Open access peer-reviewed chapter

Chiral Inversion of Active Compounds in Plant Extract

Written By

Ngoc-Van Thi Nguyen

Submitted: 30 December 2021 Reviewed: 07 January 2022 Published: 12 February 2022

DOI: 10.5772/intechopen.102537

From the Edited Volume

Secondary Metabolites - Trends and Reviews

Edited by Ramasamy Vijayakumar and Suresh Selvapuram Sudalaimuthu Raja

Chapter metrics overview

228 Chapter Downloads

View Full Metrics

Abstract

Chiral inversion is always mediated by enzymes and varies with solvent, pH and temperature. Considerable attention should be paid to the mechanism of the inversion reaction and its pharmacological and toxicological results. This chapter will discuss the mechanism of chiral inversion of plants in secondary metabolize and its importance in creating pharmacology consequences. Plant stereoisomers of alkaloids and flavonoids exhibit a wide range of pharmacological effects. Recent advances in chiral analysis for the herbal plants in clinical research & forensic toxicology by experiments in which one enantiomer was given to the experiment subjects in a specific situation. Demonstration of metabolic chiral inversion may have consequences for the development of a new pharmaceutical entity. Hence, it helps a better understanding of chiral compounds in plants, facilitating the application for drug development from medicinal herbs and thereby reducing bioanalytical and toxicology workload.

Keywords

  • chiral inversion of plants
  • eutomer
  • distomer
  • racemization

1. Introduction

Chiral inversion is the process by which enzymes modify the three-dimensional structure of a molecule by converting one enantiomer to its antipode [1]. Racemization occurs when isomerization leads in the creation of a racemic mixture. As a result, chiral inversion influences drug stability throughout drug discovery and development. Biological activity, toxicity, shelf-life and dosage of the compound are affected by the stability of the drug [2]. The process of chiral inversion is affected by a lot of variables, consequently, the strength of chiral inversion under different situations and in various substances can vary significantly. The primary elements that were acknowledged to play a vital part in the process of chiral inversion were reported to be interspecies differences and tissue types. Some recent researches have demonstrated that additional variables, such as administration route or interaction with other xenobiotics, can also impact enantiomeric conversion.

Plants create a vast diversity of physiologically active metabolites, many of which have stereochemical variants on the same molecular scaffold. These alterations in stereochemistry have a significant influence on biological function. Notably, plant stereoisomers of alkaloids and flavonoids exhibit a wide range of pharmacological effects. Alkaloids are cyclic chemical molecules with a negative oxidation state of nitrogen. They are found throughout the flora and play an important function in plant protection, sprouting, and encouraging plant development. Plants containing alkaloids are frequently employed as traditional remedies, and these chemicals typically have specific pharmacological actions. The majority of alkaloids are in a chiral form which is often appeared in products as racemic compounds, while their enantiomers have been proved to have different pharmacological actions [3]. Flavonoids are a vast category of polyphenolic chemicals with a benzo—pyrone structure that is found in all plants. Phenylpropanoid is their produce’s pathway. Recent interest in these chemicals has been sparked because of the possible health advantages of these antioxidant polyphenolic compounds [4]. The relevance of racemic flavanones stereospecific pomological disposition has been determinated and described in the last 20 years. The majority of these studies report on the measurement of flavanones in citrus fruit juices and herbs [5].

Can all chiral compounds undergo chiral inversion? Maybe no, many compounds still can be considered stable in metabolize process. Why are some enantiomers of plants inverted by enzymes and others are not attacked? The reason lies in the structure. The intent of this chapter is to provide a comprehensive, rather than an exhaustive, appraisal of chiral bio-inversion. This chapter will discuss enzymatic chiral inversion of plants in secondary metabolize and its importance create pharmacology effect. Therefrom, it helps a better understanding of chiral compounds in plants, facilitating the application for drug development from medicinal herbs.

Advertisement

2. Mechanism of inversion

Under selective conditions, racemization or enantiomerization defined as the chiral conversion of enantiomer into its antipode may present in many plants metabolizing. When the chiral molecule enantiomers in herbals interact with a chiral macromolecule-like enzyme, they generate a pair of diastereoisomeric complexes that vary energetically. It is not surprising, then, that the results of enzyme-mediated reactions performed on a pair of enantiomers may differ in type and/or extent. Indeed, given the structure of the enzyme-substrate complex, it is plausible to believe that enantioselectivity is the rule rather than the exception in metabolism. Likewise, the binding of a prochiral substrate to an enzyme may orient two enantiotopic groups differently about the enzyme catalytic site, causing these two groups to become diastereotopic within the enzyme-substrate complex. It’s simple to see how the production of a chiral metabolite from a prochiral substrate may result in stereoselectivity for one isomeric product [6].

