Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1724 2022-07-05 14:24:04 |
2 format corrected. + 3 word(s) 1727 2022-07-06 04:17:18 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Koirala, M.;  Karimzadegan, V.;  Liyanage, N.S.;  Mérindol, N.;  Desgagné-Penix, I. Heterologous Host for Alternative Production of Amaryllidaceae Alkaloids. Encyclopedia. Available online: https://encyclopedia.pub/entry/24832 (accessed on 03 July 2024).
Koirala M,  Karimzadegan V,  Liyanage NS,  Mérindol N,  Desgagné-Penix I. Heterologous Host for Alternative Production of Amaryllidaceae Alkaloids. Encyclopedia. Available at: https://encyclopedia.pub/entry/24832. Accessed July 03, 2024.
Koirala, Manoj, Vahid Karimzadegan, Nuwan Sameera Liyanage, Natacha Mérindol, Isabel Desgagné-Penix. "Heterologous Host for Alternative Production of Amaryllidaceae Alkaloids" Encyclopedia, https://encyclopedia.pub/entry/24832 (accessed July 03, 2024).
Koirala, M.,  Karimzadegan, V.,  Liyanage, N.S.,  Mérindol, N., & Desgagné-Penix, I. (2022, July 05). Heterologous Host for Alternative Production of Amaryllidaceae Alkaloids. In Encyclopedia. https://encyclopedia.pub/entry/24832
Koirala, Manoj, et al. "Heterologous Host for Alternative Production of Amaryllidaceae Alkaloids." Encyclopedia. Web. 05 July, 2022.
Heterologous Host for Alternative Production of Amaryllidaceae Alkaloids
Edit

Amaryllidaceae alkaloids (AAs) are plant specialized metabolites with therapeutic properties exclusively produced by the Amaryllidaceae plant family. Bioengineered microbial hosts that grow rapidly can produce plant target specialized metabolites faster as compared to whole plant systems. In addition, the production of plant metabolites in heterologous hosts can reduce downstream extraction process, which eventually becomes more economically sustainable. For the successful synthesis of plant metabolites such as AAs, heterologous hosts require the introduction of reconstructed biosynthetic pathway, requiring key enzymes. This requires comprehensive knowledge of the enzymatic reactions involved in the biosynthesis of the compound of interest in the native host organisms (i.e., plants).

amaryllidaceae alkaloids bioactive molecules biotechnological approach biosynthesis in vitro cultures synthetic biology

1. Molecular Understanding of Amaryllidaceae Alkaloids Biosynthesis

Even though the pharmacological aspect of Amaryllidaceae alkaloids (AAs) has extensively been explored, the full understanding of the AA biosynthetic pathway and the characterization of enzymes responsible for catalyzing the different biosynthetic reactions demand more efforts. This knowledge would enable the establishment of improved systems or sustainable platforms for the production of these valuable biologically active compounds. Combined application of early labeling study followed by latest omics strategies have accelerated the discovery of AAs biosynthetic enzymes [1][2]. After the proposition of the biosynthetic route of different intermediates, several biosynthetic enzymes were predicted based on the nature of the biochemical reaction and by homology with enzymes involved in alkaloid biosynthesis of other plant families. Databases generated from transcriptomic and metabolic analysis of different species of Amaryllidaceae support the presence of different enzyme families involved in AAs pathway [3][4][5][6][7][8][9][10].
The AA biosynthetic pathway utilizes two common amino acids, namely L-tyrosine and L-phenylalanine, as building blocks to produce a vast range of alkaloids with diverse biological activities. The first reactions of AA biosynthesis involve the formation of the ‘precursors’ from the phenylpropanoid and tyramine pathways (Figure 1). As such, L-tyrosine is decarboxylated by the enzyme tyrosine decarboxylase (TYDC) to yield tyramine while the production of the second building block, 3,4-dihydroxybenzaldehyde (3,4- DHBA), is achieved via the phenylpropanoid pathway by the action of enzymes such as phenylalanine ammonia-lyase (PAL), cinnamate 4-hydoxylase (C4H), p-coumarate 3-hydroxylase (C3H), to name but a few. TYDC was characterized from Lycoris radiata, a galanthamine producing Amaryllidaceae plant [11]. The functional characterization of PAL and C4H in L. radiata was reported using heterologous expression in bacteria [6].
