gene cluster characterization etc. For example, chromosome-level assemblies of P. somniferum has allowed Guo et al. to discover a gene cluster (BIA gene cluster) that encodes biosynthetic pathways for two morphinans: morphine and noscapine [49]. Two additional assemblies of Papaver species produced by Yang et al. have enabled them to reconstruct the evolutionary history of Papaver karyotypes, showing morphinan biosynthetic pathways underwent punctuated evolution pattern [57]. Tu et al. [53] assembled a high quality chromosomal-scale Tripterygium wilfordii genome and found the recent duplication of triptolide biosynthetic pathway genes. Then multiple omics methods were integrated to construct gene-to-metabolite network, and finally a CYP728B70 that participated in triptolide biosynthesis was identified. The genome assemblies for C. acuminata [50] and Catharanthus roseus [33] also provide comprehensive genomic resources for the analysis of camptothecin and vinblastine biosynthesis pathway, which share common upstream to produce loganic acid, and then flux into two independent branches, respectively. The C. roseus genome, coupled with chemical investigations, enabled the discovery of the last two enzymes of precondylocarpine acetate synthase and dihydroprecondylocarpine synthase responsible for vinblastine biosynthesis [33, 58], resolving a long-standing question of how vinblastine/vincristine is synthesized, making their heterologous production possible. The C. acuminata genome found two secologanic acid synthases that converted the loganic acid flux into camptothecin production, and the downstream candidate genes set the foundation to fully discover camptothecin biosynthetic mechanism [50]. Furthermore, high quality genomes are also essential in genome-wide association studies (GWAS) to identify quantitative trait loci (QTLs) associated with PNP content. For example, Fan et al. assembled a chromosome-level genome of medicinal plant Panax notoginseng and performed GWAS on 240 cultivars to identify genes linked to dry root weight and stem thickness [59]. The availability of high-quality reference genomes provides critical resources to the elucidation of biosynthetic pathways of high-value PNPs in medicinal plants.

Discovery of metabolic gene clusters through genomic mining

Microbial genes controlling secondary metabolite biosynthesis are typically clustered in certain genomic regions, known as metabolic gene clusters (MGCs). Unlike microbes, plant biosynthesis genes are usually dispersed throughout the genome and MGCs have long been considered rare in plants. However, MGCs have recently been identified in several plants including Arabidopsis [140, 141], rice [142], maize [143], and several medicinal plants such as opium poppy [49] and Taxus [55, 139]. The MGCs contain at least three non-homologous genes, ranging from tens to several hundred kilobases in total length. To date, over 30 MGCs in plants have been reported to encode PNP biosynthetic pathways based on experimental evidence [143], encoding terpenoides [140142], alkaloids [49], steroidal glycoalkaloids [144], and fatty acids [145]. Some of these PNPs encoded by gene clusters have medicinal values such as morphine and noscapine, while many have known roles in antimicrobial, allelopathic activity, and plant defense against herbivores and pathogens. Many of these functionally characterized MGCs have been discovered even before a reference genome is available. In the post-genomic era, the availability of reference genomes expedited identification of MGCs in plants using genomic mining. Recently, computer algorithms such as Plantismash [146], Phytoclust [147] and PlantClusterFinder [148] have been developed to predict MGCs encoding potential biosynthetic pathways of secondary metabolites. A large number of potential MGCs have been found in plants encoding unknown biosynthetic pathways, highlighting the limitation of our knowledge of what and how plants can produce chemically. For example, genome mining of P. somniferum genome has revealed 84 MGCs, among which one cluster encoding the pathway for morphinan and noscapine has been functionally validated [49, 149]. Tomato (Solanum lycopersicum) has 47 predicted MGCs, four of which have been associated with alpha-tomatine [144], lycosantalonol [150], fatty acids [145], and hydroxycinnamic acid amide [151] biosynthesis. A six-gene cluster for taxadiene biosynthesis was recently identified in the Taxus genome, involving in the first two biosynthetic steps, which helps to decode the complete taxol biosynthesis in the future [139]. Wheat genome mining combined with transcriptomic analysis has recently identified six pathogen-induced biosynthetic pathways encoded by MGCs, producing flavonoids and terpenes that could potentially be used as phytoalexins in disease control [152]. Combining transcriptomic and metabolic profiling data would be useful to link MGCs with particular metabolites, such as the targeting of a four-gene cluster with the falcarindiol biosynthesis in tomato [145], although such strategy faces the challenge of lacking co-expression in many MGCs.

Despite the genomic discovery of hundreds of plant MGCs, questions remain to be answered regarding this specialized genetic architecture. First, how did plants gain gene clusters during evolution? MGCs in bacterial and fungi are commonly formed through gene duplication, translocation, and horizontal gene transfer (HGT). Despite a few exceptions [153, 154], it is uncommon for plants to undergo HGT and there must be special mechanisms for MGC formation in plants. Recent genomic analyses [57, 155] have shown that structural variation events such as whole genome duplications, chromosome fission and fusion, gene duplication, translocation and loss have been implicated in the birth and evolution of biosynthesis gene clusters, supporting the theory of punctuated evolution in forming plant specialized metabolites. Overall, investigation of plant MGC evolution remains at the infant stage, requiring comparative analysis of a large number of plant genomes. It will help us understand the major driving forces of PNP evolution and its ecological impact in nature.

Second, how do many of the MGCs actually contribute to PNP biosynthesis? As more candidate MGCs continue to be identified from plant genomes via an in silico approach, it is critical to functionally characterize predicted MGCs of unknown function and associate them with potential natural products. Expressing the candidate MGCs in a heterologous host such as yeast or E. coli will be informative to determine the synthesized product, as shown by several recent examples [151, 156]. For instance, Kong et al. used yeast as a chassis to investigate the function of a tomato gene cluster, and discovered a novel naringenin chalcone synthase responsible for the production of dihydro-coumaroyl anthranilate amide [151]. The caveat of this approach is that most predicted plant MGCs are quite large (up to hundreds of kb), presenting a huge challenge to cloning them into bacterial or yeast expression vectors. The plant MGCs successfully cloned and expressed in microbial hosts are mostly mini gene clusters of several kb containing only a few open reading frames. In addition, expressing plant proteins in yeast or E. coli cells does not always work due to different codon usage and post-translational modifications in eukaryotic versus prokaryotic cells. Alternatively, validation using plant host such as Nicotiana benthemiana enables expression of candidate MGCs via Agrobacterium infiltration of plant leaves or cells, followed by metabolomic detection, although it faces the same problem of delivering and expressing long MGC fragment into tobacco cells. Recently, a platform for high-throughput secondary metabolite discovery has been developed for filamentous fungi by cloning and expressing fragmented genomic DNAs containing MGCs into fungal artificial chromosomes followed by metabolomic profiling [157]. Application of this or a similar approach has not been reported in plants considering the low genomic fraction of plant MGCs and lack of a proper artificial chromosome cloning system. A high-throughput MGC validation platform will expedite the identification and utilization of novel PNPs.

Bioengineering of PNP through plant biotechnology

In nature, PNPs are accumulated at low abundance and only in specific tissues and developmental stages of medicinal plants for two major reasons. Firstly, biosynthesis of these compounds consumes energy and competes with normal plant vegetative growth and reproduction. Secondly, most PNPs have cell toxicity from which plants have to protect themselves by detoxification, storing them in compartments, or only producing the toxic chemicals when and where needed. Evolutionarily, PNPs have probablyundergone natural as well as human selection. For example, P. somniferum accumulates high levels of painkilling morphines in capsules instead of other tissues, and the amount of morphine produced differed among cultivars [158]. By contrast, its close relative Papaver rhoeas only produces a trace amount of morphine [57]. This suggests that the ability to produce morphine has been under natural selection in poppy plants, and likely selected by domestication and breeding process.

The naturally low content of PNPs in medicinal herbs renders a major bottleneck in drug developments and clinical therapeutics. Structures of PNPs are often too complex for a profitable production by total chemical synthesis. Therefore, plant extraction remains the primary commercial source of most PNP for pharmaceuticals, causing over-exploitation of natural resources and instability to the Earth’s ecosystem. For instance, taxol, a well-known anticancer drug ingredient derived from the bark of yew tree once put the yew on the verge of extinction due to exhaustive exploitation. The demand of medical PNPs thus stimulates the breeding of superior germplasm resources for sustainable use of medicinal herbs. There are many challengesin breeding medicinal plants for high PNP yield, including limited understanding of how PNPs are exactly made and regulated by plants, lack of high-quality genome sequence, annotation and molecular markers, long breeding cycles as well as the difficulty of genetic transformation. Herb genomic research has accelerated the identification of functional genes and genome-wide molecular markers, linked molecular markers with desired characters, and improved breeding medicinal herbs. Many efforts have been made to increase PNP yield through plant breeding and biotechnological improvement, including artemisinin in A. annua [159, 160], morphine in P. somniferum [161], THC (tetrahydrocannabinol) in Cannabis sativa [162], etc. A. annua has been a primary source of the anti-malaria drug artemisinin. A. annua transcriptome sequencing enabled construction of genetic linkage map and identification of quantitative trait loci (QTL) that control artemisinin yield [160], providing genetic resources for molecular breeding. Phenotype selection coupled with molecular breeding in the past decadehas led to the production of Artemisia F1 Seed (https://www.artemisiaf1seed.org), increasing the artemisinin yield from 5 kg per hectare to 55 kg per hectare with a 1.44% of dry weight [159]. To date, A. annua remains the sole source of artemisinin globally, although artemisinin metabolic engineering has been reported [163].

The rise of modern biotechnology provides a novel strategy for precise and expedited medicinal plant breeding. A key breakthrough is the revolutionary genome editing technology, most notably the CRISPR-cas9 system, that allows precise genome bases to obtain traits of interest at an unprecedented pace [164]. This biotechnology has powered the next generation ofplant breeding to improve crop yield and quality such as PNP content. Unlike model and crop plants, the genome editing of medical herbs is still at the infant stage, hindered by the lack of genomic information and a reliable genetic transformation system. Nevertheless, CRISPR-Cas9 based genome editing has been reported in several medicinal plants towards optimizing production of pharmacological components in P. somniferum [165], S. miltiorrhiza [166], Dendrobium officinale [167] and Camelina sativa [168]. Notably, genome editing successfully targeted three FAD2 (fatty acid desaturase 2) genes in allohexaploid Camelina sativa and enhanced seed fatty acid levels [168]. CRISPR-Cas9 mediated gene deletion significantly decreased the benzylisoquinoline alkaloid flux in transgenic opium poppies [165]. Plant genetic engineering offers clear advantages in improving the yield of PNPs [169] as plant chassis naturally carries many fundamental plant biosynthetic gene circuits, making them natural cell factories to produce PNPs of interest. A paradigmatic case is the Golden Rice [170], where the whole β-carotene biosynthetic pathway was introduced into rice endosperm using Agrobacterium-mediated co-transformation to generate rice plants with carotenoid content up to 1.6 mg/g in the endosperm. Moreover, a high-efficiency vector system was developed for transgene stacking to engineer anthocyanin biosynthesis in rice endosperm [171]. In addition, Zhu et al. [172] developed a novel method called combinatorial nuclear transformation to generate multiplex-transgenic plants allowing five carotenogenic genes to be simultaneously transferred into a white maize through biolistic transformation, resulting in transgenic plants with elevated levels of β-carotene.

