Search
2024 Volume 4
Article Contents
REVIEW   Open Access    

Lotus ( Nelumbo nucifera) benzylisoquinoline alkaloids: advances in chemical profiling, extraction methods, pharmacological activities, and biosynthetic elucidation

More Information
  • As a member of the only two species in the Nelumbonaceae family, lotus ( Nelumbo nucifera) accumulates abundant benzylisoquinoline alkaloids (BIAs) in almost all of its tissues. Evidence from both traditional and modern medicine suggest great potential of the lotus BIAs in developing novel drugs for the prevention and treatment of diverse life-threatening diseases. This review provides a comprehensive summary on the up-to-date advances in the chemical profiling, extraction methods, pharmacological activities, and biosynthesis of lotus BIAs. Currently, a total of 59 BIAs structurally belonging to the 1-benzylisoquinoline, aporphine, and bis-BIA categories have been identified in various lotus tissues, with their predominant accumulation in the leaf and plumule. In contrast to the common S-conformers in Ranunculales, most lotus BIAs are R-conformers. Solvent extraction is still the most widely used BIA extraction method in lotus at the industrial level, however, numerous greener and highly advanced extraction techniques have also been developed. High-performance liquid chromatography (HPLC) followed by electrospray ionization (ESI) and tandem mass spectrometry (MS/MS) techniques are currently the most commonly used methods for separation, quantification, and characterization of lotus BIAs. Moreover, the pharmacological activities of major BIAs isolated from lotus leaves and plumules are discussed, and their biosynthetic pathways proposed based on recent functional characterization studies of lotus BIA biosynthetic genes. Finally, a summary discussion is provided on the future research trajectories in elucidating lotus BIA biosynthesis, storage, and transportation, as well as their potential application in clinical drug development.
  • 加载中
  • [1]

    Li Y, Smith T, Svetlana P, Yang J, Jin J, et al. 2014. Paleobiogeography of the lotus plant (Nelumbonaceae: Nelumbo) and its bearing on the paleoclimatic changes. Palaeogeography, Palaeoclimatology, Palaeoecology 399:284−93

    doi: 10.1016/j.palaeo.2014.01.022

    CrossRef   Google Scholar

    [2]

    Zheng P, Sun H, Liu J, Lin J, Zhang X, et al. 2022. Comparative analyses of American and Asian lotus genomes reveal insights into petal color, carpel thermogenesis and domestication. The Plant Journal 110:1498−515

    doi: 10.1111/tpj.15753

    CrossRef   Google Scholar

    [3]

    Lin Z, Zhang C, Cao D, Damaris RN, Yang P. 2019. The latest studies on lotus ( Nelumbo nucifera)-an emerging horticultural model plant. International Journal of Molecular Sciences 20:3680

    doi: 10.3390/ijms20153680

    CrossRef   Google Scholar

    [4]

    Shen-Miller J. 2002. Sacred lotus, the long-living fruits of China Antique. Seed Science Research 12:131−43

    doi: 10.1079/ssr2002112

    CrossRef   Google Scholar

    [5]

    Qi H, Yu F, Deng J, Yang P. 2022. Studies on lotus genomics and the contribution to its breeding. International Journal of Molecular Sciences 23:7270

    doi: 10.3390/ijms23137270

    CrossRef   Google Scholar

    [6]

    Yang M, Zhu L, Pan C, Xu L, Liu Y, et al. 2015. Transcriptomic analysis of the regulation of rhizome formation in temperate and tropical lotus ( Nelumbo nucifera). Scientific Reports 5:13059

    doi: 10.1038/srep13059

    CrossRef   Google Scholar

    [7]

    Wang Z, Li Y, Ma D, Zeng M, Wang Z, et al. 2023. Alkaloids from lotus ( Nelumbo nucifera): recent advances in biosynthesis, pharmacokinetics, bioactivity, safety, and industrial applications. Critical Reviews in Food Science and Nutrition 63:4867−900

    doi: 10.1080/10408398.2021.2009436

    CrossRef   Google Scholar

    [8]

    Chen G, Zhu M, Guo M. 2019. Research advances in traditional and modern use of Nelumbo nucifera: phytochemicals, health promoting activities and beyond. Critical Reviews in Food Science and Nutrition 59:S189−S209

    doi: 10.1080/10408398.2018.1553846

    CrossRef   Google Scholar

    [9]

    Mukherjee PK, Mukherjee D, Maji AK, Rai S, Heinrich M. 2009. The sacred lotus ( Nelumbo nucifera) – phytochemical and therapeutic profile. Journal of Pharmacy and Pharmacology 61:407−22

    doi: 10.1211/jpp.61.04.0001

    CrossRef   Google Scholar

    [10]

    Itoh A, Saitoh T, Tani K, Uchigaki M, Sugimoto Y, et al. 2011. Bisbenzylisoquinoline alkaloids from Nelumbo nucifera. Chemical & Pharmaceutical Bulletin 59:947−51

    doi: 10.1248/cpb.59.947

    CrossRef   Google Scholar

    [11]

    Deng X, Zhu L, Fang T, Vimolmangkang S, Yang D, et al. 2016. Analysis of isoquinoline alkaloid composition and wound-induced variation in Nelumbo using HPLC-MS/MS. Journal of Agricultural and Food Chemistry 64:1130−36

    doi: 10.1021/acs.jafc.5b06099

    CrossRef   Google Scholar

    [12]

    Pyne ME, Gold ND, Martin VJJ. 2023. Pathway elucidation and microbial synthesis of proaporphine and bis-benzylisoquinoline alkaloids from sacred lotus ( Nelumbo nucifera). Metabolic Engineering 77:162−73

    doi: 10.1016/j.ymben.2023.03.010

    CrossRef   Google Scholar

    [13]

    Liscombe DK, Facchini PJ. 2008. Evolutionary and cellular webs in benzylisoquinoline alkaloid biosynthesis. Current Opinion in Biotechnology 19:173−80

    doi: 10.1016/j.copbio.2008.02.012

    CrossRef   Google Scholar

    [14]

    Khan AY, Suresh Kumar G. 2015. Natural isoquinoline alkaloids: binding aspects to functional proteins, serum albumins, hemoglobin, and lysozyme. Biophysical Review 7:407−20

    doi: 10.1007/s12551-015-0183-5

    CrossRef   Google Scholar

    [15]

    Facchini PJ. 2001. Alkaloid biosynthesis in plants: biochemistry, cell biology, molecular regulation, and metabolic engineering applications. Annual Review of Plant Physiology and Plant Molecular Biology 52:29−66

    doi: 10.1146/annurev.arplant.52.1.29

    CrossRef   Google Scholar

    [16]

    Kunitomo J, Yoshikaw Y, Tanaka S, Imori Y, Isoi K, et al. 1973. Alkaloids of Nelumbo nucifera. Phytochemistry 12:699−701

    doi: 10.1016/S0031-9422(00)84467-2

    CrossRef   Google Scholar

    [17]

    Sharma BR, Gautam LNS, Adhikari D, Karki R. 2017. A comprehensive review on chemical profiling of Nelumbo Nucifera: potential for drug development. Phytotherapy Research 31:3−26

    doi: 10.1002/ptr.5732

    CrossRef   Google Scholar

    [18]

    Menéndez-Perdomo IM, Facchini PJ. 2018. Benzylisoquinoline alkaloids biosynthesis in sacred lotus. Molecules 23:2899

    doi: 10.3390/molecules23112899

    CrossRef   Google Scholar

    [19]

