Natural Products: source of INNovatIoN CHIMIA 2017, 71, No. 12 851

doi:10.2533/chimia.2017.851 Chimia 71 (2017) 851–858 © Swiss Chemical Society Sunlight-driven Environmental Benign Production of Bioactive Natural Products with Focus on Diterpenoids and the Pathways Involved in their Formation

Dan Luo, Birger Lindberg Møller, and Irini Pateraki*

Abstract: Diterpenoids are high value compounds characterized by high structural complexity. They constitute the largest class of specialized metabolites produced by plants. Diterpenoids are flexible molecules able to engage in specific binding to drug targets like receptors and transporters. In this review we provide an account on how the complex pathways for diterpenoids may be elucidated. Following plant pathway discovery, the com- pounds may be produced in heterologous hosts like yeasts and E. coli. Environmentally contained production in photosynthetic cells like cyanobacteria, green algae or mosses are envisioned as the ultimate future production system. Keywords: Alcohol dehydrogenases · P450s · Ingenol mebutate · Light-driven production · Terpene synthases

1. Introduction anticancer agent widely used today as a Coleus forskohlii, is used in the treatment therapeutic in combinatorial treatments of glaucoma and heart failure based on 1.1 Plant Diterpenoids and their of ovarian cancer, breast cancer, lung can- its activity as a cyclic AMP booster.[8,9] Application as Pharmaceuticals cer, Kaposi sarcoma, cervical cancer, and Ginkgolides from the leaves of Ginkgo bi- Plants produce a wide variety of natural pancreatic cancer.[6,7] Forskolin, a labdane- loba L. are a series of diterpene lactones products that play important roles in both type diterpene produced from the roots of with anti-platelet-activating antagonist ac- general and specialized metabolism.[1,2] Terpenoids constitute the largest and most diverse class of bio-active natural prod- Fig 1. Examples of ucts in the plant kingdom.[3] Based on the common plant terpe- number of five-carbon building blocks noid subclasses. embedded in their structures, terpenoids are categorized as monoterpenoids (C10), sesquiterpenoids (C15), diterpenoids (C20), sesterterpenoids (C25), triterpe- noids (C30), tetraterpenoids (C40) and polyterpenoids (>C40) (Fig. 1). Plants produce more than 11,000 different diter- penoids[4] and they display a wide range of biological activities. Some are used di- rectly as therapeutics for treatment of hu- man diseases whereas others have served as lead compounds (Fig. 2).[5] Paclitaxel, first isolated from the bark of the Pacific yew, Taxus brevifolia, is a well-known

*Correspondence: Assist. Prof. Dr. I. Pateraki Plant Biochemistry Laboratory Center for Synthetic Biology Department of Plant and Environmental Sciences University of Copenhagen, Thorvaldsensvej 40 DK-1871 Frederiksberg C, Copenhagen, Denmark E-mail: [email protected] 852 CHIMIA 2017, 71, No. 12 Natural Products: source of INNovatIoN

tivity.[10] For many years, they have been 2. Biosynthesis of Diterpenoids phosphate (GPP, 10 carbons). Subsequent used in the treatment of cerebrovascular 1'-4 additions of isopentenyl diphosphate and cardiovascular diseases.[11] Ingenol Despite their highly diverse structures, result in formation of farnesyl pyrophos- mebutate is a macrocyclic diterpene from all terpenoids are biosynthesized from two phate (FPP, 15 carbons) and geranylgera- Euphorbia peplus exhibiting remarkable common C5 isoprenoid units, isopentenyl nyl pyrophosphate (GGPP, 20 carbons). antitumor and antileukemic activity. A gel diphosphate (IPP) and dimethylallyl di- GPP, FPP and GGPP are the precursors formulation of ingenol mebutate was ap- phosphate (DMAPP). IPP and DMAP can of monoterpenoids, sesquiterpenoids and proved for the topical treatment of actinic be coupled and elongated through sequen- diterpenoids, respectively. FPP is also in- keratosis, a precondition of squamous-cell tial head-to-tail condensations catalyzed by volved in the biosynthesis of triterpenoids carcinoma.[12] With all the complex struc- prenyltransferases, resulting in formation (30 carbons). Squalene, the general precur- tures discovered in nature and especially of linear prenyl diphosphate intermediates sor of triterpenoids contains six isoprene from plants, diterpenoids are recognized as differing in the number of incorporated units and is formed from two molecules of an especially attractive source of promis- five-carbon building blocks. Two of these FPP (Fig. 3).[13–17] ing drug leads. C5 units are condensed to form geranyl di- The universal C5 precursor isopente- nyl diphosphate (IPP) can be synthesized via two different pathways: the mevalon- ate (MVA) pathway and the 2-C-methyl- D-erythritol 4-phosphate (MEP) pathway. In plants, the MVA pathway is located in the cytosol and mainly provides the C5 iso- prenoid units for the biosynthesis of ses- quiterpenoids and triterpenoids, while the MEP pathway locates in the plastids, pro- viding the C5 isoprenoid units for mono- and diterpenoid biosynthesis (Fig. 4).[15–19] This review is focused on the biosyn- thesis of diterpenoids in plants from the general precursor GGPP generated in the chloroplastbytheMEPpathway.Metabolic crosstalk and interaction between the MEP and MVA pathways have been revealed in several plants species including Nicotiana and Arabidopsis.[20,21] In the biosynthesis Fig 2. Examples of plant diterpenoids with important biological activities and the plants producing of diterpenoids, the C20 precursor derived them. from the MEP pathway is transformed by diterpene synthases (diTPSs) into different types of backbones. This initial cyclization reaction is followed by oxidative decora- tions catalyzed by cytochrome P450 mono- oxygenases (P450s), and further modifica- tions catalyzed e.g. by short-chain alcohol dehydrogenases (ADHs), acyltransferases and glucosyltransferases.[22]