Besides modifying existing metabolic pathways, genetically engineered plant chassis offers a cheap and sustainable source to produce high-value PNPs. The de novo production of PNPs in model organisms such as Arabidopsis, tobacco, tomato, and moss has progressed rapidly recently. For example, Fuentes et al. [173] transferred the entire artemisinic acid metabolic pathway from A. annua to tobacco chloroplast genome using combinatorial supertransformation of transplastomic recipient lines (COSTREL). Plants with high artemisinic acid levels were then isolated through screening large populations of transplastomic lines. In addition, strategies to increase terpenoids including overexpression of rate-limiting enzymes, chloroplast-compartmentalized engineering, and integration of transcription factors have been applied to the production of momilactone [174] and taxadiene [175] in tobacco. Momilactones are a group of diterpenes predominantly found in rice with an allelopathic activity. Through changing the subcellular localization of prenyltransferase and diterpene synthases, the diterpene biosynthesis was rerouted from chloroplast to cytosolic MEP pathway, significantly promoting the production of momilactone in N. benthamiana [175]. Noteworthy, this strategy also enabled the discovery of missing steps in momilactone B pathway, providing insights into pathway reconstitution and elucidation for desired products.

Metabolic engineering of an in vitro plant tissue culture, such as suspension cell culture and hairy root culture, is another efficient approach to yield valuable phytochemicals. Hairy roots are induced by Agrobacterium rhizogenes mediated transformation, which can be applied as high-capacity bioreactor to produce PNPs without the need of light and hormones. Hairy root cultures were successfully induced to overproduce cannabinoids in C. sativa [176], phytosterols and ginsenosides in Panax ginseng [177], and curcumin in Atropa belladonna [178]. Suspension cell culture has also made great advances to produce valuable PNPs with high yields. Plant Cell Fermentation (PCF®) Technology (https://phytonbiotech.com/) could produce natural taxol directly from plant cells of Taxus chinensis v. marei, while the metabolic engineered grapevine cells were able to produce resveratrol derivatives when elicited with MeJA and methylated cyclodextrins [179]. In the suspension cell culture system, the addition of heterologous elicitors induces the biosynthetic gene expression and increases the production of PNPs. Glandular trichomes are hairy structures differentiated from epidermal cells, featured by their enormous capacity to synthesize, store and secret large quantities of metabolites with distinct types, making them an excellent platform for decoding the biosynthesis pathway of PNPs, and efficient phytochemical factories to produce PNPs [180]. Kortbeek et al. [181] engineered tomato glandular trichomes where a farnesyl diphosphate synthase was overexpressed, resulting in a decline of monoterpenoid production in the trichomes.

Synthetic biology: a green revolution for PNP bioengineering and industrialization

Although plant breeding can produce cultivars that accumulate higher level of metabolites than others do, it is still inefficient and environmentally unsustainable for massive production for commercial uses in most cases. Alternatively, synthetic biology where microbial (e.g. yeast and bacteria) chassis are used to massively produce PNPs offers a more effective and environment-friendly alternative. Synthetic biology aims to design microbial cell factories carrying genetic circuits made of biosynthetic genes for heterologous production of PNPs. It has several advantages over plant-based extraction, such as circumventing the requirement of growing plants, rapid production and little interference from natural environment [182]. With the development of synthetic biology tools and knowledge of biosynthetic pathways, PNPs such as artemisinic acid [183], amorphadiene [163], taxadiene [36], cannabinoids [184], morphine [38], and noscapine [185] has been successfully produced using engineered microbial cells. A prominent example is the semi-synthesis of artemisinin [183], where an engineered amorphadiene-producing yeast [38] produces artemisinic acid with the titer of 25 g/L, later converted to artemisinin by chemical synthesis. This opens a route to the industrial production of artemisinin against the urgent demand of anti-malarial drugs. Parallelly, Luo et al. [184] partitioned the cannabinoid metabolic pathway into three modules: an engineered S. cerevisiae MEP pathway to make more flux into geranyl pyrophosphate, a hexanoyl-CoA biosynthetic pathway and several Cannabis genes to accumulate more olivetolic acid, as well as a heterologous downstream pathway to form the corresponding cannabinoids. The engineered yeast strains yielded 1.6 mg/L of cannabinoid from the simple sugar galactose, laying a foundation for the large-scale production of cannabinoids.

Despite progress, it remains challenging to use microbe as a chassis to synthesize high-value PNPs [182]. First, decoding biosynthetic pathways is the prerequisite for successful heterologous production but remains a challenge for vast majority of PNPs. For instance, previous attempts to reconstruct taxol pathways in microbe without knowing the steps converting downstream taxadiene to final taxol ended up with a production of taxadiene, the first committed intermediate [185]. The combination of bioinformatics and downstream functional validations in heterologous hosts to resolve the biosynthetic pathways of PNPs from sequencing data plays a key role in synthetic biology today (Fig. 3). For example, the Taxus genome analysis identified a functional grouping of CYP725As and a taxadiene gene cluster, which will facilitate the future elucidation of taxol biosynthesis [55, 146].

Figure 3.

Figure 3

A roadmap for green revolution of herb medicine: a typical workflow for plant natural product (PNP) production via synthetic biology. A Selecting plants with superior PNP traits using traditional and transgenic breeding methods. B Whole genome and transcriptome sequencing of plant samples. C Assembly and annotations of medicinal plant genomes. D Revealing plant secondary metabolite synthesis gene cluster by omics data mining. E High-throughput assembly of PNP metabolic pathways. F Efficient production of PNPs in microorganisms or plants. G Industrial-scale planting and fermentation of transgenic microorganisms and plants. H Making PNP commodities with high commercial value.

Second, choosing and optimizing a microbial host is essential to maximize yield of PNPs. E. coli and S. cerevisiae are two of the most widely used microorganisms to engineer biosynthetic pathways, given their fast growth, well-known genetic background and well-established genetic manipulation methods [36, 38, 163, 183185]. However, it is challenging to express functional plant-derived cytochrome P450 genes in prokaryotic E. coli which lacks an intracellular organelle system, post-translational modification and electron transfer machinery, thus limiting its application in heterologous production of many PNPs [36]. For example, E. coli was engineered to produce artemisinic acid with high titer of 20 g/L amorphadiene, achieving only 1 g/L artemisinic acid [186], much lower than the aforementioned 25 g/L artemisinic acid in S. cerevisiae. Apart from the commonly used E. coli and S. cerevisiae, nonconventional chassis cells are also used such as the industrial production of 4-hydroxybenzoic acid by C. glutamicum [187] and (+)-nootkatone by Pichia pastoris [39].

Third, reconstituting an efficient module is key to heterogeneous production of PNPs. To construct heterogeneous expression cassettes, codon-optimization of plant genes is often required for microbial expression [36, 38]. Partition of complex biosynthetic pathways into several modules [36, 184] and the strains are optimized by synthetic biology tools.

Finally, the balance between microbial growth and PNP yield should be considered as many PNP and their intermediates, such as alkaloids and phenols, are toxic to the microorganisms. Taken together, with the rapid development of technologies including genome sequencing, multi-omic data mining, structure biology, directed evolution of proteins, and design of novel proteins, it is expected that the efficiency in synthesizing PNPs in microorganisms will be greatly improved in the near future.

Opportunities and challenges for future PNP research

It is an exciting time to study PNP in this golden era of chemical biology. New technologies in both experimental and computational sciences are emerging at an unprecedented pace. These technologies have enabled the untangling the complex biosynthetic pathways and networks using systems biology approach, providing insight into the mechanistic and evolutionary mechanisms behind the chemical and structural diversity of natural products. Accurate and complete assembly of medicinal plant genomes is the key to understanding genome function and evolutionary patterns and improving plant traits through breeding and synthetic biology. Horticultural plant genomes are notoriously difficult to assemble, due to their wide range of genome sizes (up to hundreds of Gb), various ploidy levels, high repeat content and heterozygosity. Yet with genome technologies advancing quickly in recent years, chromosome-scale assembly, once a rarity, is now a realistic target for most genome projects. A typical genome project now adopts a combination of different technologies including long-read and short-read sequencing data, 10× Genomics data, Hi-C sequencing data or Bionano optical genome maps. Bioinformatic algorithms are continuing to be developed to solve complex problems of genome assembly and annotation leveraging these different technologies [243]. As a result, a growing number of medicinal plants now have chromosome-level genome assembly and some offer obvious improvement over previous versions of assembly and annotation [244, 73].

Despite the progress, a long journey is ahead to resolve the complete genome sequence of medicinal plants. Twenty years after the first plant reference genome was produced, the majority of plant reference genomes initially assembled using short reads remain unimproved. Recently, a telomere to telomere (T2T) genome assembly has been produced for a human cell line CHM13, resolving the nearly complete haploid genome sequence of human being [245]. The achievement is largely reliant on the use of high fidelity PacBio long-read sequencing data (HiFi reads), in addition to the ultra-long ONT reads and Hi-C reads. This is not just a milestone of human genomics, but also has a profound impact on animal and plant genomic research by kicking off an ambitious journey to resolve the complete genome sequence of all known living organisms on this planet. In fact, shortly after the publication of the human T2T genome assembly, the nearly complete genome sequence of A. thaliana [246, 247], and the gap-free reference genomes of rice [248] and watermelon [249] were reported. The nearly complete genome sequences of plants reveal tandem repeats of satellite regions located in centromeres, and find new genes that weren’t accessible in previous versions of assemblies. T2T plant genome sequences will make huge impact on plant biology allowing researchers to grasp a full complement of genetic elements associated with various traits including growth, development, and physiology. Another technical challenge for plant genome assembly is, instead of generating a collapsed diploid assembly, the ability to produce a haploid-resolved assembly that separates the parental and maternal haplotypes, also known as genome phasing. The phased assembly allows understanding of mechanisms behind heterosis and allele-specific gene regulation that contributes to many plant biological processes. To date there are only a handful of plant genomes that have been assembled and phased, including tea [250], lychee [251], and pear [252]. Commonly, genome phasing is conducted through trio-binning sequencing where genomes of parents are used to untangle the two haploid genomes of a child, although the information of parents is often unavailable for medicinal plants. In such cases, recently genome assembly methods such as hifiasm [253] allow for the resolution of haploids by using HiFi reads and Hi-C data without relying on sequencing data of family trios.