    Nakamura S, Nakashima S, Tanabe G, Oda Y, Yokota N, et al. 2013. Alkaloid constituents from flower buds and leaves of sacred lotus ( Nelumbo nucifera, Nymphaeaceae) with melanogenesis inhibitory activity in B16 melanoma cells. Bioorganic & Medicinal Chemistry 21:779−87

    doi: 10.1016/j.bmc.2012.11.038

    CrossRef   Google Scholar

    [20]

    Do TCMV, Nguyen TD, Tran H, Stuppner H, Ganzera M. 2013. Analysis of alkaloids in Lotus ( Nelumbo nucifera Gaertn.) leaves by non-aqueous capillary electrophoresis using ultraviolet and mass spectrometric detection. Journal of Chromatography 1302:174−80

    doi: 10.1016/j.chroma.2013.06.002

    CrossRef   Google Scholar

    [21]

    Liu C, Tsai W, Shen C, Lin Y, Liao J, et al. 2006. Inhibition of (S)-armepavine from Nelumbo nucifera on autoimmune disease of MRL/MpJ- lpr/lpr mice. European Journal of Pharmacology 531:270−79

    doi: 10.1016/j.ejphar.2005.11.062

    CrossRef   Google Scholar

    [22]

    Deng X, Zhao L, Fang T, Xiong Y, Ogutu C, et al. 2018. Investigation of benzylisoquinoline alkaloid biosynthetic pathway and its transcriptional regulation in lotus. Horticulture Research 5:29

    doi: 10.1038/s41438-018-0035-0

    CrossRef   Google Scholar

    [23]

    Chen S, Zhang H, Liu Y, Fang J, Li S. 2013. Determination of lotus leaf alkaloids by solid phase extraction combined with high performance liquid chromatography with diode array and tandem mass spectrometry detection. Analytical Letters 46:2846−59

    doi: 10.1080/00032719.2013.816960

    CrossRef   Google Scholar

    [24]

    Sun H, Song H, Deng X, Liu J, Yang D, et al. 2022. Transcriptome-wide characterization of alkaloids and chlorophyll biosynthesis in lotus plumule. Frontiers in Plant Science 13:885503

    doi: 10.3389/fpls.2022.885503

    CrossRef   Google Scholar

    [25]

    Furukawa H. 1966. Studies on the alkaloids of Nelumbo nucifera Gaertn. NMR spectra of liensinine type alkaloids. Yakugaku Zasshi 86:883−86

    doi: 10.1248/yakushi1947.86.10_883

    CrossRef   Google Scholar

    [26]

    Kashiwada Y, Aoshima A, Ikeshiro Y, Chen Y, Furukawa H, et al. 2005. Anti-HIV benzylisoquinoline alkaloids and flavonoids from the leaves of Nelumbo nucifera, and structure-activity correlations with related alkaloids. Bioorganic & Medicinal Chemistry 13:443−48

    doi: 10.1016/j.bmc.2004.10.020

    CrossRef   Google Scholar

    [27]

    Zelenski SG. 1977. Alkaloids of Nelumbo lutea (Willd.) pers. (Nymphaeaceae). Journal of Pharmaceutical Sciences 66:1627−28

    doi: 10.1002/jps.2600661132

    CrossRef   Google Scholar

    [28]

    Chen J, Gao K, Liu T, Zhao H, Wang J, et al. 2013. Aporphine alkaloids: a kind of alkaloids' extract source, chemical constitution and pharmacological actions in different botany: a review. Asian Journal of Chemistry 25:10015−27

    doi: 10.14233/ajchem.2013.15890

    CrossRef   Google Scholar

    [29]

    Yang M, Zhu L, Li L, Li J, Xu L, et al. 2017. Digital gene expression analysis provides insight into the transcript profile of the genes involved in aporphine alkaloid biosynthesis in lotus ( Nelumbo nucifera). Frontiers in Plant Science 8:80

    doi: 10.3389/fpls.2017.00080

    CrossRef   Google Scholar

    [30]

    Tian W, Zhi H, Yang C, Wang L, Long J, et al. 2018. Chemical composition of alkaloids of Plumula nelumbinis and their antioxidant activity from different habitats in China. Industrial Crops and Products 125:537−48

    doi: 10.1016/j.indcrop.2018.09.045

    CrossRef   Google Scholar

    [31]

    Zhou M, Jiang M, Ying X, Cui Q, Han Y, et al. 2013. Identification and comparison of anti-inflammatory ingredients from different organs of Lotus nelumbo by UPLC/Q-TOF and PCA coupled with a NF-κB reporter gene assay. PLoS ONE 8:e81971

    doi: 10.1371/journal.pone.0081971

    CrossRef   Google Scholar

    [32]

    Chen S, Guo W, Qi X, Zhou J, Liu Z, et al. 2019. Natural alkaloids from lotus plumule ameliorate lipopolysaccharide-induced depression-like behavior: integrating network pharmacology and molecular mechanism evaluation. Food & Function 10:6062−73

    doi: 10.1039/c9fo01092k

    CrossRef   Google Scholar

    [33]

    Kato E, Inagaki Y, Kawabata J. 2015. Higenamine 4′-O-β-d-glucoside in the lotus plumule induces glucose uptake of L6 cells through β2-adrenergic receptor. Bioorganic & Medicinal Chemistry 23:3317−21

    doi: 10.1016/j.bmc.2015.04.054

    CrossRef   Google Scholar

    [34]

    Tomita M, Furukawa H. 1962. On the alkaloids of Nelumbo nucifera Gaertn. V. Alkaloids of "Ohga-hasu". Yakugaku Zasshi 82:1458−60

    Google Scholar

    [35]

    Fang Y, Li Q, Shao Q, Wang B, Wei Y. 2017. A general ionic liquid pH-zone-refining countercurrent chromatography method for separation of alkaloids from Nelumbo nucifera Gaertn. Journal of Chromatography A 1507:63−71

    doi: 10.1016/j.chroma.2017.05.048

    CrossRef   Google Scholar

    [36]

    Grienke U, Mair CE, Saxena P, Baburin I, Scheel O, et al. 2015. Human ether-à-go-go related gene (hERG) channel blocking aporphine alkaloids from lotus leaves and their quantitative analysis in dietary weight loss supplements. Journal of Agriculture and Food Chemistry 63:5634−39

    doi: 10.1021/acs.jafc.5b01901

    CrossRef   Google Scholar

    [37]

    Shoji N, Umeyama A, Saito N, Luchi A, Takemoto T, et al. 1987. Asimilobine and lirinidine, serotonergic receptor antagonists, from Nelumbo nucifera. Journal of Natural Products 50:773−74

    doi: 10.1021/np50052a044

    CrossRef   Google Scholar

    [38]

    Liu C, Kao C, Wu H, Li W, Huang C, et al. 2014. Antioxidant and anticancer aporphine alkaloids from the leaves of Nelumbo nucifera Gaertn. cv. Rosa-plena. Molecules 19:17829−38

    doi: 10.3390/molecules191117829

    CrossRef   Google Scholar

    [39]

    Liu S, Lu T, Su C, Lay I, Lin H, et al. 2014. Lotus leaf ( Nelumbo nucifera) and its active constituents prevent inflammatory responses in macrophages via JNK/NF-κB signaling pathway. The American Journal of Chinese Medicine 42:869−89

    doi: 10.1142/S0192415X14500554

    CrossRef   Google Scholar

    [40]