Figure 1. Biosynthetic routes to main types (boxed) of Amaryllidaceae alkaloid (AA). Arrows without labeling reflect chemical reactions where no enzyme was characterized. Enzymes that have been identified are labeled in blue. A solid arrow shows one enzymatic step, whereas a broken arrow symbolizes multiple enzymatic reactions. Following 4′O-methylnorbelladine, the regioselective phenol-phenol’ coupling reaction is indicated in the broken arrow, leading to various AA-types. Enzyme abbreviations: 3,4-DHBA, 3,4-dihydroxybenzaldehyde; PAL, phenylalanine ammonia-lyase; C4H, cinnamate 4-hydroxylase; C3H, coumarate 3-hydroxylase; TYDC, tyrosine decarboxylase; NBS, norbelladine synthase; NR, noroxomaritidine/norcraugsodine reductase; N4OMT, norbelladine 4′-O-methyltransferase; CYP96T1, cytochrome P450 monooxygenase 96T1.
Despite having a remarkable diversity in structure and biological activity, all AAs are derived from a common intermediate, norbelladine. The condensation of tyramine and 3,4-DHBA yields norbelladine and was shown to be catalyzed either by norbelladine synthase (NBS) or by noroxomaritidine/norcraugsodine reductase (NR), in both cases with low yield [6][12]. NBS was characterized from N. pseudonarcisus king Alfred and L. aestivum [6]. GFP-tagged LaNBS and CFP-tagged NR showed that both enzymes are localized to the cytosol, which suggests that the first committed step of AA biosynthesis probably occurs in the cytosol [6][13].
Norbelladine can either be utilized directly to generate norbelladine- and cherylline-type AAs or be further methylated by norbelladine 4′-O-methyltransferase (N4OMT) to give 4′-O-methylnorbelladine (Figure 1). The structural feature of cherylline-type of AAs suggests the occurrence of 3′-O-methylation during the biosynthesis of these types of AAs, although it remains to be proven. The specific synthesis of both 3′-O-methylated and 4′-O-methylated AAs suggest that regioselective methylation is important to determine the types of the end product of AAs biosynthesis route. The characterization of norbelladine OMT from Narcissus sp. aff. pseudonarsissus suggests that methylation by NpN4OMT happens specifically at 4′-O position of norbelladine [7]. However, later studies on L. radiata OMT (LrOMT) propose that methylation can occur either in the 3′-O or 4′-O position of norbelladine, 3,4-DHBA, or caffeic acid. Kinetic study of LrOMT indicates that it has a higher affinity for 3,4-DHBA as substrate compared to norbelladine. The methylated forms of 3,4-DHBA (i.e., vanillin and isovanillin) could also be condensed with tyramine to generate 3′ or 4′-O-methylnorbelladine. However, up until now, none of the possible methylated forms of 3, 4-DHBA were tested as a substrate for NBS.
One step deeper in the AA pathway, and depending on the type of phenol-coupling reaction, the 4′-O-methylnorbelladine can be directed to 1) galanthamine-type through para-ortho’, 2) lycorine-type AAs by ortho-para’, and 3) crinine-type of AAs by para-para’ phenol coupling reactions (Figure 1). These types of C-C phenol-coupling reactions are putatively catalyzed by members of the cytochrome P450 enzyme family. For example, NpCYP96T1 was shown to catalyze the para-para’ oxidative reaction of 4′-O-methylnorbelladine into noroxomaritidine and was also shown to catalyzed formation of the para-ortho’ phenol coupled product, N-demethylnarwedine, as less than 1% of the total product [14]. Aside from CYP96T1 and NR, there are no other steps (genes or enzymes) that have been identified in the formation of phenol-coupled AA-types to date (Figure 1).