3. Diterpene Synthases (diTPSs)

The initial step in diterpenoid biosyn- thesis is the formation of the basic back- bone, which is mostly carried out by a family of known as diterpene synthases or cyclases (diTPSs). DiTPSs facilitate cyclization and/or rearrange- ment of the linear C20 prenyl diphosphate substrate into an enormous diversity of diterpene backbones through different carbocation reactions. These carboca- tion-driven reactions encompass a series of similar electrophilic steps: firstly, the generation, transformation, and stabiliza- tion of highly reactive carbocations; then the ultimate quenching of reactive carbo- cations by deprotonation or nucleophile capture. The structural features of the di- Fig 3. Scheme of the general terpenoid classes and corresponding precursors (OPP, diphosphate; terpene backbones formed arise from the PPi, inorganic pyrophosphate). Figure adapted with permission from Y. Gao, R. B. Honzatko, R. J. diverse conformations of substrate and Peters, Nat. Prod. Rep. 2012, 29, 1153. © Royal Society of Chemistry intermediates enforced by each individual Natural Products: source of INNovatIoN CHIMIA 2017, 71, No. 12 853

and isomerization.[32] As one of the larg- est super-families of enzymes, genes en- coding P450s are found in the genomes of all plants. In angiosperms, the number of P450s within each species has exploded, with an average of 300 members per spe- cies, which may be divided into 50 families and subfamilies based on their sequence homology. The sequence identity of P450s from different families, even present in the same species, is often quite low and even less than 20%.[33] Despite their variability, the structural fold of P450 proteins is high- ly conserved. As heme-thiolate proteins, a Mitochondria conserved structural feature is the cysteine residue in the core of the protein serving as fifth (axial) ligand to the heme iron and thereby mediating heme-binding and cata- lytic properties. The thiolate function of the conserved cysteine residue, together with the iron-protoporphyrin IX (heme) con- stitutes the active center for catalysis. The catalytic cycle of a P450 requires electron transfer from NADPH carried out by an FMN/FAD containing NADPH-dependent P450 (POR). In vivo, plant P450s and PORs are membrane-anchored to the surface of the endoplasmic reticu- lum (ER) (Fig. 4).[34] The less conserved regions of the P450s are associated with the substrate specificity, type of catalytic reactions and membrane localization. The Fig. 4. Schematic representation of a plant cell and subcellular localization of terpenoid me- regions related to substrate binding and tabolism. Hemifused bilayers consisting by plastid and ER membrane, have been proposed recognition are flexible, adjusting confor- as a translocation mechanism for nonpolar terpene compounds. Figure adapted from Johan mation upon binding of substrate to allow Andersen-Ranberg’s PhD thesis. the catalytic reaction. The abundance of variations of these regions in the P450s fam- synthase. Structural features of the diTPSs ily reflects their high diversity of substrate also prevent a particular reaction series selectivity and catalytic capability.[31,33] from premature termination.[23,24] Through As their functions are extremely diverse, mastering these reaction channels, diTPSs P450s are involved in almost all metabolic are capable to catalyze the formation of pathways in plants, for example, the bio- various classes of diterpenoids. According synthesis of alkaloids, phenylpropanoids, to different cyclization levels of their cyanogenic glucosides, glucosinolates and backbones, diterpenoids can be classi- terpenoids.[4] Despite the importance of this fied into three groups: linear diterpenes, class of enzymes, functional characteriza- macrocyclic diterpenes and polycyclic tion of newly discovered P450 remains an diterpenes (Fig. 5).[25–29] The biosynthe- open field of investigation due to the large sis of macrocyclic diterpenes has been of number of P450 encoding genes within a special interest in our laboratory. DiTPSs single plant species. involved in the biosynthesis of macrocy- Fig. 5. Examples of diterpene backbones with clic diterpenes, such as casbene synthase different cyclization levels. (CBS) and taxadiene synthase, are mostly 5. Short-chain Alcohol ionization-dependent cyclases.[28,29] These Dehydrogenases in Terpenoid synthases share common features in their 4. Cytochrome P450 Biosynthesis catalytic mechanisms: an initial ionization Monooxygenases of the diphosphate group of GGPP, subse- Alcohol dehydrogenases (ADHs) are quent attack of the electrons of an internal Oxygenation is the most common mod- enzymes catalyzing oxidation of alco- double bond on the resulting carbocation ification to extend the structural diversity hols and reduction of aldehydes/ketones followed by rearrangements, and finally and complexity of plant diterpenoids.[4,31] in the presence of coenzymes NAD(H)/ stabilization of the intermediate by proton Cytochrome P450 monooxygenases NADP(H).[35] ADHs can be classified into abstraction.[30] The catalytic mechanism (P450s) are the major enzymes to generate long-, medium- and short-chain dehy- of casbene synthase is analogous to those a variety of regiospecific and stereospecif- drogenases/reductases according to their of angiosperm sesquiterpene synthases in ic oxidative modifications at physiologi- polypeptide chain length, mechanistic fea- the TPS-a subfamily, which is consistent cal conditions and ambient temperatures, tures and distinct sequence motifs.[36] In with their sequence relatedness and phy- including hydroxylation, ketonization, terpenoid metabolism, short-chain ADHs logenies.[24] epoxidation, oxidative C–C bond cleavage or SDRs, in addition to P450s, are an 854 CHIMIA 2017, 71, No. 12 Natural Products: source of INNovatIoN