Unraveling the regulatory mechanisms underlying PNP biosynthesis is beneficial to improving the quality of traditional Chinese medicine through genetic breeding and metabolic engineering. Genomics and metabolomics of plant tissues have shown that the genes of biosynthetic pathways for PNPs are often co-expressed in specific tissues [49, 139]. So far it remains elusive how PNP biosynthetic enzymes and pathways are regulated in such a spatial and temporal fine-tuned manner. Studies regarding the molecular mechanisms of transcriptional regulation related to biosynthetic pathways of PNPs are still limited, most of which being focused on the specialized medicinal plants with well-known PNPs, high-quality genomic data, and an established transgenic system. The expression of PNP biosynthetic pathways is controlled by epigenetic mechanisms such as chromatin topology dynamics as shown by Nützmann et al. in model plant Arabidopsis [254]. Thus, it will be exciting to reveal what roles 3D genomic architecture and organization play in regulating biosynthesis of flavonoids, terpenoids, and alkaloids in medicinal plants. In addition, several transcription factors have been identified to regulate the process over the years (Table 2), offering targets to enhance PNP accumulation potentially via overexpression (activator) or silencing (repressor). However, it remains a challenge to stably transform most medicinal plants and obtaining transgenic plants often takes years even if it does show promise of significantly improving the PNP level. Another caveat is that expressing an excess amount of any epigenetic or transcriptional regulators, usually entangled in complex regulatory networks, can potentially lead to undesired traits. Therefore, a systems biology approach is essential to tease apart the PNP-specific gene circuits for precision modulation of target traits. Moreover, the picture is still a blur in cell heterogeneity that accounts for the cell-type specific expression of biosynthetic enzymes, transporters, and gene regulators. Recently, cutting-edge technologies such as single-cell transcriptomics (scRNA-seq) and spatial-transcriptomics have been widely applied to mammalian and plant tissues to generate a cell atlas and identify cell types within these tissues. The use of such technologies in PNP research has yet to be reported as of the date of thisreview being written, although spatial metabolomics has been reported to investigate the distribution of plant metabolites [255, 256]. It will be very interesting to identify the specific cell-types expressing PNP biosynthetic genes using the scRNA-seq and spatio-transcriptomics, guiding a precise design and bioengineering of gene circuits for PNP improvement. The key challenge of the application of single-cell genomics in medicinal plants includes lack of high-quality reference genome and gene annotations, single-cell preparations, tissue and cell-type specific markers, and robust methods to integrative anlaysis of omic profiles at a single-cell level. Protocols and methods developed for human and mammalian samples are well in place but remain to be tested and optimized for medicinal plant studies.

Table 2.

A summary of transcription factors (TFs) regulating biosynthesis of plant natural products (PNP).

PNP types Species TFs Regulated genes PNPs
Flavonoids Citrus reticulata cv. suavissima CitERF32 [188], CitERF33 [188] CitCHIL1 Naringenin chalcone, (2S)-Naringenin
Antirrhinum majus Delila [189], Perilla [189] CHS Red anthocyanin
Gentiana triflora GtbHLH1 [190] GtF3’5’H, Gt5AT Gentiodelphin
Petunia hybrida AN1 [191] dfrA, Pmyb27 Anthocyanin
Myrica rubra MrbHLH1 [192] MrCHI, MrF3’H, MrDFR1, MrANS, MrUFGT Anthocyanin
Epimedium sagittatum EsMYBF1 [193] EsCHS, EsCHI, EsF3’H, EsF3H, EsFLS Anthocyanin, Kaempferol, Andquercetin
C. reticulata CsMYBF1 [194] Cs4CL, CsCHS, CsFLS Hydroxycinnamic acid
Ginkgo biloba GbMYBF2 [195] GbCHS, GbF3H GbPAL, GbFLS, GbANS, GbCHI Quercetin, Kaempferol, Anthocyanin
P. hybrida AN2 [191], AN4 [191] dfrA, Pmyb27 Anthocyanin
M. rubra MrMYB1 [192] MrCHI, MrF3’H, MrDFR1, MrANS, MrUFGT Anthocyanin
A. majus AmMYB305 [196], AmMYB340 [197], Rosea1 [198], Rosea2 [198], Venosa [198] PAL, CHI, F3H F3′H, FLS, DFR, UFGT, ANS Magenta anthocyanin
Gentiana triflora GtMYB3 [197] GtF3’5’H, Gt5AT Gentiodelphin
Ipomoea nil InMYB1 [199] CHS-D, CHI, F3H Anthocyanin
Medicago truncatula MTWD40–1 [200] DFR, ANS, LAR, ANR, UGT72L1, CHS Anthocyanin, Proanthocyanidin, Benzoic acids, Flavonols, Flavan-3-ols
I. nil InWDR1 [199] CHS-D, CHI, F3H Anthocyanin
Terpenoids Artemisia annua AaERF1 [201], AaERF2 [201], AaORA [202], TAR1 [203] ADS, CYP71AV1, DBR2, AaERF1 Artemisinin, Artemisinic acid, Dihydroartemisinic acid
Panax ginseng ERFs [204] DDS, CYP716A47, CYP716A53v2 Ginsenoside Rb1, Ginsenoside Rg1, Ginsenoside Re
A. annua AabZIP1 [205] ADS, CYP71AV1 Artemisinin
A. annua AaWRKY1 [206,207] ADS, CYP Artemisinin
A. annua AaNAC1 [208] ADS, DBR2, ALDH1 Artemisinin, Dihydroartemisinic acid
A. annua AabHLH1 [209] HMGR, ADS, CYP71AV1 Artemisinin
P. ginseng PGbHLHs [210] DXS, DXR, IspF, IspG, IspH, ACTT, HMGS, HMGR, MVK, PMK, FPS, SS, SE, CAS, OAS, PPDS, PPTS, DDS, β-AS Ginsenosides
Glycyrrhiza uralensis GubHLH3 [211] β-AS, CYP93E3, CYP72A566 Soyasapogenol B, Sophoradiol
Betula platyphylla BpbHLH9 [212] HMGR, FPS, SS, SE, BPY, BPW Betulinic acid, Oleanolic acid, Betulin
M. truncatula TSAR1 [213], TSAR2 [213] HMGR1, MAKIBISHI1 Hemolytic saponin, Nonhemolytic soyasaponi
A. annua AaEIN3 [214] ADS, DBR2, CYP71AV1, AaORA Artemisinin
A. annua AaMYB1 [215] FDS, ADS, CYP71AV1 Artemisinin, Dihydroartemisinic acid
P. ginseng PgMYB2 [216] PgDDS Ginsenoside
Mentha spicata MsMYB [217] MsGPPS.LSU Terpene
Salvia miltiorrhiza SmMYB36 [218], SmMYB9b [219] DXS1, DXS2, DXR, MCT, MDS, HDS, CMK, HDR1, GGPPS1, CPS1, KSL1, CYP76AHI, SmDXS2, SmDXR, SmGGPPS, SmKSL1 Phenolic acids, Tanshinones
Glycine max GmMYBZ2 [220] ZCT1, ASα, STR, ORCA3 Catharanthine
Pogostemon cablin PatSWC4 [221] PatPTS Patchoulol
P. cablin PatTHs [222], PatGT-1 [222] PatHMGR Patchoulol
Salvia miltiorrhiza SMJAZ1/2/3/9 [223, 224] interaction with MYC2 Tanshinones, Lithospermic acid B
M. spicata MsYABBY5 [225] MsWRKY75 Monoterpene (limonene and carvone)
Alkaloids Eschscholzia californica 20 EcAP2/ERF TFs [226] Ec6OMT, EcCYP719A5 Benzyl isoquinoline alkaloids
Catharanthus roseus ORCA1/2/3 [227229], CrERF5 [230], CrCR1 [231] STR, ASα, TDC, CPR, DXS, D4H, SLS, GES, SLS1, SGD, Redox1, SAT, PRX1, HL1, G10H, DAT Vindoline, Catharanthine, Vinblastine, Vincristine, Secologanin, Ajmalicine, Anhydrovinblastine, Serpentine
Nelumbo nucifera NnbHLH1 [232] NnNCS1 Benzyl isoquinoline alkaloid
Coptis japonica CjbHLH1 [233] TYDC, NCS, 6OMT, CNMT, CYP80B2, 40OMT, BBE, SMT, CYP719A1 Isoquinoline alkaloid
C. roseus CrMYC1, CrMYC2, CrBIS1, CrBIS2, CrBIS3 [234236] TDC, STR Terpenoid indole alkaloid
C. roseus ZCT1 [237], ZCT2 [237], ZCT3 [237] SSR2, C5-SD, HMGR, CAS Steroidal glycoalkaloids
Coptis japonica CjWRKY1 [238] TYDC, NCS, 6OMT, CNMT, CYP80B2, 40OMT, BBE, CYP719A1, SMT Berberine
C. roseus CrWRKY1 [239] TDC, AS, DXS, SLS, SGD Terpenoid indole alkaloid
C. roseus CrGBF1 [240], CrGBF2 [240] STR Terpenoid indole alkaloid
C. roseus CrGATA1 [241] T16H2, D4H, DAT, T3O, T3R Vindoline, Tabersonine
C. roseus CrBPF1 [242] D4H, DAT Terpenoid indole alkaloid
N. nucifera NnMYB6 [232], NnMYB12 [232], NnMYB113 [232] NnTYDC1, NnCYP80G, Nn7OMT2 Benzyl isoquinoline alkaloid
N. nucifera NnRAV1 [232] NnTYDC1, NnNCS1 Benzyl isoquinoline alkaloid

Last but not least, plant and microbe interactions also have a big influence on the profiles and abundance of PNPs. The importance of location and environment for growing TCM to their medicinal values and pharmaceutical properties has long been recognized and documented in traditional medicine scriptures. The difference could be down to a combination of factors such as the ecological environment, weather and perhaps the microbiota inhabiting in the soils where the TCM grows. Although the exact formula of microbial communities involved in modulating the PNP, as well as the mechanisms of regulation remain elusive, the association between microbiota and medicinal properties in plants has been suggested in several recent studies. An outstanding example of such association is reported in model plant A. thaliana which produces specific types of triterpenoids to electively modulate root bacteria in root microbiome [257]. Interestingly, isolation and re-inoculation of these bacteria in the roots can induce an increased production of these very PNPs. It remains to be studied how different types of microbes form microbial communities and signaling networks to interact with plants, either internally (endophytes) or externally (e.g. surface of roots and leaves), contribute to the accumulation of specific PNPs. Equally interesting is the regulation of plant microbiota by plant metabolites during plant–microbe interactions. Compared to model and crop plants, microbiome studies of medicinal plants have been quite limited. A combination of culture-based and culture-free microbiome analysis with functional metabolomics in medicinal plant rhizosphere and endophytes will help identify the association between plant microbiota and specialized metabolites. With this knowledge, it will be possible to modulate the production of special natural products in controlled settings such as greenhouse and plant factories in the future, much more efficient and effective than that which could be harvested from traditional authentic herb medicines.

Acknowledgements

This project is supported by the National Natural Science Foundation of China (Grant No. 31970317 and 32070368), Key Realm R&D Program of Guangdong Province, China (No. 2020B020221001), and the Natural Science Foundation of Shaanxi Province, China (No. 2020JZ-05). L.G. is also supported by a faculty startup package from Peking University Institute of Advanced Agricultural Sciences, and Yuandu Scholarship from municipal government of Weifang. The authors would like to thank anonymous reviewers for their comments and suggestions to improve the manuscript.