    Li S, Wu J, Chen L, Du H, Xu Y, et al. 2014. Biogenesis of C-glycosyl flavones and profiling of flavonoid glycosides in lotus ( Nelumbo nucifera). PLoS ONE 9:e108860

    doi: 10.1371/journal.pone.0108860

    CrossRef   Google Scholar

    [41]

    Ye L, He X, Kong L, Liao Y, Pan R, et al. 2014. Identification and characterization of potent CYP2D6 inhibitors in lotus leaves. Journal of Ethnopharmacology 153:190−96

    doi: 10.1016/j.jep.2014.02.014

    CrossRef   Google Scholar

    [42]

    Chang C, Ou T, Yang M, Huang C, Wang C. 2016. Nelumbo nucifera Gaertn leaves extract inhibits the angiogenesis and metastasis of breast cancer cells by downregulation connective tissue growth factor (CTGF) mediated PI3K/AKT/ERK signaling. Journal of Ethnopharmacology 188:111−22

    doi: 10.1016/j.jep.2016.05.012

    CrossRef   Google Scholar

    [43]

    Wei F, Gou X, Xu X, Wang S, Bao T. 2021. Sensitive quantification of liensinine alkaloid using a HPLC-MS/MS method and its application in microvolume rat plasma. Journal of Analytical Methods in Chemistry 2021:6629579

    doi: 10.1155/2021/6629579

    CrossRef   Google Scholar

    [44]

    Yang G, Sun J, Pan Y, Zhang J, Xiao M, et al. 2018. Isolation and identification of a tribenzylisoquinoline alkaloid from Nelumbo nucifera Gaertn, a novel potential smooth muscle relaxant. Fitoterapia 124:58−65

    doi: 10.1016/j.fitote.2017.10.020

    CrossRef   Google Scholar

    [45]

    Petruczynik A. 2012. Analysis of alkaloids from different chemical groups by different liquid chromatography methods. Central European Journal of Chemistry 10:802−35

    doi: 10.2478/s11532-012-0037-y

    CrossRef   Google Scholar

    [46]

    Choi YH, Chin Y, Kim J, Jeon SH, Yoo K. 1999. Strateges for supercritical fluid extraction of hyoscyamine and scopolamine salts using basified modifiers. Journal of Chromatography A 863:47−55

    doi: 10.1016/S0021-9673(99)00962-0

    CrossRef   Google Scholar

    [47]

    Zhang Q, Lin L, Ye W. 2018. Techniques for extraction and isolation of natural products: a comprehensive review. Chinese Medicine 13:20

    Google Scholar

    [48]

    Li P, Xu G, Li S, Wang Y, Fan T, et al. 2008. Optimizing ultra performance liquid chromatographic analysis of 10 diterpenoid compounds in Salvia miltiorrhiza using central composite design. Journal of Agriculture and Food Chemistry 56:1164−71

    doi: 10.1021/jf073020u

    CrossRef   Google Scholar

    [49]

    Zhou Y, Zhang Q, Li S, Yin Z, Zhang X, et al. 2012. Quality evaluation of semen oroxyli through simultaneous quantification of 13 components by high performance liquid chromatorgraphy. Current Pharmaceutical Analysis 8:206−13

    doi: 10.2174/1573412911208020206

    CrossRef   Google Scholar

    [50]

    Morikawa T, Kitagawa N, Tanabe G, Ninomiya K, Okugawa S, et al. 2016. Quantitative determination of alkaloids in lotus flower (flower buds of Nelumbo nucifera) and their melanogenesis inhibitory activity. Molecules 21:930

    doi: 10.3390/molecules21070930

    CrossRef   Google Scholar

    [51]

    Xiao J, Tian B, Xie B, Yang E, Shi J, et al. 2010. Supercritical fluid extraction and identification of isoquinoline alkaloids from leaves of Nelumbo nucifera Gaertn. European Food Research and Technology 231:407−14

    doi: 10.1007/s00217-010-1290-y

    CrossRef   Google Scholar

    [52]

    Herrero M, Mendiola JA, Cifuentes A, Ibáñez E. 2010. Supercritical fluid extraction: recent advances and applications. Journal of Chromatography A 1217:2495−511

    doi: 10.1016/j.chroma.2009.12.019

    CrossRef   Google Scholar

    [53]

    Brachet A, Christen P, Gauvrit JY, Loneray R, Lantéri P, et al. 2000. Experimental design in supercritical fluid extraction of cocaine from coca leaves. Journal of Biochemical and Biophysical Methods 43:353−66

    doi: 10.1016/S0165-022X(00)00062-2

    CrossRef   Google Scholar

    [54]

    Radcliffe C, Maguire K, Lockwood B. 2000. Applications of supercritical fluid extraction and chromatography in forensic science. Journal of Biochemical and Biophysical Methods 43:261−72

    doi: 10.1016/S0165-022X(00)00058-0

    CrossRef   Google Scholar

    [55]

    Ito Y. 2005. Golden rules and pitfalls in selecting optimum conditions for high-speed counter-current chromatography. Journal of Chromatography A 1065:145−68

    doi: 10.1016/j.chroma.2004.12.044

    CrossRef   Google Scholar

    [56]

    Ma C, Wang J, Chu H, Zhang X, Wang Z, et al. 2014. Purification and characterization of aporphine alkaloids from leaves of Nelumbo nucifera Gaertn and their effects on glucose consumption in 3T3-L1 adipocytes. International Journal of Molecular Sciences 15:3481−94

    doi: 10.3390/ijms15033481

    CrossRef   Google Scholar

    [57]

    Zheng Z, Wang M, Wang D, Duan W, Wang X, et al. 2010. Preparative separation of alkaloids from Nelumbo nucifera leaves by conventional and pH-zone-refining counter-current chromatography. Journal of Chromatography B 878:1647−51

    doi: 10.1016/j.jchromb.2010.04.020

    CrossRef   Google Scholar

    [58]

    Liu Z, Zhao Y, Peng H, Luo T, Liu Y. 2022. Countercurrent chromatography: separation, principle, mechanical design, development trends, and applications. Journal of Liquid Chromatography & Related Technologies 45:51−65

    doi: 10.1080/10826076.2022.2117190

    CrossRef   Google Scholar

    [59]

    Wu S, Sun C, Cao X, Zhou H, Zhang H, et al. 2004. Preparative counter-current chromatography isolation of liensinine and its analogues from embryo of the seed of Nelumbo nucifera GAERTN. using upright coil planet centrifuge with four multilayer coils connected in series. Journal of Chromatography A 1041:153−62

    doi: 10.1016/j.chroma.2004.05.003

    CrossRef   Google Scholar

    [60]

    Liu S, Wang B, Li X, Qi L, Liang Y. 2009. Preparative separation and purification of liensinine, isoliensinine and neferine from seed embryo of Nelumbo nucifera GAERTN using high-speed counter-current chromatography. Journal of Separation Science 32:2476−81

    doi: 10.1002/jssc.200800766

    CrossRef   Google Scholar

    [61]

    Wang X, Liu J, Geng Y, Wang D, Dong H, et al. 2010. Preparative separation of alkaloids from Nelumbo nucifera Gaertn by pH-zone-refining counter-current chromatography. Journal of Separation Science 33:539−44

    doi: 10.1002/jssc.200900561

    CrossRef   Google Scholar

    [62]

    Duanmu Q, Li A, Sun A, Liu R, Li X. 2010. Semi-preparative high-speed counter-current chromatography separation of alkaloids from embryo of the seed of Nelumbo nucifera Gaertn by pH-gradient elution. Journal of Separation Science 33:1746−51

    doi: 10.1002/jssc.200900872

    CrossRef   Google Scholar

    [63]