important family of enzymes for catalyz- ailment are mostly targeting large biologi- the early intermediates of diterpenoid syn- ing oxidation/reduction reactions for the cal molecules such as proteins, DNA and thesis. Beyond that, a geometrically differ- modification or functionalization of ter- RNA. Inhibition, activation or other inter- ent alternative structure of the C20 precur- penoid molecules.[37–42] Enzymes in this actions with those targets elicit a biologi- sor further serves to extend the diversity family, with the typical mass of 25/30 kDa cal response in the human body, resulting of diterpenoids discovered in nature. This and a polypeptide chain composed of ap- in a positive therapeutic effect following was recently discovered, when some nov- proximately 250 amino acid residues, are administration of the right drug.[57] With el diTPSs from plants in the Solanaceae characterized by several common proper- the huge number of potential structures family were found to use the cis-prenyl di- ties: a conserved 3D structure consisting already known to be produced by plants, phosphate nerylneryl diphosphate (NNPP) of ‘Rossmann-fold’ β-sheet with α-helices diterpenoids represent a tremendous struc- as a substrate, rather than the trans-prenyl on both sides, an N-terminal dinucleotide tural repository, not only with respect to diphosphate E,E,E-geranylgeranyl diphos- binding motif and an the numerous patterns of their backbone phate (GGPP), which as described above with a catalytical residue motif YxxxK.[43] structures, but also by the suite of incor- is the substrate of most known diTPSs.[62] Despite their conserved structure, these porated functional groups (e.g. hydroxyl After formation of the different core struc- short-chain ADHs exhibit large sequence groups, keto groups, carboxyl groups, tures, membrane-bound P450s are subse- divergences. The low sequence similari- alkyl groups, sugar residues, etc.). In ad- quently recruited as versatile catalysts to ties among the plant short-chain ADHs dition, diterpenoids contain an extremely enhance the structural diversity by regio- renders functional prediction through phy- high fraction of carbons in tetrahedral and stereospecific introduction of oxygen- logenetic analysis challenging. Functional sp3 hybridization, which offers bountiful ated functional groups. Apart from diTPSs characterization of these short-chain ADHs opportunities for incorporation of chiral and P450s, enzymes from other families, should therefore be based on experimental centers within the diterpenoid structures. such as iridoid synthase-like reductases, data regarding their bioactivity, like in the These chemical features result in an un- have recently been reported to be able to case of P450s. In terpenoid biosynthesis, precedented number of unique structural use primary backbones constructed by short-chain ADHs cover a wide range of conformations, providing diterpenoid mol- diTPSs as substrates for multi-step modi- substrate spectra from monoterpenes, ses- ecules with a strong potential for properly fications and rearrangements to generate quiterpenes, diterpenes to triterpenes. The matching the shape of the binding or active cyclized structures of increased complex- majority of the short chain ADHs known sites of drug-target proteins. Building on ity.[63] Last but not least, soluble enzymes to be involved in terpenoid biosynthesis a number of comprehensive experimental like acyltransferases and glucosyltransfer- are members of the SDR110C family,[43,44] analyses, a small structural change is capa- ases, typically complete the biosynthetic including zerumbone synthase in Zingiber ble to lead to a significant alteration in the pathways by adding structural motifs de- zerumbet;[37] xanthoxin dehydrogenase in biological response, especially when these rived from fatty acid and/or carbohydrate Arabidopsis thaliana;[38] borneol dehydro- changes affect the energy of interaction biosynthesis. Together these sets of soluble genase in Lavandula x intermedia;[39] mo- of a ligand with its receptor.[58] Dramatic and membrane bound enzymes are able to milactone A synthase in Oryza sativa;[40] changes in their therapeutic activities rang- provide the catalytic abilities required for aldehyde dehydrogenase 1 in Artemisia an- ing from none to inhibitory or excitatory the formation of the multitude of diterpe- nua;[41] (–)-isopiperitenol dehydrogenase in might be induced by the replacement of noids present in the plant kingdom. Mentha x piperita.[42] In terpenoid biosyn- a single functional group.[59,60] The diver- thesis, their catalytic activities are mainly sity of functional groups in diterpenoid reported as oxidative rather than reductive. structures thus contributes greatly to their 7. Macrocyclic Diterpenoids and ability to interact with the very same target their Pharmacological Properties in different ways, resulting in completely 6. Why are Diterpenoids Favored different biological effects. This augments Some diterpenoids, such as the gibber- for Biological Activities? the opportunity for diterpenoids to be in- ellin-type phytohormones and phytol, play volved and actively targeting a wide range important roles in general metabolism and Throughout the human history, indig- of signaling pathways in the human body. are therefore present across the plant king- enous people from different areas of the The structural complexity and diver- dom.[64,65] But the large majority of plant planet have used diterpenoid-producing sity of diterpenoids reflects their biosyn- diterpenoids are specialized metabolites plants and their extracts as traditional med- thetic pathways. In comparison to the with a restricted taxonomic distribution. icines sharing many of the same properties biosynthetic pathways for other classes of Macrocyclic diterpenes and their polyes- with present day therapeutics.[45] Within natural products such as the alkaloids and ter derivatives are mainly isolated from the plant-derived terpenoids, diterpenoids phenylpropanoids, the biosynthetic path- the Thymelaeaceae and Euphorbiaceae are especially rich in structures which are way for diterpenoids are built based on plants, which could be considered as taxo- biologically active against a wide range modules which are used in many different nomic markers or signature molecules.[66] of human diseases, including cancer, combinations. The plethora of core struc- Euphorbiaceae, the spurge family, is one cardiovascular diseases, malaria, inflam- tures, which results from the ability of the of the largest families of flowing plants, mations, diabetes and viral and bacterial soluble diTPSs to cyclize GGPP accord- consisting of approximately 7,500 species infections.[46–51] Studies on structure–ac- ing to different model schemes, is truly subdivided into 300 genera.[67] Members of tivity relationships (SAR) of diterpenoids impressive. While diTPSs mainly produce this family produce a vast array of phyto- document that the diverse bioactivity and hydrocarbon backbones, they are also able toxins, mainly diterpenoid esters, alkaloids high efficiencies of this class of com- to generate some oxygenated diterpenes and glycosides.[68] The genus Euphorbia is pounds arise from their structurally unique directly without the involvement of other the largest genera in the spurge family with features.[52–56] From a mechanistic point of enzymes. For example, a pair of diterpene about 2,160 species. Plants in this genus view, the structural conformation of these synthases, CfTPS2 and CfTPS3, from the share a common feature, the production of small molecules is crucial as it determines Indian plant Coleus forskohlii, was found a poisonous, milky and irritant latex-like whether they bind and interact with targets to catalyze the formation of 13R-manoyl sap. The skin-irritating and caustic effects like receptors or transporters. In humans, oxide.[61] This catalytic character of some of the latex sap are attributed to the pres- drugs administered to cure a disease or diTPSs enriches the elements involved in ence of specific classes of macrocyclic Natural Products: source of INNovatIoN CHIMIA 2017, 71, No. 12 855