Conflict of interests

The authors declare that they have no conflict of interest.

Contributor Information

Li Guo, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Peking University Institute of Advanced Agricultural Sciences, Weifang, Shandong 261000, China.

Hui Yao, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.

Weikai Chen, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Peking University Institute of Advanced Agricultural Sciences, Weifang, Shandong 261000, China.

Xumei Wang, School of Pharmacy, Xi’an Jiaotong University, Xi’an 710061, China.

Peng Ye, State Key laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Laboratory For Lingnan Modern Agriculture, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China.

Zhichao Xu, College of Life Science, Northeast Forestry University, Harbin 150040, China.

Sisheng Zhang, State Key laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Laboratory For Lingnan Modern Agriculture, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China.

Hong Wu, State Key laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Laboratory For Lingnan Modern Agriculture, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China.

References

  • 1. Zaynab M, Fatima M, Abbas Set al. Role of secondary metabolites in plant defense against pathogens. Microb Pathog. 2018;124:198–202. [DOI] [PubMed] [Google Scholar]
  • 2. Adedeji AA, Babalola OO. Secondary metabolites as plant defensive strategy: a large role for small molecules in the near root region. Planta. 2020;252:61. [DOI] [PubMed] [Google Scholar]
  • 3. Li YQ, Kong D, Fu Yet al. The effect of developmental and environmental factors on secondary metabolites in medicinal plants. Plant Physiol Biochem. 2020;148:80–9. [DOI] [PubMed] [Google Scholar]
  • 4. Yang L, Wen KS, Ruan Xet al. Response of plant secondary metabolites to environmental factors. Molecules. 2018;23:762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wang SC, Alseekh S, Fernie ARet al. The structure and function of major plant metabolite modifications. Mol Plant. 2019;12:899–919. [DOI] [PubMed] [Google Scholar]
  • 6. Marchiosi R, dos Santos WD, Constantin RPet al. Biosynthesis and metabolic actions of simple phenolic acids in plants. Phytochem Rev. 2020;19:865–906. [Google Scholar]
  • 7. Koeduka T. The phenylpropene synthase pathway and its applications in the engineering of volatile phenylpropanoids in plants. Plant Biotechnol (Tokyo). 2014;31:401–7. [Google Scholar]
  • 8. Ye P, Liang S, Wang Xet al. Transcriptome analysis and targeted metabolic profiling for pathway elucidation and identification of a geraniol synthase involved in iridoid biosynthesis from Gardenia jasminoides. Ind Crop Prod. 2019;132:48–58. [Google Scholar]
  • 9. Šantić Ž, et al. The historical use of medicinal plants in traditional and scientific medicine. Psychiatr Danub. 2017;4:787–92. [PubMed] [Google Scholar]
  • 10. Lietava J, et al. Medicinal plants in a middle Paleolithic grave Shanidar IV? J Ethnopharmacol. 1992;35:263–6. [DOI] [PubMed] [Google Scholar]
  • 11. Norn S, Kruse PR, Kruse E. History of opium poppy and morphine. Dan Medicinhist Arbog. 2005;33:171–84. [PubMed] [Google Scholar]
  • 12. Sanchez-Ramos JR, et al. The rise and fall of tobacco as a botanical medicine. J Herb Med. 2020;22:100374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Warner KE, MacKay J. The global tobacco disease pandemic: nature, causes, and cures. Glob Public Health. 2006;1:65–86. [DOI] [PubMed] [Google Scholar]
  • 14. Jurna I. Serturner und Morphin? Eine historische vignette. Schmerz. 2003;17:280–3. [DOI] [PubMed] [Google Scholar]
  • 15. Achan J, Talisuna AO, Erhart Aet al. Quinine, an old anti-malarial drug in a modern world: role in the treatment of malaria. Malar J. 2011;10:144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Desborough MJR, Keeling DM. The aspirin story - from willow to wonder drug. Br J Haematol. 2017;177:674–83. [DOI] [PubMed] [Google Scholar]
  • 17. World Health Organization . World Malaria Report 2010. Geneva: WHO; 2010. [Google Scholar]
  • 18. Weaver BA. How taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25:2677–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Barnette KG, Gordon MS, Rodriguez Det al. Oral sabizabulin for high-risk, hospitalized adults with Covid-19: interim analysis. NEJM Evid. 2022;1:EVIDoa2200145. [DOI] [PubMed] [Google Scholar]
  • 20. Jorgensen SCJ, Kebriaei R, Dresser LD. Remdesivir: review of pharmacology, pre-clinical data, and emerging clinical experience for COVID-19. Pharmacotherapy 2020;40:659–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Mitjà O, Clotet B. Use of antiviral drugs to reduce COVID-19 transmission. Lancet Glob Health 2020;8:e639–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Wu CR, Liu Y, Yang Yet al. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm Sin B. 2020;10:766–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Liang SJ, et al. Study on flavonoid and bioactivity features of the pericarp of Citri Reticulatae ‘Chachi’ during storage. Arab J Chem. 2021;15:103653. [Google Scholar]
  • 24. Hu K, Guan WJ, Bi Yet al. Efficacy and safety of Lianhuaqingwen capsules, a repurposed Chinese herb, in patients with coronavirus disease 2019: a multicenter, prospective, randomized controlled trial. Phytomedicine. 2021;85:153242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Lichman BR. The scaffold-forming steps of plant alkaloid biosynthesis. Nat Pro Rep. 2021;38:103–29. [DOI] [PubMed] [Google Scholar]
  • 26. Yang CQ, Fang X, Wu XMet al. Transcriptional regulation of plant secondary MetabolismF. J Integr Plant Biol. 2012;54:703–12. [DOI] [PubMed] [Google Scholar]
  • 27. Beniddir MAB, Kang KB, Genta-Jouve Get al. Advances in decomposing complex metabolite mixtures using substructure-and network-based computational metabolomics approaches. Nat Prod Rep. 2021;38:1967–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Bohlmann J, Meyer-Gauen G, Croteau R. Plant terpenoid synthases: molecular biology and phylogenetic analysis. PNAS. 1998;95:4126–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Attieh J, Djiana R, Koonjul Pet al. Cloning and functional expression of two plant thiol ethyltransferases: a new class of enzymes involved in the biosynthesis of sulfur volatiles. Plant Mol Biol. 2002;50:511–21. [DOI] [PubMed] [Google Scholar]
  • 30. Ounaroon A, Decker G, Schmidt Jet al. (R, S)-Reticuline 7-O-methyltransferase and (R, S)-norcoclaurine 6-O-methyltransferase of Papaver somniferum - cDNA cloning and characterization of methyl transfer enzymes of alkaloid biosynthesis in opium poppy. Plant J. 2003;36:808–19. [DOI] [PubMed] [Google Scholar]
  • 31. Isabel DP. Integration of deep transcriptome and proteome analyses reveals the components of alkaloid metabolism in opium poppy cell cultures. BMC Plant Biol. 2010;10:252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Zeng JG, Liu Y, Liu Wet al. Integration of transcriptome, proteome and metabolism data reveals the alkaloids biosynthesis in Macleaya cordata and Macleaya microcarpa. PLoS One. 2013;8:e53409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kellner F, Kim J, Clavijo BJet al. Genome-guided investigation of plant natural product biosynthesis. Plant J. 2015;82:680–92. [DOI] [PubMed] [Google Scholar]
  • 34. Nett RS, Lau W, Sattely ES. Discovery and engineering of colchicine alkaloid biosynthesis. Nature. 2020;584:148–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Hong B, Grzech D, Caputi Let al. Biosynthesis of strychnine. Nature. 2022;607:617–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ajikumar PK, Xiao WH, Tyo KEJet al. Isoprenoid pathway optimization for taxol precursor overproduction in Escherichia coli. Science. 2010;330:70–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kitade Y, et al. Production of 4-hydroxybenzoic acid by an aerobic growth-arrested bioprocess using metabolically engineered Corynebacterium glutamicum. Appl Environ Microbiol. 2018;84:e02587–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Galanie S, Thodey K, Trenchard IJet al. Complete biosynthesis of opioids in yeast. Science. 2015;349:1095–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Wriessnegger T, Augustin P, Engleder Met al. Production of the sesquiterpenoid (+)-nootkatone by metabolic engineering of Pichia pastoris. Metab Eng. 2014;24:18–29. [DOI] [PubMed] [Google Scholar]
  • 40. Schultz BJ, Kim SY, Lau Wet al. Total biosynthesis for milligram-scale production of etoposide intermediates in a plant chassis. J Am Chem Soc. 2019;141:19231–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Cordero BF, Couso I, León Ret al. Enhancement of carotenoids biosynthesis in Chlamydomonas reinhardtii by nuclear transformation using a phytoene synthase gene isolated from chlorella zofingiensis. Appl Microbio Biotech. 2011;91:341–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Xue J, Niu YF, Huang Tet al. Genetic improvement of the microalga Phaeodactylum tricornutum for boosting neutral lipid accumulation. Metab Eng. 2015;27:1–9. [DOI] [PubMed] [Google Scholar]
  • 43. Wang R, Shu P, Zhang Cet al. Integrative analyses of metabolome and genome-wide transcriptome reveal the regulatory network governing flavor formation in kiwifruit (Actinidia chinensis). New Phytol. 2022;233:373–89. [DOI] [PubMed] [Google Scholar]
  • 44. Li Y, Chen Y, Zhou Let al. Microtom metabolic network: rewiring tomato metabolic regulatory network throughout the growth cycle. Mol Plant. 2020;13:1203–18. [DOI] [PubMed] [Google Scholar]
  • 45. Yang C, Shen S, Zhou Set al. Rice metabolic regulatory network spanning the entire life cycle. Mol Plant. 2022;15:258–75. [DOI] [PubMed] [Google Scholar]
  • 46. Chen F, Song Y, Li Xet al. Genome sequences of horticultural plants: past, present, and future. Hortic Res. 2019;6:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Liao B, Hu H, Xiao Set al. Global pharmacopoeia genome database is an integrated and mineable genomic database for traditional medicines derived from eight international pharmacopoeias. Sci China Life Sci. 2022;65:809–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Chen S, et al. Strategies of the study on herb genome program. Acta Pharm Sin B. 2010;45:807–12. [PubMed] [Google Scholar]
  • 49. Guo L, Winzer T, Yang Xet al. The opium poppy genome and morphinan production. Science. 2018;362:343–7. [DOI] [PubMed] [Google Scholar]
  • 50. Kang M, et al. A chromosome-level Camptotheca acuminata genome assembly provides insights into the evolutionary origin of camptothecin biosynthesis. Nat Commun. 2021;12:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Zhao Q, Yang J, Cui MYet al. The reference genome sequence of Scutellaria baicalensis provides insights into the evolution of wogonin biosynthesis. Mol Plant. 2019;12:935–50. [DOI] [PubMed] [Google Scholar]