    Chen Y, Fan G, Wu H, Wu Y, Mitchell A. 2007. Separation, identification and rapid determination of liensine, isoliensinine and neferine from embryo of the seed of Nelumbo nucifera Gaertn. by liquid chromatography coupled to diode array detector and tandem mass spectrometry. Journal of Pharmaceutical and Biomedical Analysis 43:99−104

    doi: 10.1016/j.jpba.2006.06.016

    CrossRef   Google Scholar

    [64]

    Luo X, Chen B, Liu J, Yao S. 2005. Simultaneous analysis of N-nornuciferine, O-nornuciferine, nuciferine, and roemerine in leaves of Nelumbo nucifera Gaertn by high-performance liquid chromatography–photodiode array detection–electrospray mass spectrometry. Analytica Chimica Acta 538:129−33

    doi: 10.1016/j.aca.2005.01.066

    CrossRef   Google Scholar

    [65]

    Lai H, Ouyang Y, Tian G, Zhao J, Zhang J, et al. 2022. Rapid characterization and identification of the chemical constituents and the metabolites of Du-zhi pill using UHPLC coupled with quadrupole time-of-flight mass spectrometry. Journal of Chromatography B 1209:123433

    doi: 10.1016/j.jchromb.2022.123433

    CrossRef   Google Scholar

    [66]

    Liang C, Zhang X, Diao X, Liao M, Sun Y, et al. 2018. Metabolism profiling of nevadensin in vitro and in vivo by UHPLC-Q-TOF-MS/MS. Journal of Chromatography B 1084:69−79

    doi: 10.1016/j.jchromb.2018.03.032

    CrossRef   Google Scholar

    [67]

    Guo Y, Chen X, Qi J, Yu B. 2016. Simultaneous qualitative and quantitative analysis of flavonoids and alkaloids from the leaves of Nelumbo nucifera Gaertn. using high-performance liquid chromatography with quadrupole time-of-flight mass spectrometry. Journal of Separation Science 39:2499−507

    doi: 10.1002/jssc.201501315

    CrossRef   Google Scholar

    [68]

    Khare CP. 2004. Indian herbal remedies. rational western therapy, ayurvedic and other traditional usage, botany. Heidelberg: Springer Berlin. ix, 524 pp. https://doi.org/10.1007/978-3-642-18659-2

    [69]

    Liu C, Tsai W, Lin Y, Liao J, Chen C, et al. 2004. The extracts from Nelumbo Nucifera suppress cell cycle progression, cytokine genes expression, and cell proliferation in human peripheral blood mononuclear cells. Life Science 75:699−716

    doi: 10.1016/j.lfs.2004.01.019

    CrossRef   Google Scholar

    [70]

    Guo F, Yang X, Li X, Feng R, Guan C, et al. 2013. Nuciferine prevents hepatic steatosis and injury induced by a high-fat diet in hamsters. PLoS ONE 8:e63770

    doi: 10.1371/journal.pone.0063770

    CrossRef   Google Scholar

    [71]

    Wu Y, Tan F, Zhang T, Xie B, Ran L, et al. 2020. The anti-obesity effect of lotus leaves on high-fat-diet-induced obesity by modulating lipid metabolism in C57BL/6J mice. Applied Biological Chemistry 63:61

    doi: 10.1186/s13765-020-00541-x

    CrossRef   Google Scholar

    [72]

    He Y, Tao Y, Qiu L, Xu W, Huang X, et al. 2022. Lotus ( Nelumbo nucifera Gaertn.) leaf-fermentation supernatant inhibits adipogenesis in 3T3-L1 preadipocytes and suppresses obesity in high-fat diet-induced obese rats. Nutrients 14:4348

    doi: 10.3390/nu14204348

    CrossRef   Google Scholar

    [73]

    Pearson ER. 2019. Type 2 diabetes: a multifaceted disease. Diabetologia 62:1107−12

    doi: 10.1007/s00125-019-4909-y

    CrossRef   Google Scholar

    [74]

    Nguyen HK, Ta NT, Minh H, Pham T, Nguyen TQ, et al. 2012. Nuciferine extracted from sacred lotus stimulates insulin secretion in vitro better than glyburide. Canadian Journal of Diabetes 36:S64

    doi: 10.1016/j.jcjd.2012.07.470

    CrossRef   Google Scholar

    [75]

    Zhang X, Zhang S, Liu X, Wang Y, Chang J, et al. 2018. Oxidation resistance 1 is a novel senolytic target. Aging Cell 17:e12780

    doi: 10.1111/acel.12780

    CrossRef   Google Scholar

    [76]

    Hwang D, Charchoghlyan H, Lee JS, Kim M. 2015. Bioactive compounds and antioxidant activities of the Korean lotus leaf ( Nelumbo nucifera) condiment: volatile and nonvolatile metabolite profiling during fermentation. International Journal of Food Science & Technology 50:1988−95

    doi: 10.1111/ijfs.12882

    CrossRef   Google Scholar

    [77]

    Guo C, Zhang N, Liu C, Xue J, Chu J, et al. 2020. Qualities and antioxidant activities of lotus leaf affected by different drying methods. Acta Physiologiae Plantarum 42:14

    doi: 10.1007/s11738-019-2992-9

    CrossRef   Google Scholar

    [78]

    Golkar Z, Bagasra O, Pace DG. 2014. Bacteriophage therapy: a potential solution for the antibiotic resistance crisis. Journal of Infection in Developing Countries 8:129−36

    doi: 10.3855/jidc.3573

    CrossRef   Google Scholar

    [79]

    Li M, Xu Z. 2007. The inhibition of dentifrice containing the lotus leaf-derived inhibitor on periodontitis-related bacteria in vitro. International Dental Journal 57:303−6

    doi: 10.1111/j.1875-595X.2007.tb00137.x

    CrossRef   Google Scholar

    [80]

    Liu W, Yi D, Guo J, Xiang Z, Deng L, et al. 2015. Nuciferine, extracted from Nelumbo nucifera Gaertn, inhibits tumor-promoting effect of nicotine involving Wnt/β-catenin signaling in non-small cell lung cancer. Journal of Ethnopharmacology 165:83−93

    doi: 10.1016/j.jep.2015.02.015

    CrossRef   Google Scholar

    [81]

    Kang EJ, Lee SK, Park KK, Son SH, Kim KR, et al. 2017. Liensinine and nuciferine, bioactive components of Nelumbo nucifera, inhibit the growth of breast cancer cells and breast cancer-associated bone loss. Evidence-Based Complementary and Alternative Medicine 2017:1583185

    doi: 10.1155/2017/1583185

    CrossRef   Google Scholar

    [82]

    Xiao M, Xian C, Wang Y, Qi X, Zhang R, et al. 2023. Nuciferine attenuates atherosclerosis by regulating the proliferation and migration of VSMCs through the Calm4/MMP12/AKT pathway in ApoE (-/-) mice fed with High-Fat-Diet. Phytomedicine 108:154536

    doi: 10.1016/j.phymed.2022.154536

    CrossRef   Google Scholar

    [83]

    Hoesel B, Schmid JA. 2013. The complexity of NF-κB signaling in inflammation and cancer. Molecular Cancer 12:86

    Google Scholar

    [84]

    Chen Q, Jin M, Yang F, Zhu J, Xiao Q, et al. 2013. Matrix metalloproteinases: inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediators of Inflammation 2013:928315

    doi: 10.1155/2013/928315

    CrossRef   Google Scholar

    [85]