At the substrate and product levels, xenobiotic metabolic reactions exhibit two forms of stereoselectivity. As a result, they can be classed according to their stereoselectivity or, if such selectivity is complete, their stereospecificity. Caution while using this latter phrase, because the ability to determination “specificity” is clearly dependent on the analytical approach of the research. The words substrate and product “stereospecificity” were initially introduced to the enzyme-mediated reduction of ketones by Prelog [7], and were later extended to drug metabolic processes by Jenner and Testa [8]. Substrate stereoselectivity is the preferred metabolism of one of two stereoisomers over the other, whereas product stereoselectivity is the preferential production of one stereoisomer over the other stereoisomers that may exist. These two “selectivities” may be so closely related that substrate-product stereoselectivity, i.e., the selective metabolism of one of a pair of enantiomers to form one of several possibly diastereoisomeric products, may also be seen. If the enantiomeric composition of medication or metabolite is detected in the analysis process, data collected from in vivo research on stereochemistry in plants must be regarded with caution (Table 1).

PlantEnzymeStereoselectiveStereospecificityRef.
Alkaloid Compound
Opium poppy1,2-dehydroreticuline reductase(R)-reticuline[9]
Catharanthus roseustetrahydroalstonine synthase(3S,19S,20S)-Tetrahydroalstonine[10]
Claviceps purpureaDimethylallyl tryptophan synthaseL-tryptophan[11]
Hyoscyamus nigerHyoscyamine 6β-hydroxylaseL-hyoscyamine[12]
Berberis koetineanaTetrahydrobenzyliso-quinoline-N-methyltransferase(R)-tetrahydropapaverine[13]
Flavonoid Compound
SoybeanChalcone isomerase(2S)-flavanone[14]
Dahlia variabilisFlavanone 4-reductase(2S)-flavanone(2S, 4R)-flavan-4-ol[15]
Citrus unshiuFlavonol synthase(2R,3R)-dihydroflavonol[16]
Glycyrrhiza echinataFlavanone 2-hydroxylase(2S)-flavanone[17]
Ginko biloba
Pseudotsuga menziesii
Dihydroflavonol 4-reductase(2R,3R)-dihydroflavonol(2R, 3S, 4S)-flavan-2,3-trans-3,4-cis-diol[18]
Medicago truncatulaAnthocyanidin reductase(2R, 3R)-flavan-3-ol[19]
Medicago sativaIsoflavone reductase(2R)-isoflavanone[20]
Pisum sativumHydroxymaackiain-3-Omethyltransferase(+)-6a-hydroxymaackiain[21]
Desmodium uncinatumLeucoanthocyanidin 4-reductaseflavan-2,3-trans-3,4-cisdiol(2R, 3S)-flavan-3-ol[22]

Table 1.

Stereoselective and/or specific enzymes of alkaloid and flavonoid compound biosynthesis in plant extract.

2.1 Alkaloids

Plants are thought to generate over 12,000 distinct alkaloids, which may be classified based on their carbon skeleton structures. Many catalytic stages in alkaloid biosynthesis in plants are catalyzed by enzymes from various protein families.

Since the discovery of morphine in 1806, the complex relationships between opium poppy and the human condition have fueled substantial study into the production of morphinan alkaloids [23]. During the 1960s, significant progress toward route elucidation was made, which supported a major theory [24] that morphine was generated by 1-benzylisoquinoline alkaloid metabolism [25]. Because only the (R)-conformer could undergo additional phenol coupling to the morphinan scaffold, (S)-reticuline emerged as the primary 1-benzylisoquinoline intermediate, with its stereochemical inversion to (R)-reticuline thought to be a critical gateway reaction [26].

The pathway makes use of opium poppy reticuline epimerase, a multi-domain protein composed of the P450 CYP82Y2 linked to an aldo-keto reductase (AKR). CYP82Y2 (1,2-dehydroreticuline synthase, DRS) catalyzes the conversion of (S)-reticuline to 1,2-dehydroreticuline, which is then converted to (R)-reticuline by the AKR module (1,2-dehydroreticuline reductase, DRR) [26]. A second P450 called CYP719B1 then tranforms (R)-reticuline into salutaridine [27, 28]. This procedure includes (R)-reticuline twisting, reorientating and oxidative C–C bond coupling stimulated by CYP719B1 (Figure 1).

Figure 1.

Proposed chiral inversion of (S)-reticulin to (R)-reticulin catalyzed by 1,2- dehydroreticuline reductase (DRR) and 1,2-dehydroreticuline reductase (DRR) in opium poppy [9].

Catharanthus roseus, a medicinal plant, creates three of these isomers: ajmalicine (raubasine), tetrahydroalstonine, and 19-epi-ajmalicine (mayumbine) (Figure 2) [30]. These heteroyohimbines are produced from deglycosylated strictosidine (strictosidine aglycone), as are the bulk of monoterpene indole alkaloids [31]. A glucose unit removal from strictosidine by strictosidine glucosidase (SGD) leads to the formation of a reactive dialdehyde intermediate that can rearrange to generate a variety of isomers [32]. The stability of these isomers by enzyme-catalyzed reduction is thought to be the first step toward the vast chemical variety found in monoterpene indole alkaloids. The tetrahydroalstonine synthase (THAS) is a zinc-dependent medium-chain dehydrogenase/reductase (MDR) that manufactures the heteroyohimbine tetrahydroalstonine (Figure 2) [33]. Although, these studies showed that THAS is an important enzyme for the heteroyohimbine production, the mechanism by which this enzyme controls the stereoselectivity of the reduction remained unexplained. Moreover, the fact that strictosidine aglycone is also a predrug of some alkaloid scaffolds so constitutes a major branch point in the monoterpene indole alkaloid biosynthesis process [29].