diterpenoids.[69] According to different stages of cyclization of their C back- 20 bones, they are classified into two groups: simple bicyclic casbene type and further cyclized types. The latter includes jatroph- anes, lathyranes, tiglianes, daphnanes, and ingenanes ranked according to their in- creased structural complexity. In addition to the high structural variety of the back- bones, the immense structural diversity occurring among macrocyclic diterpenoids results from the monooxygenation reac- tions catalyzed by P450s and the derived options to introduce esterification with different aliphatic (e.g. acetyl, n-butanoyl, isobutanoyl, methylbutanoyl, tiglyl, ange- loyl, isovaleroyl, etc.) and aromatic (ben- zoyl and nicotinoyl) carboxylic acids. As the best characterized metabolites of this genus, these macrocyclic diterpenoids are also called ‘Euphorbia diterpenoids’. Their specific taxonomic distribution and unique structural features have inspired chemists to search for the discovery of po- tential drug leads. Thus many macrocyclic diterpenoids have been tested for a wide range of therapeutically relevant biologi- cal activities, including antitumor, multi- drug-resistance-reversal, various vascular effects, antiviral and anti-inflammatory activity.[69,70] Euphorbia factor L10 and eu- phodendroidin D show powerful inhibition of the transport activity of P-glycoprotein, Fig. 6. A. Euphorbia peplus is the natural source of ingenol mebutate, but due to it‘s low concen- a multidrug transporter overexpressed in tration in the specific plant and the low availability of plant tissue, LEO pharma is using Euphorbia cancer cell plasma membranes as an efflux lathyris L. for the extraction of an ingenol mebutate derivative which is easily chemically converted pump conferring cellular resistance to anti- to ingenol mebutate. B. Structure of the diterpenoid ingenol mebutate. C. Typical presentation of cancer chemotherapy.[55,71] Prostratin is in a patient with actinic keratosis lesions (Photo courtesy of Dermnet.com). D. Picato Gel produced phase I human clinical trials for the treat- by LEO Pharma (Photo from http://www.qldsun.com.au/picato). ment of HIV, as it was shown to activate viral reservoirs in latently infected T cells, guided fractionation and identification of 9. Synthetic Biology-based as well as to inhibit viral replication.[72] the diterpenoids present in the latex sap Platforms for Sustainable Resiniferatoxin, now in phase II and III revealed that the active constituent of the Production of Structurally clinical trials, activates the transient vanil- sap was ingenol mebutate. In 2012, a gel Complex Diterpenoids loid receptor 1 (TRPV1) in a subpopula- containing ingenol mebutate (trade name tion of primary afferent sensory neurons, Picato) was approved by the US Food Most bioactive diterpenoids have been blocking nerves that transmit physiologi- and Drug Administration (FDA) and by isolated from non-cultivated medicinal cal pain.[73] These biological investigations the European Medicines Agency (EMA) plants or from endangered or red-listed provide natural science-based evidence for for topical treatment of actinic keratosis, species. A shortage of available wild plant the beneficial and health promoting uses a kind of common skin lesions with the material represents a serious obstacle for of many Euphorbia plants in traditional potential to transform into squamous cell therapeutic uses. Because of their struc- medicines.[74] With the improved analyti- carcinoma (SCC). The mechanism of ac- tural complexity, chemical synthesis of cal technologies now available, numer- tion for ingenol-3-angelate is described plant-derived diterpenoids, especially ous new macrocyclic diterpenoids will be as dual, combining rapid induction of particular stereoisomers, remains uneco- identified and made available for determi- primary necrosis followed by neutrophil- nomic and often inadequate for large- nation of their pharmacological activities. mediated, antibody-dependent cellular scale production. Additionally, in chemi- cytotoxicity towards residual diseased cal synthesis, the use of toxic catalysts and cells. The latter activity is mediated by solvents has raised public concerns about 8. Ingenol Mebutate the action of ingenol-3-angelate towards a the environment. The lack of a reliable and broad range of protein kinase C isoforms, environmentally benign and stable supply Ingenol mebutate (ingenol-3-angelate leading to anti-cancer CD8 T cell-based chain thus remains a major obstacle when or PEP005) is an ingenane-type macrocy- immunotherapies.[77,78] Apart from actinic the desire arises to develop a specific di- clic diterpenoid ester with anti-leukemic keratosis, ingenol-3-angelate was found to terpenoid into a medicinal drug. and anti-tumor activity. Ingenol mebutate induce apoptosis in acute myeloid leuke- Different strategies to improve the pro- was first isolated from Euphorbia peplus mia cells by activating the protein kinase duction yield of pharmaceutically inter- (Fig. 6).[75] The latex of E. peplus is a tra- C isoform PKCδ.[79] In other studies, in- esting diterpenoids have been exploited. ditional remedy for warts, skin keratosis genol mebutate was found to inhibit T cell In the early stages of taxol production, and basal cell carcinoma.[76] Activity- apoptosis by activating PKCθ.[80] the compound paclitaxel was directly 856 CHIMIA 2017, 71, No. 12 Natural Products: source of INNovatIoN