    Rai S, Wahile A, Mukherjee K, Saha BP, Mukherjee PK. 2006. Antioxidant activity of Nelumbo nucifera (sacred lotus) seeds. Journal of Ethnopharmacology 104:322−27

    doi: 10.1016/j.jep.2005.09.025

    CrossRef   Google Scholar

    [86]

    Jung HA, Jin SE, Choi RJ, Kim DH, Kim YS, et al. 2010. Anti-amnesic activity of neferine with antioxidant and anti-inflammatory capacities, as well as inhibition of ChEs and BACE1. Life Sciences 87:420−30

    doi: 10.1016/j.lfs.2010.08.005

    CrossRef   Google Scholar

    [87]

    Li G, Zhu G, Gao Y, Xiao W, Xu H, et al. 2013. Neferine inhibits the upregulation of CCL5 and CCR5 in vascular endothelial cells during chronic high glucose treatment. Inflammation 36:300−8

    doi: 10.1007/s10753-012-9547-1

    CrossRef   Google Scholar

    [88]

    Xie Y, Zhang Y, Zhang L, Zeng S, Guo Z, et al. 2013. Protective effects of alkaloid compounds from Nelumbinis Plumula on tert-butyl hydroperoxide-induced oxidative stress. Molecules 18:10285−300

    doi: 10.3390/molecules180910285

    CrossRef   Google Scholar

    [89]

    Chiu K, Hung Y, Wang S, Tsai Y, Wu N, et al. 2021. Anti-allergic and anti-inflammatory effects of neferine on RBL-2H3 cells. International Journal of Molecular Sciences 22:10994

    doi: 10.3390/ijms222010994

    CrossRef   Google Scholar

    [90]

    Deng G, Wang L, He W, Zhou W, Chen B, et al. 2018. Anti-inflammatory effects of neferine on LPS-induced human endothelium via MAPK, and NF-κβ pathways. Pharmazie 73:541−44

    doi: 10.1691/ph.2018.8443

    CrossRef   Google Scholar

    [91]

    Wu X, Guo Y, Min X, Pei L, Chen X. 2018. Neferine, a bisbenzylisoquinoline alkaloid, ameliorates dextran sulfate sodium-induced ulcerative colitis. The American Journal of Chinese Medicine 46:1263−79

    doi: 10.1142/S0192415X18500660

    CrossRef   Google Scholar

    [92]

    Chen X, Zhao H, Kang X, Tai X, Guo W, et al. 2023. Liensinine, an isoquinoline-type alkaloid in lotus seeds, suppressed TGF-beta1-induced proliferation and migration in human tenon capsule fibroblast cells through MAP3K7 gene. Revista Brasileira De Farmacognosia 33:128−36

    doi: 10.1007/s43450-022-00334-x

    CrossRef   Google Scholar

    [93]

    Shen Y, Bian R, Li Y, Gao Y, Liu Y, et al. 2019. Liensinine induces gallbladder cancer apoptosis and G2/M arrest by inhibiting ZFX-induced PI3K/AKT pathway. Acta Biochimica et Biophysica Sinica 51:606−13

    doi: 10.1093/abbs/gmz041

    CrossRef   Google Scholar

    [94]

    Yang J, Yu K, Si X, Li S, Cao Y, et al. 2019. Liensinine inhibited gastric cancer cell growth through ROS generation and the PI3K/AKT pathway. Journal of Cancer 10:6431−38

    doi: 10.7150/jca.32691

    CrossRef   Google Scholar

    [95]

    Zhang X, Liu Z, Xu B, Sun Z, Gong Y, et al. 2012. Neferine, an alkaloid ingredient in lotus seed embryo, inhibits proliferation of human osteosarcoma cells by promoting p38 MAPK-mediated p21 stabilization. European Journal of Pharmacology 677:47−54

    doi: 10.1016/j.ejphar.2011.12.035

    CrossRef   Google Scholar

    [96]

    Xiao J, Zhang Y, Feng X, Wang J, Qian J. 2006. Effects of isoliensinine on angiotensin II-induced proliferation of porcine coronary arterial smooth muscle cells. Journal of Asian Natural Products Research 8:209−16

    doi: 10.1080/1028602042000325609

    CrossRef   Google Scholar

    [97]

    Shu G, Yue L, Zhao W, Xu C, Yang J, et al. 2015. Isoliensinine, a bioactive alkaloid derived from embryos of Nelumbo nucifera, induces hepatocellular carcinoma cell apoptosis through suppression of NF-κB signaling. Journal of Agriculture and Food Chemistry 63:8793−803

    doi: 10.1021/acs.jafc.5b02993

    CrossRef   Google Scholar

    [98]

    Xiao X, Luo F, Fu M, Jiang Y, Liu S, et al. 2022. Evaluating the therapeutic role of selected active compounds in Plumula Nelumbinis on pulmonary hypertension via network pharmacology and experimental analysis. Frontiers in Pharmacology 13:977921

    doi: 10.3389/fphar.2022.977921

    CrossRef   Google Scholar

    [99]

    Wicha P, Onsa-ard A, Chaichompoo W, Suksamrarn A, Tocharus C. 2020. Vasorelaxant and antihypertensive effects of neferine in rats: an in vitro and in vivo study. Planta Medica 86:496−504

    doi: 10.1055/a-1123-7852

    CrossRef   Google Scholar

    [100]

    Liu Z, Hu L, Zhang Z, Song L, Zhang P, et al. 2021. Isoliensinine eliminates afterdepolarizations through inhibiting late sodium current and L-type calcium current. Cardiovascular Toxicology 21:67−78

    doi: 10.1007/s12012-020-09597-z

    CrossRef   Google Scholar

    [101]

    Poornima P, Weng CF, Padma VV. 2014. Neferine, an alkaloid from lotus seed embryo, inhibits human lung cancer cell growth by MAPK activation and cell cycle arrest. BioFactors 40:121−31

    doi: 10.1002/biof.1115

    CrossRef   Google Scholar

    [102]

    Marthandam Asokan S, Mariappan R, Muthusamy S, Velmurugan BK. 2018. Pharmacological benefits of neferine - a comprehensive review. Life Sciences 199:60−70

    doi: 10.1016/j.lfs.2018.02.032

    CrossRef   Google Scholar

    [103]

    Chang M, Ding S, Dong X, Shang X, Li Y, et al. 2022. Liensinine inhibits cell growth and blocks autophagic flux in nonsmall-cell lung cancer. Journal of Oncology 2022:1533779

    doi: 10.1155/2022/1533779

    CrossRef   Google Scholar

    [104]

    Park KM, Yoo YJ, Ryu S, Lee SH. 2016. Nelumbo Nucifera leaf protects against UVB-induced wrinkle formation and loss of subcutaneous fat through suppression of MCP3, IL-6 and IL-8 expression. Journal of Photochemistry and Photobiology B: Biology 161:211−16

    doi: 10.1016/j.jphotobiol.2016.04.006

    CrossRef   Google Scholar

    [105]

    Perry G, Cash AD, Smith MA. 2002. Alzheimer disease and oxidative stress. Journal of Biomedicine and Biotechnology 2:120−23

    doi: 10.1155/S1110724302203010

    CrossRef   Google Scholar

    [106]

    Xiao J, Zhang J, Chen H, Feng X, Wang J. 2005. Inhibitory effects of isoliensinine on bleomycin-induced pulmonary fibrosis in mice. Planta Medica 71:225−30

    doi: 10.1055/s-2005-837821

    CrossRef   Google Scholar

    [107]