Figure 2.

Heteroyohimbine alkaloid biosynthesis. Red highlighted compounds indicate the three diastereomers identified in Catharanthus roseus. Alkaloids derived from heteroyohimbines are also illustrated [29].

2.2 Flavonoids

Most flavonoid biosynthesis enzymes are extremely stereoselective and/or stereospecific; nonetheless, this assertion is based on just one or a few published findings for numerous enzymes. Flavonoids are produced by the phenylpropanoid pathway, which begins with the enzyme L-phenylalanine ammonia-lyase deamination of phenylalanine (PAL). D-phenylalanine is not a substrate for PAL; it is selective for the naturally occurring L-isomer of phenylalanine [34]. The process mediated by chalcone–flavanone isomerase (CHI), which sets the stereochemistry at C-2 of the flavonoid heterocyclic ring, maybe the most stereo-chemically crucial in flavonoid biosynthesis. CHI is a chemically and structurally well-characterized enzyme that creates (2S)-flavanones from chalcones (Figure 3) [14, 35].

Figure 3.

General outline of the flavonoid pathway (PAL: Phenylalanine ammonia-lyase, CHS: Chalcone synthase, CHI: Chalcone isomerase, FHT: Flavanone 3β-hydroxylase, FNS Ι: Flavone synthase Ι, FLS: Flavonols synthase, DFR: Dihydroflavonols reductase, ANS: Anthocyanidin synthase). Chiral inversion in flavonoid metabolizes was highlighted by red frame.

Unlike other flavonoid enzymes such as PAL or CHI, the 2-oxoglutarate-dependent dioxygenases flavonol synthase (FLS) and anthocyanidin synthase (ANS) have wide substrate and product selectivities in vitro (both take flavanone, dihydroflavonol, and leucoanthocyanidin as substrates). Prescott et al. have reported a detailed structural and in vitro research on recombinant flavonol synthase from Arabidopsis thaliana, with a focus on the stereochemistry of substrate and product, have provided information on how they catalyze reactions with their real substrates in vivo [36]. FLS and ANS prefer substrates with natural C-2 and C-3 stereochemistry [(i.e. (2R,3R)- dihydroquercetin for FLS and (2R,3S, 4R/S)- leucoanthocyanin for ANS], but hydroxylate both (2R)- and (2S)-naringenin equally well in vitro, indicating that the C-3 hydroxyl group is important in biasing substrate selectivity [37].

The flavan-3-ols (+)-catechin and (−)-epicatechin serve as the foundation for proanthocyanidins (condensed tannins), a family of molecules of great interest due to their effects on animal health [38]. The C-2 and C-3 stereochemistries of (+)-catechin (2,3-trans) are identical to those of flavonoid pathway intermediates, and a pathway leading from (2R, 3S, 4S)-leucoanthocyanidin to (+)-catechin, catalyzed by leucoanthocyanidin reductase (LAR), has been illustrated and affirmed by the cloning of a leucoanthocyanidin reducta [22]. LAR belongs to the Reductase–Epimerase–Dehydrogenase protein family, which also includes isoflavone reductase and similar homologs (Figure 4) [39].

Figure 4.

The pro-anthocyanidin pathway showing the LAR reaction.

The process catalyzed by anthocyanidin reductase (ANS) and anthocyanidin reductase (ANR) leads from leucocyanidin to (−)-epicatechin [40]. By operating on an achiral intermediate, ANR, an enzyme with limited sequence similarity to dihydroflavonol reductase, can introduce the 2,3-cis stereochemistry (anthocyanidin). Mechanisms for this reaction have been hypothesized, and it is plausible that more ANR-like enzymes with the potential to introduce different stereochemistries exist (Figure 5) [41].

Figure 5.

Pathway for CT biosynthesis placing BAN immediately downstream of ANS.

Advertisement

3. Factors affecting chiral inversion

Chiral inversion is always mediated by enzymes and varies with solvent, pH and temperature. When a molecule has two or more elements of chirality, one of which is configurationally labile, enantiomerization can occur. Many studies have been reported about the chiral compounds inversion such as: evodiamine in Evodia rutaecarpa [42], ephedrine and atropine (Figures 6 and 7) [43].

Figure 6.

Structures of (A): (1R,2S)-(−)-ephedrine and (1S,2R)-(+)-ephedrine; (B); (S)-(−)- hyoscyamine and (R)-(+)-hyoscyamine.

Figure 7.

Chemical structure of (1a) R-(−)-evodiamine, (1b) S-(+) evodiamine.

Chiral inversion is always mediated by enzymes. One of the most valuable synthetic features of enzymes is their ability to discriminate between enantiomers of racemic substrates [44]. The ratio of stereoisomers is changed mainly by stereospecificity and stereoselectivity of enzymes transformation. The stereoselectivity and stereospecificity of enzymes change dramatically the ratio of drug enantiomers and metabolites enantiomers in biological systems. The enzyme-mediated chiral inversion can be affected by determining expression, substrate affinity and activity of the enzyme. The difference of species and tissue can be different in the rate of the chiral inversion occursion as well as of the routes and mechanisms of inversion [2].