harvested from the bark of Pacific yew. to natural fluctuations and a biennial life in yeast and afforded forskolin titers of 40 This production process killed numerous cycle, the supply of E. lathyris L. seeds mg/L.[93] Forskolin, a structurally complex Pacific yew trees, seriously vitiating the is still quite limited. The increasing de- diterpenoid, demonstrates the potential of wild population of this Taxus species and mand for pharmaceutical-grade ingenol- synthetic biology for sustainable produc- the ecology of the forests in the Northwest 3-angelate motivated us to embark into tion of complex diterpenoids with impor- Pacific. To protect the Pacific yew and its development of a synthetic biology-based tant bioactivities. In parallel with the syn- natural habitats, two alternative approach- approach towards economic and environ- thetic biology-based approach, targeted es were developed to obtain paclitaxel.[81] mentally benign production.[88] molecular breeding efforts of the plant Early in the 1990s, cell cultures of Taxus Microbial production using synthetic species producing the desired compound species producing paclitaxel were used biology approaches has attracted wide at- may raise the production level of the target as starting materials.[82] Total synthesis tention as an alternative strategy for sus- compounds significantly and thus could be of paclitaxel was accomplished in 1994 tainable and environmentally friendly pro- used as an alternative commercial source, based on extensive efforts of synthetic duction of high-value terpenoids. Synthetic as observed in the successful work in chemists in the US.[83] The chemical syn- biology involves metabolic engineering of Artemisia annua.[94–96] thesis afforded new general knowledge the modules of nature into new combina- Light-driven biotechnological pro- on structurally complex diterpenoids, tions to produce biological components, duction (Fig. 7), which uses ‘green’ pho- but due to the low overall process yield, systems, cells and organisms that address tosynthetic cells as production hosts, is it did not form the basis for commercial society’s needs. The modules are intro- a possible future ideal and ultimate way production. A third production approach duced in combinations where each module to obtain structurally complex plant di- was based on a short semisynthetic route is functionally active receiving an input in terpenoids.[97] Different from traditional using 10-deacetylbaccatin as starting ma- the form of a substrate which the module microbial production systems such as E. terial and was shown feasible for indus- transforms into an output in the form of a coli and yeast, ‘green’ cell hosts, including trial production because the compound product serving as substrate for the next cyanobacteria algae and moss, are highly 10-deacetylbaccatin used as starting module. A basic principle is to work with ‘self-sufficient’. No external sugar sup- material could be isolated in high yields nature-based knowledge on how modules ply is required during the growth of these from the European yew, Taxus baccata.[84] may work and interact. In synthetic biol- cells. Instead, they provide themselves After that, semi-synthetic production of ogy, the modules used are shared for use with energy, reducing power, and carbon paclitaxel was further improved using in bottom-up approaches to reach sus- building blocks through the reactions of combinations of precursors isolated from tainable solutions.[89] Reconstruction of photosynthesis, channeling the energy of foliage of cultivated Taxus species and specific terpenoid pathways in microbial sunlight and CO from the atmosphere into 2 plant cell cultures.[85,86] The same situ- host organisms has been achieved, includ- metabolic pathways. While the cultivation ation also applied to ingenol mebutate. ing production of the antimalarial ses- of genetically modified plants in the field is Direct isolation from the original plant quiterpenoid artemisinin that in nature is controversial, algae and cyanobacteria can source E. peplus only yielded 1.1 mg/kg produced by the plant sweet wormwood easily be grown environmentally contained from the aerial tissue, which hampered (Artemisia annua).[90] Likewise, micro- in photo-bioreactors (Fig. 8). Therefore, commercial production. Current produc- bial routes towards production of the fra- light-driven production has the potential to tion of ingenol mebutate proceeds as a grances sclareol and santalol have been be economic feasible and sustainable for semi-synthetic approach using hydrolysis designed.[91,92] Recently, the complete large-scale production, since sunlight and products from the seed oil of an alternative biosynthetic pathway for the cyclic AMP CO are inexhaustible energy and carbon 2 Euphorbia species, E. lathyris L.[87] Due booster forskolin was stably reconstructed sources provided by nature.[98,99]

Fig. 7. Strategy for light-driven biotechnological production of ingenol mebutate (ingenol-3-angelate) A. Structure of the diterpenoid. B. Photosynthesis and biosynthesis of specialized bioactive compounds are separated at the cellular level taking place in the chloroplast and endo- plasmic reticulum, respectively. C. Co-localization of the biosynthetic pathway of ingenol-3-angelate together with the photosynthetic complexes in the chloroplasts of moss and green algae via bioengineering. Note that the same principles can be used to target P450s into the cyanobacterial thylakoid membrane and likewise drive the reactions using solar energy. Figure adapted from http://plantpower.ku.dk/about/ Natural Products: source of INNovatIoN CHIMIA 2017, 71, No. 12 857