  • 52. Jiang Z, Tu L, Yang Wet al. The chromosome-level reference genome assembly for Panax notoginseng and insights into ginsenoside biosynthesis. Plant Commun. 2021;2:100113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Tu L, Ping S, Zhang Zet al. Genome of Tripterygium wilfordii and identification of cytochrome P450 involved in triptolide biosynthesis. Nat Commun. 2020;11:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Song Z, Lin C, Xing Pet al. A highquality reference genome sequence of salvia miltiorrhiza provides insights into tanshinone synthesis in its red rhizomes. Plant Genome. 2020;13:e20041. [DOI] [PubMed] [Google Scholar]
  • 55. Cheng J, Wang X, Liu Xet al. Chromosome-level genome of Himalayan yew provides insights into the origin and evolution of the paclitaxel biosynthetic pathway. Mol Plant. 2021;14:1199–209. [DOI] [PubMed] [Google Scholar]
  • 56. He S, Dong X, Zhang Get al. High quality genome of Erigeron breviscapus provides a reference for herbal plants in Asteraceae. Mol Ecol Resour. 2021;21:153–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Yang X, Gao S, Guo Let al. Three chromosome-scale Papaver genomes reveal punctuated patchwork evolution of the morphinan and noscapine biosynthesis pathway. Nat Commun. 2021;12:6030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Caputi L, Franke J, Farrow SCet al. Missing enzymes in the biosynthesis of the anticancer drug vinblastine in Madagascar periwinkle. Science. 2018;360:1235–9. [DOI] [PubMed] [Google Scholar]
  • 59. Fan G, Liu X, Sun Set al. The chromosome level genome and genome-wide association study for the agronomic traits of Panax Notoginseng. iScience. 2020;23:101538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Song X, Sun P, Yuan Jet al. The celery genome sequence reveals sequential paleo-polyploidizations, karyotype evolution and resistance gene reduction in apiales. Plant Biotechnol J. 2021;19:731–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Wang Y, et al. Deletion and tandem duplications of biosynthetic genes drive the diversity of triterpenoids in Aralia elata. Nat Commun. 2022;13:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Song X, Wang J, Li Net al. Deciphering the high-quality genome sequence of coriander that causes controversial feelings. Plant Biotechnol J. 2020;18:1444–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Wang Z, et al. Reshuffling of the ancestral core-eudicot genome shaped chromatin topology and epigenetic modification in Panax. Nat Commun. 2022;13:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Sun X, Zhu S, Li Net al. A chromosome-level genome assembly of garlic (Allium sativum) provides insights into genome evolution and allicin biosynthesis. Mol Plant. 2020;13:1328–39. [DOI] [PubMed] [Google Scholar]
  • 65. Han B, Jing Y, Dai Jet al. A chromosome-level genome assembly of dendrobium huoshanense using long reads and hi-C data. Genome Biol Evol. 2020;12:2486–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Niu Z, Zhu F, Fan Yet al. The chromosome-level reference genome assembly for Dendrobium officinale and its utility of functional genomics research and molecular breeding study. Acta Pharm Sin B. 2021;11:2080–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Bae E, An C, Kang MJet al. Chromosome-level genome assembly of the fully mycoheterotrophic orchid Gastrodia elata. G3. 2022;12:jkab433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Fan W, Wang S, Wang Het al. The genomes of chicory, endive, great burdock and yacon provide insights into Asteraceae palaeo-polyploidization history and plant inulin production. Mol Ecol Resour. 2022;22:3124–40. [DOI] [PubMed] [Google Scholar]
  • 69. Liao B, Shen X, Xiang Let al. Allele-aware chromosome-level genome assembly of Artemisia annua reveals the correlation between ADS expansion and artemisinin yield. Mol Plant. 2022;15:1310–28. [DOI] [PubMed] [Google Scholar]
  • 70. Miao Y, Luo D, Zhao Tet al. Genome sequencing reveals chromosome fusion and extensive expansion of genes related to secondary metabolism in Artemisia argyi. Plant Biotechnol J. 2022;20:1902–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Wu Z, Liu H, Zhan Wet al. The chromosome-scale reference genome of safflower (Carthamus tinctorius) provides insights into linoleic acid and flavonoid biosynthesis. Plant Biotechnol J. 2021;19:1725–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Song C, Liu Y, Song Aet al. The chrysanthemum nankingense genome provides insights into the evolution and diversification of chrysanthemum flowers and medicinal traits. Mol Plant. 2018;11:1482–91. [DOI] [PubMed] [Google Scholar]
  • 73. Jia Y, Chen S, Chen Wet al. A chromosome-level reference genome of Chinese balloon flower (Platycodon grandiflorus). Front Genet. 2022;13:869784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Kang M, Wu H, Yang Qet al. A chromosome-scale genome assembly of Isatis indigotica, an important medicinal plant used in traditional Chinese medicine. Hortic Res. 2020;7:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zhang J, Tian Y, Yan Let al. Genome of plant maca (Lepidium meyenii) illuminates genomic basis for high-altitude adaptation in the Central Andes. Mol Plant. 2016;9:1066–77. [DOI] [PubMed] [Google Scholar]
  • 76. Li C, et al. Assembly and annotation of a draft genome of the medicinal plant Polygonum cuspidatum. Front Plant Sci. 2019;10:1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Yang Y, Sun P, Lv Let al. Prickly waterlily and rigid hornwort genomes shed light on early angiosperm evolution. Nat Plants. 2020;6:215–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Xie D, Xu Y, Wang Jet al. The wax gourd genomes offer insights into the genetic diversity and ancestral cucurbit karyotype. Nat Commun. 2019;10:5158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Huang D, Ming R, Xu Set al. Chromosome-level genome assembly of Gynostemma pentaphyllum provides insights into gypenoside biosynthesis. DNA Res. 2021;28:dsab018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Wu H, Zhao G, Gong Het al. A high-quality sponge gourd (Luffa cylindrica) genome. Hortic Res. 2020;7:128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Cui J, Yang Y, Luo Set al. Whole-genome sequencing provides insights into the genetic diversity and domestication of bitter gourd (Momordica spp.). Hortic Res. 2020;7:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Xia M, et al. Improved de novo genome assembly and analysis of the Chinese cucurbit Siraitia grosvenorii, also known as monk fruit or luo-han-guo. Gigascience. 2018;7:giy067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Pu X, Li Z, Tian Yet al. The honeysuckle genome provides insight into the molecular mechanism of carotenoid metabolism underlying dynamic flower coloration. New Phytol. 2020;227:930–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Akagi T, Shirasawa K, Nagasaki Het al. The persimmon genome reveals clues to the evolution of a lineage-specific sex determination system in plants. PLoS Genet. 2020;16:e1008566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Xia E, Zhang HB, Sheng Jet al. The tea tree genome provides insights into tea flavor and independent evolution of caffeine biosynthesis. Mol Plant. 2017;10:866–77. [DOI] [PubMed] [Google Scholar]
  • 86. Mochida K, Sakurai T, Seki Het al. Draft genome assembly and annotation of Glycyrrhiza uralensis, a medicinal legume. Plant J. 2017;89:181–94. [DOI] [PubMed] [Google Scholar]
  • 87. Kang S, et al. Genome-enabled discovery of anthraquinone biosynthesis in Senna tora. Nat Commun. 2020;11:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Qin S, Wu L, Wei Ket al. A draft genome for Spatholobus suberectus. Sci Data. 2019;6:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Li Y, Wei H, Yang Jet al. High-quality de novo assembly of the Eucommia ulmoides haploid genome provides new insights into evolution and rubber biosynthesis. Hortic Res. 2020;7:183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Hoopes GM, Hamilton JP, Kim Jet al. Genome assembly and annotation of the medicinal PlantCalotropis gigantea, a producer of anticancer and antimalarial cardenolides. G3. 2018;8:385–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Liu Y, Tang Q, Cheng Pet al. Whole-genome sequencing and analysis of the Chinese herbal plant Gelsemium elegans. Acta Pharm Sin B. 2020;10:374–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Xu Z, et al. Tandem gene duplications drive divergent evolution of caffeine and crocin biosynthetic pathways in plants. BMC Biol. 2020;18:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Wang J, Xu S, Mei Yet al. A high-quality genome assembly of Morinda officinalis, a famous native southern herb in the Lingnan region of southern China. Hortic Res. 2021;8:135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Rai A, et al. Chromosome-level genome assembly of Ophiorrhiza pumila reveals the evolution of camptothecin biosynthesis. Nat Commun. 2021;12:1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Liang Y, Chen S, Wei Ket al. Chromosome level genome assembly of Andrographis paniculata. Front Genet. 2020;11:701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Vining KJ, Johnson SR, Ahkami Aet al. Draft genome sequence of Mentha longifolia and development of resources for mint cultivar improvement. Mol Plant. 2017;10:323–39. [DOI] [PubMed] [Google Scholar]
  • 97. Bornowski N, Hamilton JP, Liao Pet al. Genome sequencing of four culinary herbs reveals terpenoid genes underlying chemodiversity in the Nepetoideae. DNA Res. 2020;27:dsaa016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Zhang Y, et al. Incipient diploidization of the medicinal plant Perilla within 10,000 years. Nat Commun. 2021;12:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Shen Y, Li W, Zeng Yet al. Chromosome-level and haplotype-resolved genome provides insight into the tetraploid hybrid origin of patchouli. Nat Commun. 2022;13:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Zheng X, Chen D, Chen Bet al. Insights into salvianolic acid B biosynthesis from chromosome-scale assembly of the salvia bowleyana genome. Acta Bot Sin. 2021;63:1309–23. [DOI] [PubMed] [Google Scholar]
  • 101. Jia K, Liu H, Zhang RGet al. Chromosome-scale assembly and evolution of the tetraploid Salvia splendens (Lamiaceae) genome. Hortic Res. 2021;8:177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Xu Z, Gao R, Pu Xet al. Comparative genome analysis of Scutellaria baicalensis and Scutellaria barbata reveals the evolution of active flavonoid biosynthesis. Genom Proteom Bioinform 2020;18:230–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Li L, Cushman SA, He YXet al. Genome sequencing and population genomics modeling provide insights into the local adaptation of weeping forsythia. Hortic Res. 2020;7:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Li M, Zhang D, Gao Qet al. Genome structure and evolution of Antirrhinum majus L. Nat Plants. 2019;5:174–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Shang J, et al. The chromosome-level wintersweet (Chimonanthus praecox) genome provides insights into floral scent biosynthesis and flowering in winter. Genome Biol. 2020;21:1–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Chaw SM, Liu YC, Wu YWet al. Stout camphor tree genome fills gaps in understanding of flowering plant genome evolution. Nat Plants. 2019;5:63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Li J, Lv M, Du Let al. An enormous Paris polyphylla genome sheds light on genome size evolution and polyphyllin biogenesis. BioRxiv 2020. preprint: not peer reviewed. [Google Scholar]