On another hand, the development of strategies that improves the stereoselectivity of enzyme-catalyzed resolutions has been researching. Modification of the substrate, recycling of the product and changing of the reaction conditions are the three most common ways. From now, even enzymes with modest stereoselectivity can be used successfully [44]. Configurational stability depends mainly on the structure and the conditions, especially with solvent, pH and temperature [2].

According to Ngoc Van Thi Nguyen et al. (2013) research, extraction conditions are also can affect the enantiomerization while this study investigated the optimization of the extraction procedure, more specifically the solvent, pH and temperature [42].

3.1 Solvent

In the metabolic chiral inversion research, avoiding spontaneous or chemical racemization of enantiomers is one of the important things [2]. The organic solvent characterism is one other parameter that can significantly interfere with this chiral inversion [45]. The study of Yang SK [46] has shown that racemization half-lives t1/2 of enantiomeric oxazepam were 4.8 min in methanol, while it was 840 min in diethyl ether, and 5000 min in hexane, 4500 min in acetonitrile, etc.

3.2 pH

Based on the result of the study of Glass Amanda M. et al. (2012), the data collectively prove that pH has a minute effect on the chiral inversion rate (Figure 8) [48].

Figure 8.

Time-dependent changes of D-luciferin substrate. Luciferin racemization under various pHs of 150 mM GTA buffer, under ddH2O, and under a medium was monitored for 12 days. The results are highlighted by colors: pH 5 (purple), pH 6 (blue), pH 7 (orange), pH 8 (wine red), pH 8.5 (green), ddH2O (pink) and DMEM (brown). Even under acidic to neutral conditions, obvious racemization that could not be ignored for long-term experiments were observed. The best condition for inhibiting racemization to maintain D-luciferin optical purity was dissolution in ddH2O [47].

The pH effect on proton extraction to give the enolate-form of CoA-thioester resulting in chiral inversion [47]. Chiral inversion and sufficient emission intensity were observed at basic pH 8 and 8.9, respectively, whereas only little emission was observed under neutral to acidic conditions.

3.3 Temperature

Enzyme activity is also affected by temperature, which can lead to the chiral inversion efficiency. The research of the effect of temperature on enzyme activity showed that the hydrogen peroxidase activity’s best temperature is 41°C. When this condition is decreased to 37° C, the enzyme activity decreased. Continuing to decrease to 9°C can decrease dramatically the activity of the enzyme. The influence on enzyme flexibility is because of the temperature effect on hydrogen bonds and covalent (Figure 9) [49].

Figure 9.

The effect of temperature on enzyme activity [49].

Advertisement

4. Pharmacological consequences

One of the three majorities of racemic pharmaceuticals are the racemic drugs that only have one eutomer, but the distomer could be transformed into its bioactive antipode by chiral inversion in the body (Table 2) [60].

PlantStereoisomer compoundPharmacological effectTest modelRef.
Alkaloids
Huperzia serrataHuperzine A and BAnticholinesterase activityAcetylcholinesterase (AChE) inhibitory assay[50]
Narcissus jonquilla quailJonquailineAnticancerHuman cancer cell line: A549; OE21; Hs683 U373;SKMEL B16F10[51]
Uncaria rhynchophyllaSpeciophyllineAntiplasmodial activityPlasmodium falciparum[52]
Isatis indigoticaIsatindigotindolineInhibitory effects on β-amyloid aggregationThioflavin T (ThT)-binding assay[53]
Flavonoids
Centaurea maculosaTrans-flavan-3-ol (+)-catechinAntibacterialXanthomonas campestris, Pseudomonas fluorescens, Erwinia carotovora[54]
Citrus fruitNarirutin, naringin, hesperidin and neohesperidinAntioxidantDPPH assy[55]
Psidium guajavaQuercetin-3-O-α-L-arabinopyranosideanti-Streptococcus mutans activityS. mutans[56]
Rhus retinorrhoeaPersicogeninAnticancerMCF-7, HeLa, and HT-29 cells[57]
Silybum marianumSilibinin A and Silibinin BAnticancerMDA-MB-468 breast cancer cells of the control mice[58]
Leucosceptrum canumS-(+)- and R-(−)-leucoflavoninesAticholinesterase activityAcetylcholinesterase (AChE) inhibitory assay[59]

Table 2.

Pharmacologic effect of stereoisomer compound in plant extract.

4.1 Alkaloids

Based on many studies about unnatural alkaloid enantiomers, and the results reviewed here the pharmacological effect of natural isomers is enantioselective. However, unnatural enantiomers also have a pharmacological effect of their own which can be discovered in the future. Morphinans of the unnatural (+)-series, in contrast to the (−)-series which are chemically connected with natural morphine, were found to be do not have pharmacological effects as analgesics in vivo, instead, presented useful antitussive properties (Figure 10) [62].

Figure 10.

Biosynthesis of morphine in plants. * These metabolic conversions are highly stereoselective [61].