candidate genes represented in the tran- and the development and successful intro- scriptomes, homology-based screening is duction of new products manufactured us- carried out using previously characterized ing renewable resources and by the use of diTPS and P450 encoding gene sequences yet to be developed novel green technolo- from other species involved in terpenoid gies that are economically viable and have metabolism. transformative power. In our research, we Next, to determine which of the select- are trying to contribute to this transition ed cDNAs participate in the target biosyn- by developing sunlight-driven production thetic pathway, functional characterization platforms for high value products. Because of candidate genes has to be performed; for the photosynthetic cells optimized for pro- this step different screening and expression duction of high value compounds are ge- systems may be used. These include in netically engineered, we decided not to vivo experiments using the Nicotiana ben- work with plants grown in the field or in thamiana/agrobacterium transient expres- greenhouses. Instead all production will sion system, engineered Saccharomyces be environmentally contained using cya- cerevisiae (yeast) strains and in vitro as- nobacteria, algae or moss cells grown in says with recombinant enzymes purified suspension cultures in transparent plastic from Escherichia coli strains or coupled bags. The major challenge we currently in transcription/translation assays. The face is to optimize the photosynthetic pro- N. benthamiana/agrobacterium transient duction systems to efficiently compete Fig. 8. Microalgae are cultured in photo-biore- expression system offers fast functional with classical production systems based actors at Novagreen using natural light. screening of candidate genes.[102] This on fermentation processes like yeast or E. plant-based expression system has great coli cultures, whose production is based on advantage in heterologous expression of direct use of petrochemicals. We are con- 10. Strategies used for Gene plant enzymes owing to the ‘close to na- fident that within the next decade or two, Discovery tive’ internal environment in which the the growing demand for sunlight-driven proteins will be produced and functionally environmental benign production systems To enable heterologous production of embedded. Because N. benthamiana plants will attract the necessary huge investments high-value diterpenoids in non-photosyn- produce diterpenoids for their own growth required to make this vision a reality. thetic as well as photosynthetic microbial and self-defense, the machinery for GGPP cells as mentioned above, knowledge of synthesis exists naturally in this model- Acknowledgements the genes encoding the enzymes involved plant host. An additional benefit of this Executive Assistant Dr. Anna Khodosevich in the biosynthetic pathways giving rise ‘green’ host is that it favors the expression is thanked for careful proof reading and han- to compound formation is essential. and functionality of heterologous plant dling of the manuscript. The authors grate- fully acknowledge financial support from Fortunately, new technologies facilitating P450s.[103] Due to issues such as membrane the VILLUM Foundation to the VILLUM and guiding plant pathway discovery have localization of the protein and availability Research Center ‘Plant Plasticity’ (BLM), been developed.[100] of reducing power, it can be quite challeng- from the UCPH Excellence Program for The genomes of numerous medicinal ing to functionally express plant P450s in Interdisciplinary Research to Center for plants are currently being sequenced but E.coli and yeast.[104] Despite these benefits, Synthetic Biology ‘bioSYNergy’ (BLM), from most of them are not yet publicly avail- the Nicotiana expression system also has the European Research Council Advanced able. This also applies for C. forskohlii, E. its limits. Intermediates and products of Grant number 323034: ‘Light-driven Chemical peblus and E. lathyris. As a fast and cheap interest synthesized from heterologously Synthesis using Cytochrome P450s’ (BLM) alternative, RNA sequencing (RNAseq) introduced pathways might be further and from the ERC Proof of Concept Grant based transcriptome libraries have been metabolized by endogenous enzymes and number 680896: ‘SUNLIGHTING’ (BLM). shown very useful in the field of pathway other detoxification mechanisms in N. Received: October 23, 2017 discovery in these non-model plants, of- benthamiana, since this plant species also fering the opportunity to explore unknown produce oxygenated diterpenoids formed and functional space involved by endogenous diTPSs and P450s.[105] To [1] H. P. Makkar, P. Siddhuraju, K. Becker, Meth. in a biosynthetic pathway. Additionally, confirm the results observed using N. ben- Mol. Biol. 2007, 393, 1. these RNAseq libraries can provide rela- thamiana as host, it is therefore advisable [2] K. Springob, T. M. Kutchan, ‘Introduction to tive transcript abundances of the cDNAs to carry out parallel experiments in engi- the Different Classes of Natural Products’, in ‘Plant-derived Natural Products, Synthesis, expressed in the selected plant tissue or neered yeast strains or in vitro enzyme as- Function and Applications’, Eds. A. E. Osbourn, cell type, which can lead the selection of says (where possible) as controls. V. Lanzotti, Springer-Verlag New York, 2009, candidate cDNAs.[100,101] Biosynthesis and 3–50. accumulation of specialized diterpenoids [3] B. Singh, R. A. Sharma, 3 Biotech 2015, 5, 129. [4] B. Hamberger, S. Bak, Philos. Trans. R. Soc. [61] 11. Conclusion is typically limited to specific cell types. Lond. B Biol. Sci. 2013, 368, 20120426. Investigation of the formation of a specific [5] S. Mafu, P. Zerbe, Phytochem. Rev. 2017, DOI: diterpenoid in the course of plant ontogeny The global challenges that humankind 10.1007/s11101-017-9513-5. and within a specific cell type using tracer is facing worldwide related to the use of [6] M. C. Wani, H. L. Taylor, M. E. Wall, P. Coggon, studies, metabolite profiling or mass spec- fossil fuels, climate change, food security, A. T. McPhail, J. Am. Chem. Soc. 1971, 93, 2325. trometry identifies the most suitable tis- environmental preservation, mass migra- [7] G. M. Cragg, Med. Res. Rev. 1998, 18, 315. sues for pathway elucidation, where genes tion and financial inequality are all well [8] S. Doseyici, I. Mehmetoglu, A. Toker, F. H. encoding enzymes involved in the target known. In order to address these challeng- Yerlikaya, E. Erbay, Biotech. Histochem. 2014, pathway are probably highly expressed. es, we should first shift our main produc- 89, 388. tion systems away from petrochemicals. [9] K. B. Seamon, W. Padgett, J. W. Daly, Proc. Next-generation sequencing technology Natl. Acad. Sci. USA 1981, 78, 3363. is then performed to obtain the transcrip- This transition towards a truly bio-based [10] K. Stromgaard, K. Nakanishi, Angew. Chem. Int. tomes of the chosen tissues. To select society requires multidisciplinary research Ed. 2004, 43, 1640. 858 CHIMIA 2017, 71, No. 12 Natural Products: source of INNovatIoN