Plants synthesize specialized metabolites by using complex biosynthetic routes that derive from primary metabolic pathways. AAs biosynthesis is a multifaceted process that involves different regulatory elements and gene functions. The expression of certain genes involved in plant metabolism also changes with different climatic and environmental factors [15]. Furthermore, it also varies within different developmental stage of plant [3]. It remains challenging to correlate gene expression and metabolite accumulation in planta, as the site of metabolite synthesis may differ from the site of accumulation. For example, nicotine biosynthesis occurs in the root of tobacco but accumulates in the aerial part of the plant [16], whereas morphine biosynthesis starts in sieve elements of the phloem but accumulates in adjacent laticifers cells in opium poppy [17]. As such, in vitro cultures have been an essential tool to decipher the alkaloid biosynthesis pathway. In 2011, Tahchy et al. used deuterium-labeled precursors fed to in vitro cultures of L. aestivum. In this study, the authors followed the transfer of labeled precursor 4′-O-methyl-d3-norbelladine from media into shoot and then its metabolization into lycorine and galanthamine. This research demonstrated that 4′-O-methylated-norbelladine was a key intermediate AAs [18]. Until now, AAs specific genes such as NBS, N4OMT, CYP96T1, and NR have been characterized and confirmed from Leucojum sp., Narcissus sp., Lycoris sp. cultures [3][19][20][21], however, the researchers' molecular understanding regarding this complex biosynthesis route of AAs and its regulation is still unclear. Furthermore, the pattern of relative expression of putative AAs biosynthetic genes (in fields versus in vitro and in differentiated versus undifferentiated tissues of Narcissus development) added some clear knowledge regarding their role in alkaloid biosynthesis [22]. A study performed on callus culture of L. radiata showed how different factors, such as temperature (cold treatment), osmotic pressure (PEG treatment), or elicitor treatment (methyl jasmonate), can influence LrOMT gene expression pattern [23]. Thus, in vitro system cultures are a powerful tool to uncover AAs biosynthesis and gene regulation that should be thoroughly exploited.

2. Synthetic Biology for AA Biosynthesis

Although the complete biosynthetic pathway of AAs is not resolved, and up to now the AAs demand has not been sufficiently fulfilled by a plant source, a synthetic biological approach could be a powerful approach to produce AAs. Recent achievements in synthetic biological approaches include the production of complex biomolecules such as noscapine (a benzylisoquinoline alkaloid from opium poppy) and its halogenated derivatives (anticancer) in Saccharomyces cerevisiae, assembling 30 biosynthetic enzymes from plant, bacteria, and mammal, with yeast itself including seven plant endoplasmic reticulum localized genes [24]. This success gives hope for producing complex biomolecules such as AAs by using a synthetic biological approach.
Proper selection of host organism is the starting point of synthetic biological approach. The chosen organism should be producing (or easily modified to produce) enough core metabolites such as aromatic amino acids, L-phenylalanine, and L-tyrosine, precursors needed for the biosynthesis of target specialized metabolite such as AAs. Selection of host species will also rely on prior knowledge of their ease of engineering, established cloning tools, culture techniques, and possibly scaling up to industrial requirements. Due to rapid growth and easy handling, microbial hosts such as yeast (Saccharomyces cerevisiae), and to a lesser extent Escherichia coli, were used to produce plant-derived high value alkaloids like morphinan alkaloids [25][26][27]. Furthermore, the production of aromatic amino acid (precursor for AAs) and associated upstream gene/enzyme were well studies in these hosts [28]. Precursors such as L-tyrosine and p-coumaric have been already produced in E. coli [29][30]. Recently, unicellular photosynthetic organisms such as microalgae and cyanobacteria became interesting research platforms because of their unicellular physiology, together with their photosynthetic, heterotrophic, and mixotrophic lifestyles [31]. Moreover, plant-based genetic engineering technique is also emerging in model plants such as Nicotiana tabacum and N. benthamiana [32].