(+)- and (−)-spondomine-racemic and dimeric indole alkaloids have been reported in the study of Tian-Yun Jin (2021) [63], especially, (+)-spondomine displayed cytotoxic against the K562 cell line and exhibited Wnt and HIF1. Moreover, all of them were found to be active in promoted angiogenesis and moderate antiinflammation.

Oleracein E (OE) (8,9-dihydroxy-1,5,6,10b-tetrahydro-2H25 pyrrolo[2,1-a]isoquinoline-3-one), an alkaloid possessing tetrahydroisoquinoline and pyrrolidone skeletons. It was reported to have a lot of pharmacological effects such as: anti-bacterial, anti-inflammatory, anti-aging, anti-hypoxia, anti-oxidant, skeleton-relaxant, hypolipidaemic, analgesic, hypoglycemic, cognition-improvement and neuroprotective functions, especially the optical isomer of (+)-oleracein E (OE) called (−)-trolline has remarkable antibacterial as well as moderate antiviral activity against influenza viruses A and B [64].

4.2 Flavonoids

According to Blair, Lachlan M. (2016) [65], (−)-Foveoglin A (5) exhibited cytotoxicity against a panel of cancer cell lines, while (+)-isofoveoglin (7) and (−)-cyclofoveoglin (8) were weakly cytotoxic, and (+)-foveoglin B (6) was inactive (Figures 11 and 12).

Figure 11.

Aglain and aglaforbesin flavoalkaloids 1–7, 10–12 [65].

Figure 12.

Aglain and aglaforbesin flavoalkaloids 8, 9 [65].

Characterize the stereoselective pharmacokinetics of pinocembrin and pinostrobin and their bioactivity in some in vitro investigation with relevant roles in heart disease, colon cancer, and diabetes etiology and pathophysiology [66]. These investigations have revealed that chiral differences in the chemical structure of these compounds result in significant pharmacodynamic differences. Pinocembrin and pinostrobin demonstrated concentration-dependent alpha amylase inhibitory activity. While pinocembrin also has anti-inflammatory antioxidant in the pure S-enantiomer and racemate.

Racemic liquiritigenin is proved to be a dose-dependent inhibition of alpha-amylase enzyme whereas its pure enantiomers did not have this bioactivity. Racemic liquiritigenin showed moderate antiproliferative activity on an HT-29 cancer cell line that was also dose-dependent and had inhibitory effects on the cyclooxygenase-2 enzyme [67].

Racemic liquiritigenin, which was dose-dependent, has been proved its moderate antiproliferative activity on a cancer cell line_ HT-29, and inhibitory effects on the cyclooxygenase-2 enzyme [67]. The nature type of naringenin, hesperetin and hesperidin is S - enantiomer, but both R and S enantiomers can have biological activities such as: antitumor, antioxidant and anti-inflammatory [68]. The two enantiomers of equol: R-(+)-equol and S-(−)-equol have been researched in antitumor activity which shown a significant decrease in the number of palpable tumors presented in animals feeding R-(+)-equol compared to the S-(−)-equol’s result (Figure 13).

Figure 13.

Chemical structure of daidzein (a) and its metabolites (b and c).

Advertisement

5. Conclusion

Chiral inversion is always mediated by enzymes and varies with solvent, pH and temperature. Considerable attention should be paid to the mechanism of the inversion reaction and its pharmacological and toxicological results. Recent advances in chiral analysis for the herbal plants in clinical research & forensic toxicology by experiments in which one enantiomer was given to the experiment subjects in a specific situation. Demonstration of metabolic chiral inversion demonstration may give an answer for the development of a new pharmaceutical entity. Understanding more about the factors facilitating such interconversions may considerably aid herbal plant development thanks to this feature determination at an early stage and thereby reducing bioanalytical and toxicology workload.