  • 108. Chan AP, Crabtree J, Zhao Qet al. Draft genome sequence of the oilseed species Ricinus communis. Nat Biotechnol. 2010;28:951–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Zhou W, et al. Whole-genome sequence data of Hypericum perforatum and functional characterization of melatonin biosynthesis by N-acetylserotonin O-methyltransferase. J Pineal Res. 2021;70:e12709. [DOI] [PubMed] [Google Scholar]
  • 110. Wang Z, Hobson N, Galindo Let al. The genome of flax (Linum usitatissimum) assembled de novo from short shotgun sequence reads. Plant J. 2012;72:461–73. [DOI] [PubMed] [Google Scholar]
  • 111. Xia Z, Huang D, Zhang Set al. Chromosome-scale genome assembly provides insights into the evolution and flavor synthesis of passion fruit (Passiflora edulis Sims). Hortic Res. 2021;8:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Nong W, Law STS, Wong AYPet al. Chromosomal-level reference genome of the incense tree Aquilaria sinensis. Mol Ecol Resour. 2020;20:971–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Qin G, Xu C, Ming Ret al. The pomegranate (Punica granatum L.) genome and the genomics of punicalagin biosynthesis. Plant J. 2017;91:1108–28. [DOI] [PubMed] [Google Scholar]
  • 114. Zhang L, Chen F, Zhang Xet al. The water lily genome and the early evolution of flowering plants. Nature. 2020;577:79–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Cui X, Meng F, Pan Xet al. Chromosome-level genome assembly of Aristolochia contorta provides insights into the biosynthesis of benzylisoquinoline alkaloids and aristolochic acids. Hortic Res. 2022;9:uhac005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Hu L, Xu Z, Wang Met al. The chromosome-scale reference genome of black pepper provides insight into piperine biosynthesis. Nat Commun. 2019;10:4702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Guo C, Wang Y, Yang Aet al. The Coix genome provides insights into Panicoideae evolution and papery hull domestication. Mol Plant. 2020;13:309–20. [DOI] [PubMed] [Google Scholar]
  • 118. Shi T, Rahmani RS, Gugger PFet al. Distinct expression and methylation patterns for genes with different fates following a single whole-genome duplication in flowering plants. Mol Biol Evol. 2020;37:2394–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Liu X, Liu Y, Huang Pet al. The genome of medicinal plant Macleaya cordata provides new insights into benzylisoquinoline alkaloids metabolism. Mol Plant. 2017;10:975–89. [DOI] [PubMed] [Google Scholar]
  • 120. Liu Y, et al. Analysis of the Coptis chinensis genome reveals the diversification of protoberberine-type alkaloids. Nat Commun. 2021;12:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Grassa CJ, Wenger JP, Dabney Cet al. A complete cannabis chromosome assembly and adaptive admixture for elevated cannabidiol (CBD) content. New Phytol. 2021;230:1665–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Peng X, Liu H, Chen Pet al. A chromosome-scale genome assembly of paper mulberry (Broussonetia papyrifera) provides new insights into its forage and papermaking usage. Mol Plant. 2019;12:661–77. [DOI] [PubMed] [Google Scholar]
  • 123. Xuan Y, Ma B, Li Det al. Chromosome restructuring and number change during the evolution of Morus notabilis and Morus alba. Hortic Res. 2022;9:uhab030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Liu M, Zhao J, Cai QLet al. The complex jujube genome provides insights into fruit tree biology. Nat Commun. 2014;5:5315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Jiang S, An H, Xu Fet al. Chromosome-level genome assembly and annotation of the loquat (Eriobotrya japonica) genome. GigaScience. 2020;9:giaa015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Raymond O, Gouzy J, Just Jet al. The Rosa genome provides new insights into the domestication of modern roses. Nat Genet. 2018;50:772–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Luan M, Jian JB, Chen Pet al. Draft genome sequence of ramie, Boehmeria nivea (L.) Gaudich. Mol Eco Resour. 2018;18:639–45. [DOI] [PubMed] [Google Scholar]
  • 128. Zeng L, Tu XL, Dai Het al. Whole genomes and transcriptomes reveal adaptation and domestication of pistachio. Genome Biol. 2019;20:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Ma Q, Sun T, Li Set al. The Acer truncatum genome provides insights into nervonic acid biosynthesis. Plant J. 2020;104:662–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Liang Q, Li H, Li Set al. The genome assembly and annotation of yellowhorn (Xanthoceras sorbifolium Bunge). GigaScience. 2019;8:giz071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Fu Y, Li L, Hao Set al. Draft genome sequence of the Tibetan medicinal herb Rhodiola crenulata. Gigascience. 2017;6:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Hoshino A, Jayakumar V, Nitasaka Eet al. Genome sequence and analysis of the Japanese morning glory Ipomoea nil. Nat Commun. 2016;7:13295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. De-la-Cruz IM, Hallab A, Olivares-Pinto Uet al. Genomic signatures of the evolution of defence against its natural enemies in the poisonous and medicinal plant Datura stramonium (Solanaceae). Sci Rep. 2021;11:882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Yang P, Zhao HY, Wei JSet al. Chromosome-level genome assembly and functional characterization of terpene synthases provide insights into the volatile terpenoid biosynthesis of Wurfbainia villosa. Plant J. 2022. [DOI] [PubMed] [Google Scholar]
  • 135. Li H, Wu L, Dong Zet al. Haplotype-resolved genome of diploid ginger (Zingiber officinale) and its unique gingerol biosynthetic pathway. Hortic Res. 2021;8:189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Xu Z, Xin T, Bartels Det al. Genome analysis of the ancient tracheophyte Selaginella tamariscina reveals evolutionary features relevant to the acquisition of desiccation tolerance. Mol Plant. 2018;11:983–94. [DOI] [PubMed] [Google Scholar]
  • 137. Liu H, Wang X, Wang Get al. The nearly complete genome of Ginkgo biloba illuminates gymnosperm evolution. Nat Plants. 2021;7:748–56. [DOI] [PubMed] [Google Scholar]
  • 138. Wan T, Liu ZM, Li LFet al. A genome for gnetophytes and early evolution of seed plants. Nat Plants. 2018;4:82–9. [DOI] [PubMed] [Google Scholar]
  • 139. Xiong X, Gou J, Liao Qet al. The Taxus genome provides insights into paclitaxel biosynthesis. Nat Plants. 2021;7:1026–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Field B, Osbourn AE. Metabolic diversification-independent assembly of operon-like gene clusters in different plants. Science. 2008;320:543–7. [DOI] [PubMed] [Google Scholar]
  • 141. Field B, Fiston-Lavier AS, Kemen Aet al. Formation of plant metabolic gene clusters within dynamic chromosomal regions. PNAS. 2011;108:16116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Mao LF, Kawaide H, Higuchi Tet al. Genomic evidence for convergent evolution of gene clusters for momilactone biosynthesis in land plants. PNAS. 2020;117:12472–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Frey M, Chomet P, Glawischnig Eet al. Analysis of a chemical plant defense mechanism in grasses. Science. 1997;277:696–9. [DOI] [PubMed] [Google Scholar]
  • 144. Itkin M, Heinig U, Tzfadia Oet al. Biosynthesis of antinutritional alkaloids in Solanaceous crops is mediated by clustered genes. Science. 2013;341:175–9. [DOI] [PubMed] [Google Scholar]
  • 145. Jeon JE, Kim JG, Fischer CRet al. A pathogen-responsive gene cluster for highly modified fatty acids in tomato. Cell. 2020;180:176–187.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Kautsar SA, Suarez Duran HG, Blin Ket al. PlantiSMASH: automated identification, annotation and expression analysis of plant biosynthetic gene clusters. Nucleic Acids Res. 2017;45:W55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Töpfer N, Fuchs LM, Aharoni A. The PhytoClust tool for metabolic gene clusters discovery in plant genomes. Nucleic Acids Res. 2017;45:7049–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Schläpfer P, Zhang P, Wang Cet al. Genome-wide prediction of metabolic enzymes, pathways, and gene clusters in plants. Plant Physiol. 2017;173:2041–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Winzer T, Gazda V, He Zet al. A Papaver somniferum 10-gene cluster for synthesis of the anticancer alkaloid noscapine. Science. 2012;336:1704–8. [DOI] [PubMed] [Google Scholar]
  • 150. Matsuba Y, Zi J, Jones ADet al. Biosynthesis of the diterpenoid lycosantalonol via nerylneryl diphosphate in Solanum lycopersicum. PLoS One. 2015;10:e0119302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Kong D, Li S, Smolke CD. Discovery of a previously unknown biosynthetic capacity of naringenin chalcone synthase by heterologous expression of a tomato gene cluster in yeast. Sci Adv. 2020;6:eabd1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Polturak G, Dippe M, Stephenson MJet al. Pathogen-induced biosynthetic pathways encode defense-related molecules in bread wheat. PNAS. 2022;119:e2123299119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Wu D, Jiang B, Ye CYet al. Horizontal transfer and evolution of the biosynthetic gene cluster for benzoxazinoids in plants. Plant Commun. 2022;3:100320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Ma J, Wang S, Zhu Xet al. Major episodes of horizontal gene transfer drove the evolution of land plants. Mol Plant. 2022;15:857–71. [DOI] [PubMed] [Google Scholar]
  • 155. Fan P, Wang P, Lou YRet al. Evolution of a plant gene cluster in Solanaceae and emergence of metabolic diversity. elife. 2020;9:e56717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Kotopka BJ, Smolke CD. Production of the cyanogenic glycoside dhurrin in yeast. Metab Eng Commun. 2019;9:e00092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Clevenger KD, Bok JW, Ye Ret al. A scalable platform to identify fungal secondary metabolites and their gene clusters. Nat Chem Biol. 2017;13:895–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Li Q, Ramasamy S, Singh Pet al. Gene clustering and copy number variation in alkaloid metabolic pathways of opium poppy. Nat Commun. 2020;11:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Wetzstein HY, Porter JA, Janick Jet al. Selection and clonal propagation of high artemisinin genotypes of Artemisia annua. Front Plant Sci. 2018;9:358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Graham IA, Besser K, Blumer Set al. The genetic map of Artemisia annua L. identifies loci affecting yield of the antimalarial drug artemisinin. Science. 2010;327:328–31. [DOI] [PubMed] [Google Scholar]