    Chen M, Zhang J, Wang J, Gao L, Chen X, et al. 2015. Anti-fibrotic effects of neferine on carbon tetrachlorideinduced hepatic fibrosis in mice. The American Journal of Chinese Medicine 43:231−40

    doi: 10.1142/S0192415X15500159

    CrossRef   Google Scholar

    [108]

    Menéndez-Perdomo IM, Facchini PJ. 2023. Elucidation of the ( R)-enantiospecific benzylisoquinoline alkaloid biosynthetic pathways in sacred lotus ( Nelumbo nucifera). Scientific Reports 13:2955

    doi: 10.1038/s41598-023-29415-0

    CrossRef   Google Scholar

    [109]

    Rueffer M, Zenk MH. 1987. Distant precursors of benzylisoquinoline alkaloids and their enzymatic formation. Verlag der Zeitschrift für Naturforschung 42C:319−32

    doi: 10.1515/znc-1987-0402

    CrossRef   Google Scholar

    [110]

    Hagel JM, Facchini PJ. 2013. Benzylisoquinoline alkaloid metabolism: a century of discovery and a brave new world. Plant and Cell Physiology 54:647−72

    doi: 10.1093/pcp/pct020

    CrossRef   Google Scholar

    [111]

    Mizutani M, Sato F. 2011. Unusual P450 reactions in plant secondary metabolism. Archives of Biochemistry and Biophysics 507:194−203

    doi: 10.1016/j.abb.2010.09.026

    CrossRef   Google Scholar

    [112]

    Ming R, VanBuren R, Liu Y, Yang M, Han Y, et al. 2013. Genome of the long-living sacred lotus ( Nelumbo nucifera Gaertn.). Genome Biology 14:R41

    doi: 10.1186/gb-2013-14-5-r41

    CrossRef   Google Scholar

    [113]

    Gui S, Peng J, Wang X, Wu Z, Cao R, et al. 2018. Improving Nelumbo nucifera genome assemblies using high-resolution genetic maps and BioNano genome mapping reveals ancient chromosome rearrangements. The Plant Journal 94:721−34

    doi: 10.1111/tpj.13894

    CrossRef   Google Scholar

    [114]

    Shi T, Rahmani RS, Gugger PF, Wang M, Li H, et al. 2020. Distinct expression and methylation patterns for genes with different fates following a single whole-genome duplication in flowering plants. Molecular Biology and Evolution 37:2394−413

    doi: 10.1093/molbev/msaa105

    CrossRef   Google Scholar

    [115]

    Qi H, Yu F, Deng J, Zhang L, Yang P. 2023. The high-quality genome of lotus reveals tandem duplicate genes involved in stress response and secondary metabolites biosynthesis. Horticulture Research 10:uhad040

    doi: 10.1093/hr/uhad040

    CrossRef   Google Scholar

    [116]

    Liscombe DK, Macleod BP, Loukanina N, Nandi OI, Facchini PJ. 2005. Evidence for the monophyletic evolution of benzylisoquinoline alkaloid biosynthesis in angiosperms. Phytochemistry 66:1374−93

    doi: 10.1016/j.phytochem.2005.04.029

    CrossRef   Google Scholar

    [117]

    Vimolmangkang S, Deng X, Owiti A, Meelaph T, Ogutu C, et al. 2016. Evolutionary origin of the NCSI gene subfamily encoding norcoclaurine synthase is associated with the biosynthesis of benzylisoquinoline alkaloids in plants. Scientific Reports 6:26323

    doi: 10.1038/srep26323

    CrossRef   Google Scholar

    [118]

    Nelson DR, Schuler MA. 2013. Cytochrome P450 genes from the sacred lotus genome. Tropical Plant Biology 6:138−51

    doi: 10.1007/s12042-013-9119-z

    CrossRef   Google Scholar

    [119]

    Menéndez-Perdomo IM, Facchini PJ. 2020. Isolation and characterization of two O-methyltransferases involved in benzylisoquinoline alkaloid biosynthesis in sacred lotus ( Nelumbo nucifera). Journal of Biological Chemistry 295:1598−612

    doi: 10.1074/jbc.RA119.011547

    CrossRef   Google Scholar

    [120]

    Yu Y, Liu Y, Dong G, Jiang J, Leng L, et al. 2023. Functional characterization and key residues engineering of a regiopromiscuity O-methyltransferase involved in benzylisoquinoline alkaloid biosynthesis in Nelumbo nucifera. Horticulture Research 10:uhac276

    doi: 10.1093/hr/uhac276

    CrossRef   Google Scholar

    [121]

    Li J, Xiong Y, Li Y, Ye S, Yin Q, et al. 2019. Comprehensive analysis and functional studies of WRKY transcription factors in Nelumbo nucifera. International Journal of Molecular Sciences 20:5006

    doi: 10.3390/ijms20205006

    CrossRef   Google Scholar

    [122]

    Li J, Li Y, Dang M, Li S, Chen S, et al. 2022. Jasmonate-responsive transcription factors NnWRKY70a and NnWRKY70b positively regulate benzylisoquinoline alkaloid biosynthesis in lotus ( Nelumbo nucifera). Frontiers in Plant Science 13:862915

    doi: 10.3389/fpls.2022.862915

    CrossRef   Google Scholar

  • Cite this article

    Wei X, Zhang M, Yang M, Ogutu C, Li J, et al. 2024. Lotus (Nelumbo nucifera) benzylisoquinoline alkaloids: advances in chemical profiling, extraction methods, pharmacological activities, and biosynthetic elucidation. Vegetable Research 4: e005 doi: 10.48130/vegres-0024-0004
    Wei X, Zhang M, Yang M, Ogutu C, Li J, et al. 2024. Lotus ( Nelumbo nucifera) benzylisoquinoline alkaloids: advances in chemical profiling, extraction methods, pharmacological activities, and biosynthetic elucidation. Vegetable Research 4: e005 doi: 10.48130/vegres-0024-0004

Figures(8)  /  Tables(4)

Article Metrics

Article views(1105) PDF downloads(171)

REVIEW   Open Access    

Lotus ( Nelumbo nucifera) benzylisoquinoline alkaloids: advances in chemical profiling, extraction methods, pharmacological activities, and biosynthetic elucidation

Vegetable Research  4 Article number: e005  (2024)  |  Cite this article

Abstract: As a member of the only two species in the Nelumbonaceae family, lotus ( Nelumbo nucifera) accumulates abundant benzylisoquinoline alkaloids (BIAs) in almost all of its tissues. Evidence from both traditional and modern medicine suggest great potential of the lotus BIAs in developing novel drugs for the prevention and treatment of diverse life-threatening diseases. This review provides a comprehensive summary on the up-to-date advances in the chemical profiling, extraction methods, pharmacological activities, and biosynthesis of lotus BIAs. Currently, a total of 59 BIAs structurally belonging to the 1-benzylisoquinoline, aporphine, and bis-BIA categories have been identified in various lotus tissues, with their predominant accumulation in the leaf and plumule. In contrast to the common S-conformers in Ranunculales, most lotus BIAs are R-conformers. Solvent extraction is still the most widely used BIA extraction method in lotus at the industrial level, however, numerous greener and highly advanced extraction techniques have also been developed. High-performance liquid chromatography (HPLC) followed by electrospray ionization (ESI) and tandem mass spectrometry (MS/MS) techniques are currently the most commonly used methods for separation, quantification, and characterization of lotus BIAs. Moreover, the pharmacological activities of major BIAs isolated from lotus leaves and plumules are discussed, and their biosynthetic pathways proposed based on recent functional characterization studies of lotus BIA biosynthetic genes. Finally, a summary discussion is provided on the future research trajectories in elucidating lotus BIA biosynthesis, storage, and transportation, as well as their potential application in clinical drug development.