Advertisement

Acknowledgments

The authors would like to express their hearty gratitude to Can Tho University of Medicine and Pharmacy. We also thank all of our colleagues for their excellent assistance.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Khan SJ. Biologically mediated chiral inversion of emerging contaminants. In: Lambropoulou DA, Nollet LML, editors. Transformation Products of Emerging Contaminants in the Environment: Analysis, Processes, Occurrence, Effects and Risks. Chichester, United Kingdom, UK: John Wiley and Sons Ltd; 2014. pp. 261-279
  2. 2. Wsól V, Skálová L, Szotáková B. Chiral inversion of drugs: Coincidence or principle? Current Drug Metabolism. 2004;5(6):517-533
  3. 3. Nguyen NVT, Nguyen KNH, Nguyen KT, Kim KH, Aboul-Enein HY. The impact of chirality on the analysis of alkaloids in plant. Pharmacia. 2021;68(3):643-656
  4. 4. Kumar S, Pandey AK. Chemistry and biological activities of flavonoids: An overview. ScientificWorldJournal. 2013;2013:162750
  5. 5. Yáñez JA, Andrews PK, Davies NM. Methods of analysis and separation of chiral flavonoids. Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences. 2007;848(2):159-181
  6. 6. Caldwell J. Stereochemical determinants of the nature and consequences of drug metabolism. Journal of Chromatography A. 1995;694(1):39-48
  7. 7. Prelog V. Specification of the stereospecificity of some oxido-reductases by diamond lattice sections. Pure and Applied Chemistry. 1964;9(1):119-130
  8. 8. Jenner P, Testa B. The influence of stereochemical factors on drug disposition. Drug Metabolism Reviews. 1973;2(2):117-184
  9. 9. Girvan HM, Munro AW. Applications of microbial cytochrome P450 enzymes in biotechnology and synthetic biology. Current Opinion in Chemical Biology. 2016;31:136-145
  10. 10. Hemscheidt T, Zenk MH. Partial purification and characterization of a NADPH dependent tetrahydroalstonine synthase from Catharanthus roseus cell suspension cultures. Plant Cell Reports. 1985;4(4):216-219
  11. 11. Gebler JC, Poulter CD. Purification and characterization of dimethylallyl tryptophan synthase from Claviceps purpurea. Archives of Biochemistry and Biophysics. 1992;296(1):308-313
  12. 12. Hashimoto T, Yamada Y. Purification and characterization of hyoscyamine 6 beta-hydroxylase from root cultures of Hyoscyamus niger L. hydroxylase and epoxidase activities in the enzyme preparation. European Journal of Biochemistry. 1987;164(2):277-285
  13. 13. Thomas F, Meinhart HZ. Purification and characterization of three isoforms of S-adenosyl-l-methionine: (R,S)-Tetrahydrobenzylisoquinoline-N-methyltransferase from Berberis koetineana cell cultures. Phytochemistry. 1990;29(11):3491-3497
  14. 14. Bednar RA, Hadcock JR. Purification and characterization of chalcone isomerase from soybeans. The Journal of Biological Chemistry. 1988;263(20):9582-9588
  15. 15. Fischer D, Stich K, Britsch L, Grisebach H. Purification and characterization of (+)dihydroflavonol (3-hydroxyflavanone) 4-reductase from flowers of Dahlia variabilis. Archives of Biochemistry and Biophysics. 1988;264(1):40-47
  16. 16. Lukacin R, Wellmann F, Britsch L, Martens S, Matern U. Flavonol synthase from Citrus unshiu is a bifunctional dioxygenase. Phytochemistry. 2003;62(3):287-292
  17. 17. Akashi T, Aoki T, Ayabe S. Identification of a cytochrome P450 cDNA encoding (2S)-flavanone 2-hydroxylase of licorice (Glycyrrhiza echinata L.; Fabaceae) which represents licodione synthase and flavone synthase II. FEBS Letters. 1998;431(2):287-290
  18. 18. Stafford HA, Lester HH. Flavan-3-ol biosynthesis: The conversion of (+)-Dihydromyricetin to its Flavan-3,4-diol (Leucodelphinidin) and to (+)-Gallocatechin by reductases extracted from tissue cultures of Ginkgo biloba and Pseudotsuga menziesii. Plant Physiology. 1985;78(4):791-794
  19. 19. Xie DY, Sharma SB, Dixon RA. Anthocyanidin reductases from Medicago truncatula and Arabidopsis thaliana. Archives of Biochemistry and Biophysics. 2004;422(1):91-102
  20. 20. Paiva NL, Edwards R, Sun YJ, Hrazdina G, Dixon RA. Stress responses in alfalfa (Medicago sativa L.) 11. Molecular cloning and expression of alfalfa isoflavone reductase, a key enzyme of isoflavonoid phytoalexin biosynthesis. Plant Molecular Biology. 1991;17(4):653-667
  21. 21. Preisig CL, Matthews DE, Vanetten HD. Purification and characterization of S-Adenosyl-l-methionine:6a-Hydroxymaackiain 3-O-methyltransferase from Pisum sativum. Plant Physiology. 1989;91(2):559-566
  22. 22. Tanner GJ, Francki KT, Abrahams S, Watson JM, Larkin PJ, Ashton AR. Proanthocyanidin biosynthesis in plants. Purification of legume leucoanthocyanidin reductase and molecular cloning of its cDNA. The Journal of Biological Chemistry. 2003;278(34):31647-31656