Once a host organism is selected, availability of precursor molecules can be enhanced by modifications to its metabolic pathway, such as gene deletions, swapping of endogenous enzymes with more active homologues, or overexpression of endogenous metabolic genes. Then, a route to the desired specialized metabolites can be planned and implemented. A candidate pathway is first outlined through selection of stepwise chemical intermediates leading from host metabolism to the target compound, followed by selection of enzymes to carry out each specified reaction [33][34]. Even though the lack of knowledge in the AAs biosynthetic pathway hinders this approach as of yet, it could be partially overcome by creating libraries of gap-filling genes candidate generated from huge plant transcriptomic database, as available for thousands of plants or as part of the PhytoMetaSyn project [35]. In addition, the decrease in the cost of DNA synthesis helps accelerate gene characterization from its native source and ultimately facilitate the production of complex biomolecules like AAs [35][36][37]. Such work was done to produce polyketides. Soon, platform of synthetic approach will not only provide techniques to produce AAs but also help in the biosynthesis of novel AAs derivatives with improved biological and physiological properties. In example, once the complete identification of genes encoding enzymes required for the biosynthesis of galanthamine is achieved, one more enzyme could be added in the transgenic construct that could involve glycosylation, shifting the polarity of parent molecule, and eventually improving drug uptake by the human body.

References

  1. Desgagné-Penix, I. Biosynthesis of alkaloids in Amaryllidaceae plants: A review. Phytochem. Rev. 2020, 20, 409–431.
  2. Kilgore, M.B.; Kutchan, T.M. The Amaryllidaceae alkaloids: Biosynthesis and methods for enzyme discovery. Phytochem. Rev. Proc. Phytochem. Soc. Eur. 2016, 15, 317–337.
  3. Hotchandani, T.; de Villers, J.; Desgagne-Penix, I. Developmental Regulation of the Expression of Amaryllidaceae Alkaloid Biosynthetic Genes in Narcissus papyraceus. Genes 2019, 10, 594.
  4. Li, Q.; Xu, J.; Zheng, Y.; Zhang, Y.; Cai, Y. Transcriptomic and Metabolomic Analyses Reveals That Exogenous Methyl Jasmonate Regulates Galanthamine Biosynthesis in Lycoris longituba Seedlings. Front. Plant Sci. 2021, 12, 713795.
  5. Singh, A.; Desgagne-Penix, I. Transcriptome and metabolome profiling of Narcissus pseudonarcissus ‘King Alfred’ reveal components of Amaryllidaceae alkaloid metabolism. Sci. Rep. 2017, 7, 17356.
  6. Tousignant, L.; Diaz-Garza, A.M.; Majhi, B.B.; Gelinas, S.E.; Singh, A.; Desgagne-Penix, I. Transcriptome analysis of Leucojum aestivum and identification of genes involved in norbelladine biosynthesis. Planta 2022, 255, 30.
  7. Kilgore, M.B.; Augustin, M.M.; Starks, C.M.; O’Neil-Johnson, M.; May, G.D.; Crow, J.A.; Kutchan, T.M. Cloning and Characterization of a Norbelladine 4′-O-Methyltransferase Involved in the Biosynthesis of the Alzheimer’s Drug Galanthamine in Narcissus sp. aff. pseudonarcissus. PLoS ONE 2014, 9, e103223.
  8. Park, C.H.; Yeo, H.J.; Park, Y.E.; Baek, S.A.; Kim, J.K.; Park, S.U. Transcriptome Analysis and Metabolic Profiling of Lycoris Radiata. Biology 2019, 8, 63.
  9. Hu, J.; Li, W.; Liu, Z.; Zhang, G.; Luo, Y. Molecular cloning and functional characterization of tyrosine decarboxylases from galanthamine-producing Lycoris radiata. Acta Physiol. Plantarum 2021, 43, 84.
  10. Li, Q.; Xu, J.; Yang, L.; Zhou, X.; Cai, Y.; Zhang, Y. Transcriptome Analysis of Different Tissues Reveals Key Genes Associated With Galanthamine Biosynthesis in Lycoris longituba. Front. Plant Sci. 2020, 11, 519752.
  11. Singh, A.; Massicotte, M.A.; Garand, A.; Tousignant, L.; Ouellette, V.; Berube, G.; Desgagne-Penix, I. Cloning and characterization of norbelladine synthase catalyzing the first committed reaction in Amaryllidaceae alkaloid biosynthesis. BMC Plant Biol. 2018, 18, 338.