    • Lotus, Nelumbo nucifera Gaertn. (commonly known as Asian lotus), is one of the most commercially significant aquatic plants in the world. It belongs to the Nelumbonaceae family, which contains a single genus Nelumbo, with the American lotus ( Nelumbo lutea Pear) as the only other extant species. The Asian lotus is geographically widely distributed in most Asian countries, as well as in Australia and Russia ( Fig. 1). The American lotus, also known as yellow lotus, occurs primarily in eastern regions of North American and the Caribbean [ 1] . Due to geographical isolation by the Pacific Ocean, the two lotus species exhibit distinct morphologies in plant size, leaf, and petal shape, particularly in their petal colors. The Asian lotus displays diverse flower colors, including white, pink, red, pale yellow, as well as a variety of color patterns, while the American lotus only exhibit yellow flowers ( Fig. 1). The two species, however, are genetically crossable and share rather similar aquatic habits and life cycles. Both of the two species have eight pairs of chromosomes (2n = 16), with approximately 800 Mb genome sizes, highly conserved gene contents, and little chromosomal rearrangements [ 2, 3] .

      Figure 1. 

      Geographical distribution of lotus accessions collected in the Wuhan National Lotus Germplasm Bank at the Wuhan Botanical Garden of the Chinese Academy of Sciences. The different colored dots indicate cultivars belonging to different categories.

      While the American lotus predominantly grows naturally in the wild, the Asian lotus has been domesticated and cultivated over 3,000 years, for its nutritional, ornamental, and medicinal attributes [ 4] . The total lotus cultivation area in China is estimated to cover over 530,000 ha, and so far at least 4,500 lotus cultivar germplasm resources have been conserved [ 5] . Most of these cultivars are hybrids developed by traditional crossing, while approximately 300 are local genotypes collected from wild lakes all over the world ( Fig. 1) [ 2] . These lotus cultivars are grouped into four categories based on their agricultural uses, including the flower lotus for ornamental purpose; the rhizome lotus for edible rhizome production; the seed lotus for harvesting fresh or mature seeds; and the medicinal lotus for pharmaceutical uses [ 6] .

      As an ancient traditional herb, all lotus plant tissues have been used in the treatment of various diseases, such as fever, sunstroke, sweating, obesity, ischemia, cancer, diabetes, hepatopathy, and hypertension [ 710] . Its medicinal properties are largely ascribed to the abundantly accumulated active components in different lotus organs, including alkaloids, flavonoids, terpenoids, polysaccharides, saponins, and minerals. Of these compounds, benzylisoquinoline alkaloids (BIAs) are regarded as the most important pharmacological ingredients [ 11, 12] . Here, we summarize the recent advances in the chemical profiles, pharmacological activities, extraction methods, and elucidations of its biosynthesis pathway.

    • BIAs are a class of tyrosine-derived nitrogenous secondary metabolites with over 2,500 known structures [ 13] . The core chemical structure of BIAs consist of a benzene ring, a pyridine ring, and a second aromatic ring ( Fig. 2) [ 14] . Like other plant-derived alkaloids, most BIAs are significantly bioactive [ 15] . Typical pharmacologically significant BIAs include morphine and codeine with analgesic traits, sanguinarine and berberine with antimicrobial properties, as well as noscapine, papaverine, and tubocurarine with antitussive, vasodilative, and muscle relaxant properties, respectively. The occurrence of BIAs in plants is restricted primarily in the order Ranunculales and eumagnoliids. However, their accumulation has been detected in other families, including Rutaceae, Lauraceae, Cornaceae, and Nelumbonaceae.

      Figure 2. 

      1-Benzylisoquinoline alkaloids reported in Nelumbo nucifera.

      The lotus plant richly accumulate BIAs in almost all its organs [ 11] . To date, at least 61 alkaloids ( Table 1) have been identified in lotus since the first isolation of nuciferine, roemerine, and O-nornuciferine alkaloids from the plant in the 1960s [ 16, 17] . Notably, all reported lotus alkaloids are BIAs except Oleracein E and Methylcorypalline, which contain only basic isoquinoline structures. Based on their structures, these BIAs can further be divided into three subclasses, including 1-benzylisoquinolines, aporphines, and bisbenzylisoquinolines ( Figs 24). 1-Benzylisoquinoline alkaloids are basically intermediates in the biosynthesis of aporphines and bisbenzylisoquinolines, and are mostly accumulated in trace amounts in lotus tissues [ 18] . In contrast, aporphines and bisbenzylisoquinolines are the end BIA products in lotus, with their accumulation levels accounting for over 95% of the total BIA content [ 11] .

      Table 1.  BIAs detected in the lotus ( Nelumbo nucifera) tissues. Alkaloids are assigned with numbers as shown in Figures 25.

      No. Alkaloid Formula Enantiomer Organ Reference
      1-BENZYLISOQUINOLINE
      1 Lotusine C 19H 24NO 3 + E, S, F [ 25]
      2 Methyl lotusine L
      3 Armepavine C 19H 23NO 3 (−)-R and (+)-S S, F, L [ 21]
      4 Coclaurine C 17H 19NO 3 (+)-R S, L [ 26]
      5 N-norarmepavine C 18H 21NO 3 (+)-R L [ 27]
      6 N-methylisococlaurine C 18H 21NO 3 L [ 16, 28]
      7 N-methylcoclaurine C 18H 21NO 3 (−)-R L, S [ 16]
      8 Isococlaurine C 19H 24NO 3+ L
      9 Methylhigenamine S [ 17]
      10 Norcoclaurine-6- O-glucoside S
      11 Norcoclaurine C 16H 17NO 3 (+)-R and (+)-S S, L [ 26, 29]
      12 Argemexirine S, L
      13 6-demethy-4-methyl- N-methylcoclaurine C 18H 21NO 3 S [ 30]
      14 Nor- O-methylarmepavine C 20H 25NO 3 S [ 30]
      15 4’- N-methylcoclaurine C 19H 23NO 3 L, S [ 17]
      16 4’-methyl coclaurine L, S [ 17]
      17 Bromo methyl armepavine L, S [ 17]
      18 Methoxymethy lisoquinoline L, S [ 17]
      19 Higenamine P [ 31, 32]
      20 Higenamine glucoside P [ 33]
      APORPHINE
      21 Nuciferine C 19H 21NO 2 (−)-R S, F, L [ 34, 35]
      22 N-nornuciferine C 18H 19NO 2 (−)-R S, L [ 26]
      23 Roemerine C 18H 17NO 2 (−)-R S, F, L [ 16, 26, 32]
      24 O-nornuciferine C 18H 19NO 2 (−)-R S, F, L [ 32, 36]
      25 Anonaine C 17H 15NO 2 (−)-R S, L [ 16, 26]
      26 Lirinidine C 18H 19NO 2 (−)-R S, L [ 37]
      27 Nuciferine- N-Methanol F
      28 Nuciferine- N-Acetyl F
      29 Anonaine- N-Acetyl F
      30 Caaverine C 17H 17NO 2 (−)-R S, L [ 20, 38]
      31 Oxidation-nuciferine S, L, F [ 19, 30]
      32 Asimilobine C 17H 17NO 2 (−)-R S, F, L [ 17, 20]
      33 Methyl asimilobine S, L [ 17]
      34 N-methyl asimilobine S, L [ 16, 39]
      35 Roemerine- N-oxide S, L [ 16, 40]
      36 N-methyl asimilobine- N-oxide S, L, F [ 16, 19]
      37 Nuciferine- N-oxide S, L, F [ 16, 19]
      38 Dehydroanonaine C 17H 13NO 2 L [ 16]
      39 Dehydronuciferine C 19H 19NO 2 L [ 16]
      40 Dehydroaporphine C 18H 15NO 2 L [ 41]
      41 Nelumnucine S, L
      42 Dehydroroemerine L [ 16]
      43 Liriodenine C 17H 9NO 3 L [ 16, 40]
      44 7-hydroxydehy dronuciferine C 19H 19NO 3 L [ 16]
      45 Pronuciferine C 19H 21NO 3 (+)-R and (−)-S S, F, L [ 16, 26, 40]
      46 Glaziovine S
      47 Lysicamine C 18H 13NO 3 L [ 38, 42]
      48 Cepharadione L [ 38]
      BISBENZYLISOQUINOLINE
      49 Neferine C 38H 44N 2O 6 1R, 1'S S, F, E [ 17, 31]
      50 Liensinine C 37H 42N 2O 6 1R, 1'R S, F, E [ 35]
      51 Isoliensinine 1R, 1'S S, F, E [ 25]
      52 N-norisoliensinine C 36H 40N 2O 6 S, F, E [ 25]
      53 6-hydroxynorisoliensinine C 36H 40N 2O 6 S, F, E
      54 Methyl neferine S, E [ 10, 17]
      55 Nelumboferine C 36H 40N 2O 6 S, E [ 10, 17]
      56 Negferine C 38H 44N 2O 6 L [ 17, 26]
      57 Nelumborine F [ 17]
      58 Dauricine S, F [ 43]
      TRIBENZYLISOQUINOLINE
      59 Neoliensinine C 63H 70N 3O 10 1R, 1'S, 1''R E [ 44]
      L, Leaf; E, embryo; F, flowers; S, seeds; R, rhizome; LS: leaf sap; NS, not specified.