  23. 23. Hagel JM, Facchini PJ. Benzylisoquinoline alkaloid metabolism: A century of discovery and a brave new world. Plant & Cell Physiology. 2013;54:647-672
  24. 24. Gulland JMR, R. Constitution of codeine and thebaine. Manchester Literary and Philosophical Society. 1925;69:79-68
  25. 25. Kirby GW. Biosynthesis of the morphine alkaloids. Science. 1967;155:170-173
  26. 26. Farrow SC, Hagel JM, Beaudoin GA, Burns DC, Facchini PJ. Stereochemical inversion of (S)-reticuline by a cytochrome P450 fusion in opium poppy. Nature Chemical Biology. 2015;11(9):728-732
  27. 27. Galanie S, Thodey K, Trenchard IJ, Filsinger Interrante M, Smolke CD. Complete biosynthesis of opioids in yeast. Science. 2015;349(6252):1095-1100
  28. 28. Gesell A, Rolf M, Ziegler J, Díaz Chávez ML, Huang FC, Kutchan TM. CYP719B1 is salutaridine synthase, the C-C phenol-coupling enzyme of morphine biosynthesis in opium poppy. The Journal of Biological Chemistry. 2009;284(36):24432-24442
  29. 29. Stavrinides A, Tatsis E, Caputi L, et al. Structural investigation of heteroyohimbine alkaloid synthesis reveals active site elements that control stereoselectivity. Nature Communications. 2016;7:1-14
  30. 30. Stoeckigt J, Husson HP, Kan-Fan C, Zenk MH. Cathenamine, a central intermediate in the cell free biosynthesis of ajmalicine and related indole alkaloids. Journal of the Chemical Society, Chemical Communications. 1997;164–166
  31. 31. O’Connor SE, Maresh JJ. Chemistry and biology of monoterpene indole alkaloid biosynthesis. Natural Product Reports. 2006;23:532-547
  32. 32. Gerasimenko I, Sheludko Y, Ma X, Stöckigt J. Heterologous expression of a Rauvolfia cDNA encoding strictosidine glucosidase, a biosynthetic key to over 2000 monoterpenoid indole alkaloids. European Journal of Biochemistry. 2002;269:2204-2213
  33. 33. Stavrinides A, Tatsis EC, Foureau E, Caputi L, Kellner F, Courdavault V, et al. Unlocking the diversity of alkaloids in Catharanthus roseus: Nuclear localization suggests metabolic channeling in secondary metabolism. Chemistry & Biology. 2015;22(3):336-341
  34. 34. Koukol J, Conn EE. The metabolism of aromatic compounds in higher plants. The Journal of Biological Chemistry. 1961;136:2692-2698
  35. 35. Jez JM, Bowman ME, Dixon RA, Noel JP. Structure and mechanism of the evolutionarily unique plant enzyme chalcone isomerase. Nature Structural Biology. 2000;7(9):786-791
  36. 36. Prescott AG, Stamford NP, Wheeler G, Firmin JL. In vitro properties of a recombinant flavonol synthase from Arabidopsis thaliana. Phytochemistry. 2002;60(6):589-593
  37. 37. Turnbull JJ, Nakajima J, Welford RW, Yamazaki M, Saito K, Schofield CJ. Mechanistic studies on three 2-oxoglutarate-dependent oxygenases of flavonoid biosynthesis: Anthocyanidin synthase, flavonol synthase, and flavanone 3beta-hydroxylase. The Journal of Biological Chemistry. 2004;279(2):1206-1216
  38. 38. Dixon RA, Xie DY, Sharma SB. Proanthocyanidins--a final frontier in flavonoid research? The New Phytologist. 2005;165(1):9-28
  39. 39. Min T, Kasahara H, Bedgar DL, Youn B, Lawrence PK, Gang DR, et al. Crystal structures of pinoresinol-lariciresinol and phenylcoumaran benzylic ether reductases and their relationship to isoflavone reductases. The Journal of Biological Chemistry. 2003;278(50):50714-50723
  40. 40. Xie DY, Sharma SB, Paiva NL, Ferreira D, Dixon RA. Role of anthocyanidin reductase, encoded by BANYULS in plant flavonoid biosynthesis. Science. 2003;299(5605):396-399
  41. 41. Marais JPJ, Deavours B, Dixon RA, Ferreira D. The stereochemistry of flavonoids. In: Grotewold E, editor. The Science of Flavonoids. New York, NY: Springer; 2006. DOI: 10.1007/978-0-387-28822-2_1
  42. 42. Nguyen NV, Lee KR, Lee YJ, Choi S, Kang JS, Mar W, et al. Chiral high-performance liquid chromatographic separation of evodiamine enantiomers and rutaecarpine, isolated from Evodiae fructus. Journal of Pharmaceutical and Biomedical Analysis. 2013;81-82:151-159
  43. 43. Rajender K, Jürgen M, Ravi B. Enantiomerization study of atropine and its Semipreparative Enantioseparation along with (1RS,2SR)-(±)-ephedrine on polyacrylamide column using high-performance liquid chromatography. Journal of Liquid Chromatography & Related Technologies. 2015;38(1):111-116
  44. 44. Margolin AL. Enzymes in the synthesis of chiral drugs. Enzyme and Microbial Technology. 1993;15(4):266-280
  45. 45. Pham-Huy C, Villain-Pautet G, Hua H, Chikhi-Chorfi N, Galons H, Thevenin M, et al. Separation of oxazepam, lorazepam, and temazepam enantiomers by HPLC on a derivatized cyclodextrin-bonded phase: Application to the determination of oxazepam in plasma. Journal of Biochemical and Biophysical Methods. 2002;54(1–3):287-299