  • 161. Celik I, Camci H, Kose Aet al. Molecular genetic diversity and association mapping of morphine content and agronomic traits in Turkish opium poppy (Papaver somniferum) germplasm. Mol Breeding. 2016;36:46. [Google Scholar]
  • 162. Naim-Feil E, et al. The characterization of key physiological traits of medicinal cannabis (Cannabis sativa L.) as a tool for precision breeding. BMC Plant Biol. 2021;21:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Westfall PJ, Pitera DJ, Lenihan JRet al. Production of amorphadiene in yeast, and its conversion to dihydroartemisinic acid, precursor to the antimalarial agent artemisinin. PNAS. 2012;109:E111–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Gao CX. Genome engineering for crop improvement and future agriculture. Cell. 2021;184:1621–35. [DOI] [PubMed] [Google Scholar]
  • 165. Alagoz Y, et al. Manipulating the biosynthesis of bioactive compound alkaloids for next-generation metabolic engineering in opium poppy using CRISPR-Cas 9 genome editing technology. Sci Rep. 2016;6:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Li B, Cui G, Shen Get al. Targeted mutagenesis in the medicinal plant salvia miltiorrhiza. Sci Rep. 2017;7:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Kui L, Chen H, Zhang Wet al. Building a genetic manipulation tool box for orchid biology: identification of constitutive promoters and application of CRISPR/Cas9 in the orchid, Dendrobium officinale. Front Plant Sci. 2017;7:2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Jiang WZ, Henry IM, Lynagh PGet al. Significant enhancement of fatty acid composition in seeds of the allohexaploid, Camelina sativa, using CRISPR/Cas9 gene editing. Plant Biotechnol J. 2017;15:648–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Zhu X, Liu X, Liu Tet al. Synthetic biology of plant natural products: from pathway elucidation to engineered biosynthesis in plant cells. Plant Commun. 2021;2:100229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Ye XD, al-Babili S, Klöti Aet al. Engineering the provitamin a (beta-carotene) biosynthetic pathway into (carotenoid-free) rice endosperm. Science. 2000;287:303–5. [DOI] [PubMed] [Google Scholar]
  • 171. Zhu Q, Yu S, Zeng Det al. Development of “purple endosperm rice” by engineering anthocyanin biosynthesis in the endosperm with a high-efficiency transgene stacking system. Mol Plant. 2017;10:918–29. [DOI] [PubMed] [Google Scholar]
  • 172. Zhu C, Naqvi S, Breitenbach Jet al. Combinatorial genetic transformation generates a library of metabolic phenotypes for the carotenoid pathway in maize. PNAS. 2008;105:18232–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Fuentes P, Zhou F, Erban Aet al. A new synthetic biology approach allows transfer of an entire metabolic pathway from a medicinal plant to a biomass crop. elife. 2016;5:e13664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. De La Peña R, Sattely ES. Rerouting plant terpene biosynthesis enables momilactone pathway elucidation. Nat Chem Biol. 2021;17:205–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Li J, Mutanda I, Wang Ket al. Chloroplastic metabolic engineering coupled with isoprenoid pool enhancement for committed taxanes biosynthesis in Nicotiana benthamiana. Nat Commun. 2019;10:4850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Farag S, Kayser O. Cannabinoids production by hairy root cultures of Cannabis sativa L. J Plant Sci USA. 2015;6:1874–84. [Google Scholar]
  • 177. Kim YK, Kim YB, Uddin MRet al. Enhanced triterpene accumulation in Panax ginseng hairy roots overexpressing mevalonate-5-pyrophosphate decarboxylase and farnesyl pyrophosphate synthase. ACS Synth Biol. 2014;3:773–9. [DOI] [PubMed] [Google Scholar]
  • 178. Singh S, Pandey P, Akhtar MQet al. A new synthetic biology approach for the production of curcumin and its glucoside in Atropa belladonna hairy roots. J Biotechnol. 2021;328:23–33. [DOI] [PubMed] [Google Scholar]
  • 179. Martínez-Márquez A, Morante-Carriel JA, Ramírez-Estrada Ket al. Production of highly bioactive resveratrol analogues pterostilbene and piceatannol in metabolically engineered grapevine cell cultures. Plant Biotechnol J. 2016;14:1813–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Schuurink R, Tissier A. Glandular trichomes: micro-organs with model status? New Phytol. 2020;225:2251–66. [DOI] [PubMed] [Google Scholar]
  • 181. Kortbeek RW, Xu J, Ramirez Aet al. Engineering of tomato glandular trichomes for the production of specialized metabolites. Method Enzymol. 2016;576:305–31. [DOI] [PubMed] [Google Scholar]
  • 182. Xu X, Liu Y, Du Get al. Microbial chassis development for natural product biosynthesis. Trends Biotechnol. 2020;38:779–96. [DOI] [PubMed] [Google Scholar]
  • 183. Paddon CJ, Westfall PJ, Pitera DJet al. High-level semi-synthetic production of the potent antimalarial artemisinin. Nature. 2013;496:528–32. [DOI] [PubMed] [Google Scholar]
  • 184. Luo X, Reiter MA, d’Espaux Let al. Complete biosynthesis of cannabinoids and their unnatural analogues in yeast. Nature. 2019;567:123–6. [DOI] [PubMed] [Google Scholar]
  • 185. Li Y, Li S, Thodey Ket al. Complete biosynthesis of noscapine and halogenated alkaloids in yeast. PNAS. 2018;115:E3922–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Tsuruta H, Paddon CJ, Eng Det al. High-level production of amorpha-4, 11-diene, a precursor of the antimalarial agent artemisinin, in Escherichia coli. PLoS One. 2009;4:e4489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Osorio-Montalvo P, Sáenz-Carbonell L, de-la-Peña C. 5-Azacytidine: a promoter of epigenetic changes in the quest to improve plant somatic embryogenesis. Int J Mol Sci. 2018;19:3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Zhao C, Liu X, Gong Qet al. Three AP2/ERF family members modulate flavonoid synthesis by regulating type IV chalcone isomerase in citrus. Plant Biotechnol J. 2021;19:671–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Goodrich J, Carpenter R, Coen ES. A common gene regulates pigmentation pattern in diverse plant species. Cell. 1992;68:955–64. [DOI] [PubMed] [Google Scholar]
  • 190. Nakatsuka T, Haruta KS, Pitaksutheepong Cet al. Identification and characterization of R2R3-MYB and bHLH transcription factors regulating anthocyanin biosynthesis in gentian flowers. Plant Cell Physiol. 2008;49:1818–29. [DOI] [PubMed] [Google Scholar]
  • 191. Spelt C, Quattrocchio F, Mol JNMet al. anthocyanin1of petunia encodes a basic helix-loop-helix protein that directly activates transcription of structural anthocyanin genes. Plant Cell. 2000;12:1619–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Liu X, Yin XR, Allan ACet al. The role of MrbHLH1 and MrMYB1 in regulating anthocyanin biosynthetic genes in tobacco and Chinese bayberry (Myrica rubra) during anthocyanin biosynthesis. Plant Cell Tiss Org. 2013;115:285–98. [Google Scholar]
  • 193. Huang W, et al. A R2R3-MYB transcription factor regulates the flavonol biosynthetic pathway in a traditional Chinese medicinal plant, Epimedium sagittatum. Front Plant Sci. 2016;7:1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Liu C, Long J, Zhu Ket al. Characterization of a citrus R2R3-MYB transcription factor that regulates the flavonol and hydroxycinnamic acid biosynthesis. Sci Rep. 2016;26:2535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Xu F, Ning Y, Zhang Wet al. An R2R3-MYB transcription factor as a negative regulator of the flavonoid biosynthesis pathway in Ginkgo biloba. Funct Integr Genomics. 2014;14:177–89. [DOI] [PubMed] [Google Scholar]
  • 196. Sablowski RW, Moyano E, Culianez-Macia FAet al. A flower-specific Myb protein activates transcription of phenylpropanoid biosynthetic genes. EMBO J. 1994;13:128–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Moyano E, Martínez-Garcia JF, Martin C. Apparent redundancy in myb gene function provides gearing for the control of flavonoid biosynthesis in antirrhinum flowers. Plant Cell. 1996;8:1519–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Schwinn K, Venail J, Shang Yet al. A small family of MYB-regulatory genes controls floral pigmentation intensity and patterning in the genus antirrhinum. Plant Cell. 2006;18:831–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Morita Y, Saitoh M, Hoshino Aet al. Isolation of cDNAs for R2R3-MYB, bHLH and WDR transcriptional regulators and identification of c and ca mutations conferring white flowers in the Japanese morning glory. Plant Cell Physiol. 2006;47:457–70. [DOI] [PubMed] [Google Scholar]
  • 200. Pang Y, Wenger JP, Saathoff Ket al. A WD40 repeat protein from Medicago truncatula is necessary for tissue-specific anthocyanin and proanthocyanidin biosynthesis but not for trichome development. Plant Physiol. 2009;151:1114–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Yu ZX, Li JX, Yang CQet al. The jasmonate-responsive AP2/ERF transcription factors AaERF1 and AaERF2 positively regulate artemisinin biosynthesis in Artemisia annua L. Mol Plant. 2012;5:353–65. [DOI] [PubMed] [Google Scholar]
  • 202. Lu X, Zhang L, Zhang Fet al. AaORA, a trichome-specific AP2/ERF transcription factor of Artemisia annua, is a positive regulator in the artemisinin biosynthetic pathway and in disease resistance to Botrytis cinerea. New Phytol. 2013;198:1191–202. [DOI] [PubMed] [Google Scholar]
  • 203. Tan H, Xiao L, Gao Set al. Trichome and artemisinin regulator 1 is required for trichome development and artemisinin biosynthesis in Artemisia annua. Mol Plant. 2015;8:1396–411. [DOI] [PubMed] [Google Scholar]
  • 204. Zhang J, et al. Expression analysis of transcription factor ERF gene family of Panax ginseng. China J Chin Mater Med. 2020;45:2515–22. [DOI] [PubMed] [Google Scholar]
  • 205. Zhang F, Fu X, Lv Zet al. A basic leucine zipper transcription factor, AabZIP1, connects abscisic acid signaling with artemisinin biosynthesis in Artemisia annua. Mol Plant. 2015;8:163–75. [DOI] [PubMed] [Google Scholar]
  • 206. Ma D, Pu G, Lei Cet al. Isolation and characterization of AaWRKY1, an Artemisia annua transcription factor that regulates the amorpha-4,11-diene synthase gene, a key gene of artemisinin biosynthesis. Plant Cell Physiol. 2009;50:2146–61. [DOI] [PubMed] [Google Scholar]
  • 207. Jiang W, Fu X, Pan Qet al. Overexpression of AaWRKY1 leads to an enhanced content of artemisinin in Artemisia annua. Biomed Res Int. 2016;2016:7314971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Lv Z, Wang S, Zhang Fet al. Overexpression of a novel NAC domain-containing transcription factor gene (AaNAC1) enhances the content of artemisinin and increases tolerance to drought and botrytis cinerea in Artemisia annua. Plant Cell Physiol. 2016;57:1961–71. [DOI] [PubMed] [Google Scholar]
  • 209. Ji Y, Xiao J, Shen Yet al. Cloning and characterization of AabHLH1, a bHLH transcription factor that positively regulates artemisinin biosynthesis in Artemisia annua. Plant Cell Physiol. 2014;55:1592–604. [DOI] [PubMed] [Google Scholar]