[11] K. M. Nash, Z. A. Shah, Integr. Med. Insights [46] M. T. Islam, Phytotherapy Res. 2017, 31, 691 [81] J. Goodman, V. Walsh, ‘The story of taxol: na- 2015, 10, 1. [47] A. M. de Oliveira, C. R. Tirapelli, S. R.Ambrosio, ture and politics in the pursuit of an anti-cancer [12] G. Siller, K. Gebauer, P. Welburn, J. Katsamas, S. F. B. da Costa, Recent Pat. Cardiovasc. Drug drug’, Cambridge University Press, 2001. M. Ogbourne, Aust. J. Dermatol. 2009, 50, 16. Discov. 2008, 3, 1. [82] Y. Yukimune, H. Tabata,Y. Higashi,Y. Hara, Nat. [13] Y. Gao, R. B. Honzatko, R. J. Peters, Nat. Prod. [48] R. Batista, J. Silva Ade, Jr., A. B. de Oliveira, Biotechnol. 1996, 14, 1129. Rep. 2012, 29, 1153. Molecules 2009, 14, 3037. [83] K. Nicolaou, Z. Yang, J. Liu, H. Ueno, P. [14] T. W. Goodwin, Annu. Rev. Plant Physiol. 1979, [49] C. Cardenas, A. R. Quesada, M. A. Medina, Nantermet, R. Guy, C. Claiborne, J. Renaud, E. 30, 369. PLoS One 2011, 6, e23407. Couladouros, K. Paulvannan, Nature 1994, 367, [15] A. X. Cheng, Y. G. Lou, Y. B. Mao, S. Lu, L. J. [50] A. Nagarajan, P. Brindha, J. Pharma. Res. 2012, 630. Wang, X. Y. Chen, J. Integ. Plant Biol. 2007, 49, 5, 4530. [84] P. G. Wuts, Curr. Opin. Drug Discov. Devel. 179. [51] A. Ulubelen, S. Öksüz, G. Topcu, A. C. Gören, 1998, 1, 329. [16] E. Vranova, D. Coman, W. Gruissem, Mol. Plant W. Vo elter, J. Nat. Prod. 2001, 64, 549. [85] S. A. Wilson, S. C. Roberts, Plant Biotechnol. J. 2012, 5, 318,. [52] K. B. Seamon, J. W. Daly, H. Metzger, N. J. de 2012, 10, 249. [17] D. Tholl, Curr. Op. Plant Biol. 2006, 9, 297. Souza, J. Reden, J. Med. Chem. 1983, 26, 436. [86] S.-H. Pyo, H.-B. Park, B.-K. Song, B.-H. Han, [18] E. Vranova, D. Coman, W. Gruissem, Annu. Rev. [53] L. Moujir, A. M. Gutiérrez-Navarro, L. San J.-H. Kim, Proc. Biochem. 2004, 39, 1985. Plant Biol. 2013, 64, 665. Andrés, J. G. Luis, Phytochem. 1993, 34, 1493. [87] X. Liang, G. Grue-Sørensen, A. K. Petersen, T. [19] D. Tholl, S. Lee, The Arabidopsis Book 2011, 9, [54] G. Krauter, C. W. Vo n der Lieth, R. Schmidt, E. Högberg, Synlett 2012, 2647. e0143. Hecker, Eur. J. Biochem. 1996, 242, 417. [88] G. Appendino, Angew. Chem. Int. Ed. 2014, 53, [20] O. Laule, A. Fürholz, H.-S. Chang, T. Zhu, X. [55] G. Corea, E. Fattorusso, V. Lanzotti, O. 927. Wang, P. B. Heifetz, W. Gruissem, M. Lange, Taglialatela-Scafati, G. Appendino, M. Ballero, [89] E. Andrianantoandro, S. Basu, D. K. Karig, R. Proc. Natl. Acad. Sci. USA 2003, 100, 6866. P. N. Simon, C. Dumontet, A. Di Pietro, J. Med. Weiss, Mol. Syst. Biol. 2006, 2, 2006 0028. [21] A. Hemmerlin, J.-F. Hoeffler, O. Meyer, D. Chem. 2003, 46, 3395. [90] C. J. Paddon, P. J. Westfall, D. J. Pitera, K. Tritsch, I. A. Kagan, C. Grosdemange-Billiard, [56] G. Corea, E. Fattorusso, V. Lanzotti, R. Motti, M. Rohmer, T. J. Bach, J. Biol. Chem. 2003, 278, P.-N. Simon, C. Dumontet, A. Di Pietro, J. Med. Benjamin, K. Fisher, D. McPhee, M. D. Leavell, 26666. Chem. 2004, 47, 988. A. Tai, A. Main, D. Eng, D. R. Polichuk, K. H. [22] P. Zerbe, J. Bohlmann, Trends Biotechnol. 2015, [57] J. P. Hughes, S. Rees, S. B. Kalindjian, K. L. Teoh, D. W. Reed, T. Treynor, J. Lenihan, M. 33, 419. Philpott, Br. J. Pharmacol. 2011, 162, 1239. Fleck, S. Bajad, G. Dang, D. Dengrove, D. Diola, [23] E. Davis, R. Croteau, Biosynthesis 2000, 53. [58] A. C. Anderson, Chem. Biol. 2003, 10, 787. G. Dorin, K. W. Ellens, S. Fickes, J. Galazzo, S. [24] J. Bohlmann, G. Meyer-Gauen, R. Croteau, [59] D. Colquhoun, Br. J. Pharmacol. 1998, 125, 923. P. Gaucher, T. Geistlinger, R. Henry, M. Hepp, Proc. Natl. Acad. Sci. USA 1998, 95, 4126. [60] S. Souza, M. Trindade, J. R. G. d. S. Almeida, T. Horning, T. Iqbal, H. Jiang, L. Kizer, B. Lieu, [25] D. M. Martin, J. Faldt, J. Bohlmann, Plant A. A. Souza Araujo, M. C. Duarte, D. P. Gelain, D. Melis, N. Moss, R. Regentin, S. Secrest, H. Physiol. 2004, 135, 1908. J. C. F. Moreira, M. R. V. Santos, L. J. Quintans Tsuruta, R. Vazquez, L. F. Westblade, L. Xu, M. [26] M. Herde, K. Gartner, T. G. Kollner, B. Fode, W. Júnior, Basic Clin. Pharmacol. Toxicol. 2014, Yu,Y. Zhang, L. Zhao, J. Lievense, P. S. Covello, Boland, J. Gershenzon, C. Gatz, D. Tholl, Plant 115, 244. J. D. Keasling, K. K. Reiling, N. S. Renninger, J. Cell 2008, 20, 1152. [61] I. Pateraki, J. Andersen-Ranberg, B. Hamberger, D. Newman, Nature 2013, 496, 528. [27] C. I. Keeling, H. K. Dullat, M.Yuen, S. G. Ralph, A. M. Heskes, H. J. Martens, P. Zerbe, S. S. [91] M. Schalk, L. Pastore, M. A. Mirata, S. Khim, S. Jancsik, J. Bohlmann, Plant Physiol. 2010, Bach, B. L. Moller, J. Bohlmann, B. Hamberger, M. Schouwey, F. Deguerry, V. Pineda, L. Rocci, 152, 1197. Plant Physiol. 2014, 164, 1222. L. Daviet, J. Am. Chem. Soc. 2012, 134, 18900. [28] C. Mau, C. A. West, Proc. Natl. Acad. Sci. USA [62] Y. Matsuba, J. Zi, A. D. Jones, R. J. Peters, E. [92] G. Scalcinati, S. Partow, V. Siewers, M. Schalk, 1994, 91, 8497. Pichersky, PLoS One 2015, 10, e0119302. L. Daviet, J. Nielsen, Microbial Cell Factories [29] M. Hezari, N. G. Lewis, R. Croteau, Arch. [63] F. Geu-Flores, N. H. Sherden, V. Courdavault, V. 2012, 11, 117. Biochem. Biophys. 