  12. Kilgore, M.B.; Holland, C.K.; Jez, J.M.; Kutchan, T.M. Identification of a Noroxomaritidine Reductase with Amaryllidaceae Alkaloid Biosynthesis Related Activities. J. Biol. Chem. 2016, 291, 16740–16752.
  13. Majhi, B.B.; Gélinas, S.-E.; Mérindol, N.; Desgagne-Penix, I. Characterization of norbelladine synthase and noroxomaritidine/norcraugsodine reductase reveals a novel catalytic route for the biosynthesis of Amaryllidaceae alkaloids including the Alzheimer’s drug galanthamine. Plant J. 2022; submitted.
  14. Kilgore, M.B.; Augustin, M.M.; May, G.D.; Crow, J.A.; Kutchan, T.M. CYP96T1 of Narcissus sp. aff. pseudonarcissus Catalyzes Formation of the Para-Para’ C-C Phenol Couple in the Amaryllidaceae Alkaloids. Front. Plant Sci. 2016, 7, 225.
  15. Ncube, B.; Nair, J.J.; Rárová, L.; Strnad, M.; Finnie, J.F.; Van Staden, J. Seasonal pharmacological properties and alkaloid content in Cyrtanthus contractus N.E. Br. S. Afr. J. Botany 2015, 97, 69–76.
  16. Katoh, A.; Ohki, H.; Inai, K.; Hashimoto, T. Molecular regulation of nicotine biosynthesis. Plant Biotechnol. 2005, 22, 389–392.
  17. Onoyovwe, A.; Hagel, J.M.; Chen, X.; Khan, M.F.; Schriemer, D.C.; Facchini, P.J. Morphine biosynthesis in opium poppy involves two cell types: Sieve elements and laticifers. Plant Cell 2013, 25, 4110–4122.
  18. El Tahchy, A.; Ptak, A.; Boisbrun, M.; Barre, E.; Guillou, C.; Dupire, F.; Chretien, F.; Henry, M.; Chapleur, Y.; Laurain-Mattar, D. Kinetic study of the rearrangement of deuterium-labeled 4′-O-methylnorbelladine in Leucojum aestivum shoot cultures by mass spectrometry. Influence of precursor feeding on amaryllidaceae alkaloid accumulation. J. Nat. Prod. 2011, 74, 2356–2361.
  19. Ren, Z.; Lin, Y.; Lv, X.; Zhang, J.; Zhang, D.; Gao, C.; Wu, Y.; Xia, Y. Clonal bulblet regeneration and endophytic communities profiling of Lycoris sprengeri, an economically valuable bulbous plant of pharmaceutical and ornamental value. Sci. Hortic. 2021, 279, 109856.
  20. Ka, S.; Masi, M.; Merindol, N.; Di Lecce, R.; Plourde, M.B.; Seck, M.; Gorecki, M.; Pescitelli, G.; Desgagne-Penix, I.; Evidente, A. Gigantelline, gigantellinine and gigancrinine, cherylline- and crinine-type alkaloids isolated from Crinum jagus with anti-acetylcholinesterase activity. Phytochemistry 2020, 175, 112390.
  21. Fennell, C.W.; Elgorashi, E.E.; van Staden, J. Alkaloid production in Crinum moorei cultures. J. Nat. Prod. 2003, 66, 1524–1526.
  22. Aleya, F.; Xianmin, C.; Anthony, H.; Meriel, J. Relative expression of putative genes involved in galanthamine and other Amaryllidaceae alkaloids biosynthesis in Narcissus field and in vitro tissues. Gene 2021, 774, 145424.
  23. Sun, B.; Wang, P.; Wang, R.; Li, Y.; Xu, S. Molecular Cloning and Characterization of a meta/para-O-Methyltransferase from Lycoris aurea. Int. J. Mol. Sci. 2018, 19, 1911.