  • 210. Chu Y, Xiao S, Su Het al. Genome-wide characterization and analysis of bHLH transcription factors in Panax ginseng. Acta Pharm Sin B. 2018;8:666–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Tamura K, Yoshida K, Hiraoka Yet al. The basic helix-loop-helix transcription factor GubHLH3 positively regulates soyasaponin biosynthetic genes in Glycyrrhiza uralensis. Plant Cell Physiol. 2018;59:778–91. [DOI] [PubMed] [Google Scholar]
  • 212. Yin J, Li X, Zhan Yet al. Cloning and expression of BpMYC4 and BpbHLH9 genes and the role of BpbHLH9 in triterpenoid synthesis in birch. BMC Plant Biol. 2017;17:214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Mertens J, Pollier J, vanden Bossche Ret al. The bHLH transcription factors TSAR1 and TSAR2 regulate triterpene saponin biosynthesis in Medicago truncatula. Plant Physiol. 2016;170:194–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Tang Y, Li L, Yan Tet al. AaEIN3 mediates the downregulation of artemisinin biosynthesis by ethylene signaling through promoting leaf senescence in Artemisia annua. Front Plant Sci. 2018;9:413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Matías-Hernández L, Jiang W, Yang Ket al. AaMYB1 and its orthologue AtMYB61 affect terpene metabolism and trichome development in Artemisia annua and Arabidopsis thaliana. Plant J. 2017;90:520–34. [DOI] [PubMed] [Google Scholar]
  • 216. Liu T, Luo T, Guo Xet al. PgMYB2, a MeJA-responsive transcription factor, positively regulates the dammarenediol synthase gene expression in Panax ginseng. Int J Mol Sci. 2019;20:2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Reddy VA, Wang Q, Dhar Net al. Spearmint R2R3-MYB transcription factor MsMYB negatively regulates monoterpene production and suppresses the expression of geranyl diphosphate synthase large subunit (MsGPPS.LSU). Plant Biotechnol J. 2017;15:1105–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Ding K, Pei T, Bai Zet al. SmMYB36, a novel R2R3-MYB transcription factor, enhances tanshinone accumulation and decreases phenolic acid content in salvia miltiorrhiza hairy roots. Sci Rep. 2017;7:5104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Zhang J, Zhou L, Zheng Xet al. Overexpression of SmMYB9b enhances tanshinone concentration in salvia miltiorrhiza hairy roots. Plant Cell Rep. 2017;36:1297–309. [DOI] [PubMed] [Google Scholar]
  • 220. Zhou ML, Hou HL, Zhu XMet al. Soybean transcription factor GmMYBZ2 represses catharanthine biosynthesis in hairy roots of Catharanthus roseus. Appl Microbiol Biot. 2011;91:1095–105. [DOI] [PubMed] [Google Scholar]
  • 221. Chen X, Li J, Liu Yet al. PatSWC4, a methyl jasmonate-responsive MYB (v-myb avian myeloblastosis viral oncogene homolog)-related transcription factor, positively regulates patchoulol biosynthesis in Pogostemon cablin. Ind Crop Prod. 2020;154:112672. [Google Scholar]
  • 222. Li J, Chen X, Zhou Xet al. Identification of trihelix transcription factors in Pogostemon cablin reveals PatGT-1 negatively regulates patchoulol biosynthesis. Ind Crop Prod. 2021;161:113182. [Google Scholar]
  • 223. Shi M, Zhou W, Zhang Jet al. Methyl jasmonate induction of tanshinone biosynthesis in salvia miltiorrhiza hairy roots is mediated by jasmonate zim-domain repressor proteins. Sci Rep. 2016;6:20919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Zhou Y, Zhou X, Li Qet al. Molecular cloning, bioinformatics analysis, and transcriptional profiling of JAZ1 and JAZ2 from salvia miltiorrhiza. Biotechnol Appl Bioc. 2017;64:27–34. [DOI] [PubMed] [Google Scholar]
  • 225. Wang Q, Reddy VA, Panicker Det al. Metabolic engineering of terpene biosynthesis in plants using a trichome-specific transcription factor MsYABBY5 from spearmint (Mentha spicata). Plant Biotechnol J. 2016;14:1619–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Yamada Y, Nishida S, Shitan Net al. Genome-wide identification of AP2/ERF transcription factor-encoding genes in California poppy (Eschscholzia californica) and their expression profiles in response to methyl jasmonate. Sci Rep. 2020;10:18066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Wang CT, Liu H, Gao XSet al. Overexpression of G10H and ORCA3 in the hairy roots of Catharanthus roseus improves catharanthine production. Plant Cell Rep. 2010;29:887–94. [DOI] [PubMed] [Google Scholar]
  • 228. Fits L, Memelink J. ORCA3, a jasmonate-responsive transcriptional regulator of plant primary and secondary metabolism. Science. 2000;289:295–7. [DOI] [PubMed] [Google Scholar]
  • 229. Menke FL, Champion A, Kijne JWet al. A novel jasmonate- and elicitor-responsive element in the periwinkle secondary metabolite biosynthetic gene Str interacts with a jasmonate- and elicitor-inducible AP2-domain transcription factor, ORCA2. EMBO J. 1999;18:4455–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Pan Q, Wang Q, Yuan Fet al. Overexpression of ORCA3 and G10H in Catharanthus roseus plants regulated alkaloid biosynthesis and metabolism revealed by NMR-metabolomics. PLoS One. 2012;7:e43038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Liu J, Gao F, Ren Jet al. A novel AP2/ERF transcription factor CR1 regulates the accumulation of vindoline and serpentine in Catharanthus roseus. Front Plant Sci. 2017;8:2082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Deng X, Zhao L, Fang Tet al. Investigation of benzylisoquinoline alkaloid biosynthetic pathway and its transcriptional regulation in lotus. Hortic Res. 2018;5:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Yamada Y, Kokabu Y, Chaki Ket al. Isoquinoline alkaloid biosynthesis is regulated by a unique bHLH-type transcription factor in Coptis japonica. Plant Cell Physiol. 2011;52:1131–41. [DOI] [PubMed] [Google Scholar]
  • 234. Chatel G, Montiel G, Pré Met al. CrMYC1, a Catharanthus roseus elicitor- and jasmonate-responsive bHLH transcription factor that binds the G-box element of the strictosidine synthase gene promoter. J Exp Bot. 2003;54:2587–8. [DOI] [PubMed] [Google Scholar]
  • 235. Zhang H, Hedhili S, Montiel Get al. The basic helix-loop-helix transcription factor CrMYC2 controls the jasmonate-responsive expression of the ORCA genes that regulate alkaloid biosynthesis in Catharanthus roseus. Plant J. 2011;67:61–71. [DOI] [PubMed] [Google Scholar]
  • 236. Singh SK, et al. bHLH iridoid synthesis 3 is a member of a bHLH gene cluster regulating terpenoid indole alkaloid biosynthesis in Catharanthus roseus. Plant Direct. 2021;5:e00305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Pauw B, Hilliou FAO, Martin VSet al. Zinc finger proteins act as transcriptional repressors of alkaloid biosynthesis genes in Catharanthus roseus. J Biol Chem. 2004;279:52940–8. [DOI] [PubMed] [Google Scholar]
  • 238. Kato N, Dubouzet E, Kokabu Yet al. Identification of a WRKY protein as a transcriptional regulator of benzylisoquinoline alkaloid biosynthesis in Coptis japonica. Plant Cell Physiol. 2007;48:8–18. [DOI] [PubMed] [Google Scholar]
  • 239. Suttipanta N, Pattanaik S, Kulshrestha Met al. The transcription factor CrWRKY1 positively regulates the terpenoid indole alkaloid biosynthesis in Catharanthus roseus. Plant Physiol. 2011;157:2081–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Sibéril Y, Benhamron S, Memelink Jet al. Catharanthus roseus G-box binding factors 1 and 2 act as repressors of strictosidine synthase gene expression in cell cultures. Plant Mol Biol. 2001;45:477–88. [DOI] [PubMed] [Google Scholar]
  • 241. Liu Y, Patra B, Pattanaik Set al. GATA and PIF transcription factors regulate light-induced biosynthesis of vindoline in Catharanthus. Plant Physiol. 2019;180:1316–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Li CY, et al. CrBPF1 overexpression alters transcript levels of terpenoid indole alkaloid biosynthetic and regulatory genes. Front Plant Sci. 2015;6:818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Rice ES, Green RE. New approaches for genome assembly and scaffolding. Annu Rev Anim Biosci. 2019;7:17–40. [DOI] [PubMed] [Google Scholar]
  • 244. Wang J, Li J, Li Zet al. Genomic insights into longan evolution from a chromosome-level genome assembly and population genomics of longan accessions. Hortic Res. 2022;9:uhac021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Miga KH, Koren S, Rhie Aet al. Telomere-to-telomere assembly of a complete human X chromosome. Nature. 2020;585:79–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Wang B, Yang X, Jia Yet al. High-quality Arabidopsis thaliana genome assembly with Nanopore and HiFi long reads. Genom Proteom Bioinf. 2022;20:4–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Naish M, Alonge M, Wlodzimierz Pet al. The genetic and epigenetic landscape of theArabidopsiscentromeres. Science. 2021;374:eabi7489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Song JM, Xie WZ, Wang Set al. Two gap-free reference genomes and a global view of the centromere architecture in rice. Mol Plant. 2021;14:1757–67. [DOI] [PubMed] [Google Scholar]
  • 249. Deng Y, Liu S, Zhangi Yet al. A telomere-to-telomere gap-free reference genome of watermelon and its mutation library provide important resources for gene discovery and breeding. Mol Plant. 2022;15:1268–84. [DOI] [PubMed] [Google Scholar]
  • 250. Zhou Z, Tan H, Li Qet al. Trichome and ARTEMISININ regulator 2positively regulates trichome development and artemisinin biosynthesis inArtemisia annua. New Phytol. 2020;228:932–45. [DOI] [PubMed] [Google Scholar]
  • 251. Chalvin C, Drevensek S, Dron Met al. Genetic control of glandular trichome development. Trends Plant Sci. 2020;25:477–87. [DOI] [PubMed] [Google Scholar]
  • 252. Li Y, Kong D, Bai Met al. Correlation of the temporal and spatial expression patterns of HQT with the biosynthesis and accumulation of chlorogenic acid in Lonicera japonica flowers. Hortic Res. 2019;6:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Cheng H, Concepcion GT, Feng Xet al. Haplotype-resolved de novo assembly using phased assembly graphs with hifiasm. Nat Methods. 2021;18:170–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Nützmann HW, Doerr D, Ramírez-Colmenero Aet al. Active and repressed biosynthetic gene clusters have spatially distinct chromosome states. Proc Natl Acad Sci U S A. 2020;117:13800–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Susniak K, Krysa M, Gieroba Bet al. Recent developments of MALDI MSI application in plant tissues analysis. Acta Biochim Pol. 2020;67:277–81. [DOI] [PubMed] [Google Scholar]
  • 256. Sun C, Ma S, Li Let al. Visualizing the distributions and spatiotemporal changes of metabolites in Panax notoginseng by MALDI mass spectrometry imaging. J Ginseng Res. 2021;45:726–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Huang AC, Jiang T, Liu YXet al. A specialized metabolic network selectively modulates Arabidopsisroot microbiota. Science. 2019;364:eaau6389. [DOI] [PubMed] [Google Scholar]

Articles from Horticulture Research are provided here courtesy of Oxford University Press

RESOURCES