  46. 46. Yang SK, Lu XL. Resolution and stability of oxazepam enantiomers. Chirality. 1992;4:443-446
  47. 47. Juri M, Dai-ichiro K, Masatoshi O, Masahiro T, Seiji N, Kazunari A, et al. Biosynthesis-inspired deracemizative production of d -luciferin by combining luciferase and thioesterase. Biochimica et Biophysica Acta (BBA) - General Subjects. 2017;1861(8):2112-2118
  48. 48. Glass AM, Krause ME, Laurence JS, Jackson TA. Controlling the chiral inversion reaction of the Metallopeptide Ni-asparagine-cysteine-cysteine with dioxygen. Inorganic Chemistry. 2012;51(18):10055-10063
  49. 49. Eed J. Factors affecting enzyme activity. Essai. 2012;10(1):19
  50. 50. Jia-Sen L, Yuan-Long Z, Chao-Mei Y, You-Zuo Z, Yan-Yi H, Feng-Wu W, et al. The structures of huperzine a and B, two new alkaloids exhibiting marked anticholinesterase activity. Canadian Journal of Chemistry. 1986;64(4):837-839
  51. 51. Vergura S, Santoro E, Masi M, Evidente A, Scafato P, Superchi S, et al. Absolute configuration assignment to anticancer Amaryllidaceae alkaloid jonquailine. Fitoterapia. 2018;129:78-84
  52. 52. Pierrot D, Sinou V, Bun SS, Parzy D, Taudon N, Rodriguez J, et al. Design and synthesis of simplified speciophylline analogues and β-carbolines as active molecules against plasmodium falciparum. Drug Development Research. 2019;80(1):133-137
  53. 53. Si-Fan L, Bin L, Yu-Fei X, Le Z, Li-Li L, Xiao-Xiao H, et al. Bioactive spiropyrrolizidine oxindole alkaloid enantiomers from Isatis indigotica fortune. Organic & Biomolecular Chemistry. 2018;16(48):9430-9439
  54. 54. Bais HP, Walker TS, Stermitz FR, Hufbauer RA, Vivanco JM. Enantiomeric-dependent phytotoxic and antimicrobial activity of (+/−)-catechin. A rhizosecreted racemic mixture from spotted knapweed. Plant Physiology. 2002;128(4):1173-1179
  55. 55. Zheng H, Zhen XT, Chen Y, Zhu SC, Ye LH, Yang SW, et al. In situ antioxidation-assisted matrix solid-phase dispersion microextraction and discrimination of chiral flavonoids from citrus fruit via ion mobility quadrupole time-of-flight high-resolution mass spectrometry. Food Chemistry. 2021;343:128422
  56. 56. Prabu GR, Gnanamani A, Sadulla S. Guaijaverin -- a plant flavonoid as potential antiplaque agent against Streptococcus mutans. Journal of Applied Microbiology. 2006;101(2):487-495
  57. 57. Saquib Q, Ahmed S, Ahmad MS, Al-Rehaily AJ, Siddiqui MA, Faisal M, et al. Anticancer efficacies of persicogenin and homoeriodictyol isolated from Rhus retinorrhoea. Process Biochemistry. 2020;95:186-196
  58. 58. Jahanafrooz Z, Motamed N, Rinner B, Mokhtarzadeh A, Baradaran B. Silibinin to improve cancer therapeutic, as an apoptotic inducer, autophagy modulator, cell cycle inhibitor, and microRNAs regulator. Life Sciences. 2018;213:236-247
  59. 59. Feng L, Zhang Y, Liu YC, Liu Y, Luo SH, Huang CS, et al. Leucoflavonine, a new bioactive racemic flavoalkaloid from the leaves of Leucosceptrum canum. Bioorganic & Medicinal Chemistry. 2019;27(2):442-446
  60. 60. Nguyen LA, He H, Pham-Huy C. Chiral drugs: An overview. International Journal of Biomedical Sciences. 2006;2(2):85-100
  61. 61. Rainer L, Meinhart HZ. Closure of the oxide bridge in morphine biosynthesis. 1994;35(23):3897-3900
  62. 62. Arnold B. Chapter 3: Biological activity of unnatural alkaloid enantiomers. In: Cordell GA, editor. The Alkaloids: Chemistry and Biology. Vol. 50. Academic Press; 1998. pp. 109-139
  63. 63. Jin T, Li P, Wang C, Tang X, Cheng M, Zong Y, et al. Racemic Bisindole alkaloids: Structure, bioactivity, and computational study. Chinese Journal of Chemistry. 2021;39(9):2588-2598
  64. 64. Wang PP, Sun HX, Liu CJ, Hu MH, He XQ, Yue S, et al. Racemic oleracein E increases the survival rate and attenuates memory impairment in D-galactose/NaNO₂-induced senescent mice. Phytomedicine. 2016;23(5):460-467
  65. 65. Blair LM, Calvert MB, Sperry J. Flavoalkaloids-isolation, biological activity, and Total synthesis. The Alkaloids. Chemistry and Biology. 2017;77:85-115
  66. 66. Sayre CL, Alrushaid S, Martinez SE, Anderson HD, Davies NM. Pre-clinical pharmacokinetic and Pharmacodynamic characterization of selected chiral flavonoids: Pinocembrin and Pinostrobin. Journal of Pharmacy & Pharmaceutical Sciences. 2015;18(4):368-395
  67. 67. Alrushaid S, Davies NM, Martinez SE, Sayre CL. Pharmacological characterization of liquiritigenin, a chiral flavonoid in licorice. Research in Pharmaceutical Sciences. 2016;11(5):355-365
  68. 68. Pinto C, Cidade H, Pinto M, Tiritan ME. Chiral flavonoids as antitumor agents. Pharmaceuticals. 2021;14(12):1267

Written By

Ngoc-Van Thi Nguyen

Submitted: 30 December 2021 Reviewed: 07 January 2022 Published: 12 February 2022