  24. Li, Y.; Li, S.; Thodey, K.; Trenchard, I.; Cravens, A.; Smolke, C.D. Complete biosynthesis of noscapine and halogenated alkaloids in yeast. Proc. Natl. Acad. Sci. USA 2018, 115, E3922–E3931.
  25. Diamond, A.; Desgagne-Penix, I. Metabolic engineering for the production of plant isoquinoline alkaloids. Plant Biotechnol. J. 2016, 14, 1319–1328.
  26. Facchini, P.J.; Bohlmann, J.; Covello, P.S.; De Luca, V.; Mahadevan, R.; Page, J.E.; Ro, D.K.; Sensen, C.W.; Storms, R.; Martin, V.J. Synthetic biosystems for the production of high-value plant metabolites. Trends Biotechnol. 2012, 30, 127–131.
  27. Fossati, E.; Narcross, L.; Ekins, A.; Falgueyret, J.P.; Martin, V.J. Synthesis of Morphinan Alkaloids in Saccharomyces cerevisiae. PLoS ONE 2015, 10, e0124459.
  28. Cao, M.; Gao, M.; Suástegui, M.; Mei, Y.; Shao, Z. Building microbial factories for the production of aromatic amino acid pathway derivatives: From commodity chemicals to plant-sourced natural products. Metab. Eng. 2020, 58, 94–132.
  29. Li, Y.; Li, J.; Qian, B.; Cheng, L.; Xu, S.; Wang, R. De Novo Biosynthesis of p-Coumaric Acid in E. coli with a trans-Cinnamic Acid 4-Hydroxylase from the Amaryllidaceae Plant Lycoris aurea. Molecules 2018, 23, 3185.
  30. Patnaik, R.; Zolandz, R.R.; Green, D.A.; Kraynie, D.F. L-tyrosine production by recombinant Escherichia coli: Fermentation optimization and recovery. Biotechnol. Bioeng. 2008, 99, 741–752.
  31. Slattery, S.S.; Diamond, A.; Wang, H.; Therrien, J.A.; Lant, J.T.; Jazey, T.; Lee, K.; Klassen, Z.; Desgagne-Penix, I.; Karas, B.J.; et al. An Expanded Plasmid-Based Genetic Toolbox Enables Cas9 Genome Editing and Stable Maintenance of Synthetic Pathways in Phaeodactylum tricornutum. ACS Synth. Biol. 2018, 7, 328–338.
  32. Farhi, M.; Marhevka, E.; Ben-Ari, J.; Algamas-Dimantov, A.; Liang, Z.; Zeevi, V.; Edelbaum, O.; Spitzer-Rimon, B.; Abeliovich, H.; Schwartz, B.; et al. Generation of the potent anti-malarial drug artemisinin in tobacco. Nat. Biotechnol. 2011, 29, 1072–1074.
  33. Kitney, R.; Freemont, P. Synthetic biology—The state of play. FEBS Lett. 2012, 586, 2029–2036.
  34. Auslander, S.; Auslander, D.; Fussenegger, M. Synthetic Biology-The Synthesis of Biology. Angew. Chem. Int. Ed. Engl. 2017, 56, 6396–6419.
  35. Matasci, N.; Hung, L.H.; Yan, Z.; Carpenter, E.J.; Wickett, N.J.; Mirarab, S.; Nguyen, N.; Warnow, T.; Ayyampalayam, S.; Barker, M.; et al. Data access for the 1000 Plants (1KP) project. Gigascience 2014, 3, 17.
  36. Xiao, M.; Zhang, Y.; Chen, X.; Lee, E.J.; Barber, C.J.; Chakrabarty, R.; Desgagne-Penix, I.; Haslam, T.M.; Kim, Y.B.; Liu, E.; et al. Transcriptome analysis based on next-generation sequencing of non-model plants producing specialized metabolites of biotechnological interest. J. Biotechnol. 2013, 166, 122–134.
  37. Pyne, M.E.; Narcross, L.; Martin, V.J.J. Engineering Plant Secondary Metabolism in Microbial Systems. Plant Physiol. 2019, 179, 844–861.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 482
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 06 Jul 2022
1000/1000
Video Production Service