1995, 322, 437. Burlat, W. S. Glenn, C. Wu, E. Nims, Y. Cui, S. [93] I. Pateraki, J. Andersen-Ranberg, N. B. Jensen, [30] R. Croteau, E. Davis, T. Hartmann, T. E. O’Connor, Nature 2012, 492, 138. S. G. Wubshet, A. M. Heskes, V. Forman, B. Hemscheidt, J. Sanz-Cervera, B. Shen, E. [64] R. J. Peters, in ‘Isoprenoid synthesis in plants Hallstrom, B. Hamberger, M. S. Motawia, C. Stocking, R. Williams, ‘Biosynthesis: Aromatic and microorganisms’, Springer, 2012, p. 233. E. Olsen, D. Staerk, J. Hansen, B. L. Moller, Polyketides, Isoprenoids, Alkaloids’, Vol. 209, [65] T. Ischebeck, A. M. Zbierzak, M. Kanwischer, P. B. Hamberger, eLife 2017, 6, DOI: 10.7554/ Springer, 2003. Dormann, J. Biol. Chem. 2006, 281, 2470. eLife.23001. [31] I. Pateraki, A. M. Heskes, B. Hamberger, Adv. [66] R. F. Keeler, in ‘Handbook of Natural Toxins: [94] I. A. Graham, K. Besser, S. Blumer, C. A. Biochem. Eng. Biotechnol. 2015, 148, 107. Toxicology of Plant and Fungal Compounds’, Branigan, T. Czechowski, L. Elias, I. Guterman, [32] F. P. Guengerich, A. W. Munro, J. Biol. Chem. Vol. 6, CRC Press, 1991, p. 217. D. Harvey, P. G. Isaac, A. M. Khan, T. R. Larson, 2013, 288, 17065. [67] A. Takhtajan, ‘Flowering plants’, Springer Y. Li, T. Pawson, T. Penfield, A. M. Rae, D. A. [33] D. Werck-Reichhart, R. Feyereisen, Genome Science & Business Media, 2009. Rathbone, S. Reid, J. Ross, M. F. Smallwood, Biol. 2000, 1, REVIEWS3003. [68] A. F. M. Rizk, Bot. J. Linnean Soc. 1987, 94, 293. V. Segura, T. Townsend, D. Vyas, T. Winzer, D. [34] V. B. Urlacher, M. Girhard, Trends Biotechnol. [69] A. Vasas, J. Hohmann, Chem. Rev. 2014, 114, Bowles, Science 2010, 327, 328. 2012, 30, 26. 8579. [95] T. Townsend, V. Segura, G. Chigeza, T. Penfield, [35] Y. Kallberg, U. Oppermann, H. Jörnvall, B. [70] Q. W. Shi, X. H. Su, H. Kiyota, Chem. Rev. 2008, A. Rae, D. Harvey, D. Bowles, I. A. Graham, Persson, FEBS J. 2002, 269, 4409. 108, 4295. Plos one 2013, 8, e61989. [36] K. Kavanagh, H. Jörnvall, B. Persson, U. [71] G. Appendino, C. Della Porta, G. Conseil, O. [96] M. Peplow, Nature 2016, 530, 389. Oppermann, Cell. Mol. Life Sci. 2008, 65, 3895. Sterner, E. Mercalli, C. Dumontet, A. Di Pietro, [97] K. Jensen, P. E. Jensen, B. L. Moller, Trends [37] S. Okamoto, F. Yu, H. Harada, T. Okajima, J. J. Nat. Prod. 2003, 66 , 140. Plant Sci. 2012, 17, 60. Hattan, N. Misawa, R. Utsumi, FEBS J. 2011, [72] H. E. Johnson, S. A. Banack, P. A. Cox, J. Nat. [98] A. Z. Nielsen, B. Ziersen, K. Jensen, L. M. n. 278, 2892. Prod. 2008, 71, 2041. Lassen, C. E. Olsen, B. L. Møller, P. E. Jensen, [38] M. Gonzalez-Guzman, N. Apostolova, J. M. [73] M. Raisinghani, R. M. Pabbidi, L. S. Premkumar, ACS Syn. Biol. 2013, 2, 308. Belles, J. M. Barrero, P. Piqueras, M. R. Ponce, J. Physiol. 2005, 567, 771. J. L. Micol, R. Serrano, P. L. Rodriguez, Plant [74] M. Ernst, O. M. Grace, C. H. Saslis-Lagoudakis, [99] L. M. n. Lassen, A. Z. Nielsen, B. Ziersen, T. Cell 2002, 14, 1833. N. Nilsson, H. T. Simonsen, N. Ronsted, J. Gnanasekaran, B. L. Møller, P. E. Jensen, ACS [39] L. S. Sarker, M. Galata, Z. A. Demissie, S. S. Ethnopharmacol. 2015, 176, 90. Syn. Biol. 2013, 3, 1. Mahmoud, Arch. Biochem. Biophys. 2012, 528, [75] J. Hohmann, F. Evanics, L. Berta, T. Bartok, [100] Y. Higashi, K. Saito, Plant Cell Environ. 2013, 163. Planta Med. 2000, 66, 291. 36, 1597. [40] A. Atawong, M. Hasegawa, O. Kodama, Biosci. [76] J. Ramsay, A. Suhrbier, J. Aylward, S. Ogbourne, [101] E. Gongora-Castillo, C. R. Buell, Nat. Prod. Rep. Biotechnol. Biochem. 2002, 66, 566. S. J. Cozzi, M. Poulsen, K. Baumann, P. 2013, 30, 490. [41] M. Liu, P. Shi, X. Fu, P. E. Brodelius, Q. Shen, Welburn, G. Redlich, P. Parsons, Br. J. Dermatol. [102] S. S. Bach, J.-É. Bassard, J. Andersen-Ranberg, W. Jiang, Q. He, K. Tang, Plant Cell, Tissue and 2011, 164, 633. M. E. Møldrup, H. T. Simonsen, B. Hamberger, Organ Culture (PCTOC) 2016, 126, 469. [77] N. Kedei, D. J. Lundberg, A. Toth, P. Welburn, S. in ‘Methods in molecular biology’, Springer, [42] G. W. Turner, R. Croteau, Plant Physiol. 2004, H. Garfield, P. M. Blumberg, Cancer Res. 2004, 2014, p. 245. 136, 4215. 64, 3243. [103] K. Geisler, R. K. Hughes, F. Sainsbury, G. P. [43] H. Moummou, Y. Kallberg, L. B. Tonfack, B. [78] R. H. Rosen, A. K. Gupta, S. K. Tyring, J. Am. Lomonossoff, M. Rejzek, S. Fairhurst, C. E. Persson, B. van der Rest, BMC Plant Biol. 2012, Acad. Dermatol. 2012, 66, 486. Olsen, M. S. Motawia, R. E. Melton, A. M. 12, 219. [79] P. Hampson, H. Chahal, F. Khanim, R. Hayden, Hemmings, S. Bak, A. Osbourn, Proc. Natl. [44] S. A. McAdam, F. C. Sussmilch, T. J. Brodribb, A. Mulder, L. K. Assi, C. M. Bunce, J. M. Lord, Acad. Sci. USA 2013, 110, E3360. J. J. Ross, AoB Plants 2015, 7, DOI: 10.1093/ Blood 2005, 106, 1362. [104] H. Duan, M. A. Schuler, Phytochem. Rev. 2006, aobpla/plv091. [80] W. Y. Lee, P. Hampson, L. Coulthard, F. Ali, M. 5, 507. [45] V. Lanzotti, in ‘Natural Products’, Springer, Salmon, J. M. Lord, D. Scheel-Toellner, J. Biol. [105] E. Wang, J. T. Hall, G. J. Wagner, Mol. Breeding 2013, p. 3173. Chem. 2010, 285, 23889. 2004, 13, 49.