      Figure 3. 

      Aporphine-type alkaloids isolated from Nelumbo nucifera.

      Figure 4. 

      Bis- and tri- benzylisoquinoline alkaloids isolated from Nelumbo nucifera.

      Unlike BIAs in the Ranunculales, lotus BIAs exhibit several unique features. Firstly, lotus BIAs show strict tissue specific accumulation patterns. Although BIAs are found in all the plant tissues, highest levels of approximately 3,000 μg/g fresh weight (FW) are accumulated in the laminae and plumules, followed by petals and petioles with about 500 and 100–200 μg/g FW, respectively. Conversely, lowest BIAs levels (less than 20 μg/g FW) occur in the rhizomes and stamens. Interestingly, the lotus bleeding sap contains extremely high BIAs level of up to 10,000 μg/g FW [ 11] . In addition, lotus leaves and petals predominantly accumulate aporphine BIAs, including nuciferine, O-nornuciferine, N-nornuciferine, roemerine, and anonaine, while the plumules mostly contain bis-BIAs of liensinine, isoliensinine, and neferine [ 11] . Secondly, while BIAs prevalently assume the ( S)-enantiomer conformation, lotus show enrichment of both ( R)- and ( S)-conformers. For example, both ( R)- and ( S)-stereoisomers of armepavine have been isolated from sacred lotus [ 1921] . To date, most aporphines isolated from lotus belong to the ( R)-configurations ( Table 1). Thirdly, all aporphine BIAs isolated from lotus lack the hydroxyl and methyl modifications at the C-3' and C-4' positions, suggesting that the ( R)- N-methylcoclaurine might be the key precursor for both aporphine and bis-BIA biosynthesis in lotus [ 22] .

      Lotus cultivars exhibit considerably diverse patterns of BIA composition and accumulation. Generally, seed cultivars accumulate the highest concentration of BIAs in leaves, followed by flower cultivars, while lowest levels are observed in the rhizome cultivars [ 11, 23] . Multivariate principal component analysis (PCA) of 92 lotus cultivars showed that the seed cultivars generally accumulate high levels of nuciferine and O-nornuciferine, whereas flower cultivars primarily accumulate high N-nornuciferine, roemerine, and anonaine alkaloids [ 23] . In contrast, rhizome cultivars contain no dominant BIAs. Cluster analysis of the 92 cultivars also indicated that the American cultivar ‘Meizhouhuanglian’ could group with other Asian lotus cultivars, which suggested the lack of obvious differentiation between the American and the Asian lotus accessions based on BIA content and composition [ 23] . Developmentally, the lowest BIAs levels are accumulated in the lotus leaves at the bud stages (developmental stage S1). BIAs levels then increase steadily in the leaf growth and expansion stages, peak at S6 stage, then decrease slightly at the S7 senescence stage ( Fig. 5a) [ 11] . Accumulation of BIAs in the lotus plumules is also developmentally regulated [ 24] . Major BIAs were almost undetectable in the plumules at 12 days post pollination (DAP), which later increased consistently until 21 DAP ( Fig. 5b).

      Figure 5. 

      BIA profiles in the leaf and plumule during development. (a) Lotus leaves showing the seven defined developmental stages. (b) BIA profiles in the seven leaf developmental stages. (c) Lotus plumules showing different developmental stages. (d) BIA content in the plumule at different developmental stages. S, leaf developmental stages; DAP, days post pollination. The figure is modified from previous reports [ 11, 24] .

    • Extraction of alkaloids from raw materials is the first analytical step in their identification, quantification, and application. Currently, three main alkaloid extraction methods with varied principles are widely used, including solvent extraction, distillation, expeller pressing and sublimation methods. Solvent extraction is presently the most popularly used method. The plant alkaloids occur mostly as salts of organic and inorganic acids [ 45, 46] , thus, acidic and basic solvents are initially required to remove the non-alkaloid compounds. Subsequently, appropriate purification methods, primarily acidic extraction is performed by mixing the ground sample materials with weak acid solutions (e.g., acetic acid in water, methanol, or ethanol), to obtain a desired level of purity. The solvent extraction is carried out in four steps: 1) assimilation of solvents into the target solid matrix; 2) alkaloid dissolution in solvents; 3) solvent diffusion out of a solid matrix; 4) collection of extracted solutes [ 47] . Theoretically, any factors that enhance the above steps can facilitate alkaloid extraction. Key factors that affect the extraction efficiency, include alkaloids-solvent solubility, sample particle size, solvent to solid ratio, extraction temperature, and extraction duration [ 48, 49] .

      The lotus BIAs are weak alkalescent compounds, with poor solubility or insolubility in water, but with rapid solubility in organic solvents. Based on the law of similarity and intermiscibility (like dissolves like), crude lotus BIAs extract are usually prepared in alcohol solvents (ethanol or methanol) [ 10, 19] . Other solvents, such as hydrochloric acid, dichloromethane and trichloromethane have also been used for lotus BIA extraction [ 20] . The solvent extraction efficiency can be enhanced by finely grinding the lotus tissues, solvent refluxing, microwaving, and sonication. A recent comparative study, using three extraction solvent (methanol, 50% methanol, and water) and two different extraction conditions (reflux for 120 min twice or sonication for 30 min twice), suggested that the 'reflux in methanol' method could result in highest BIA recovery from lotus flowers ( Table 2) [ 50] . Moreover, pre-basifying lotus leaves with 10% ammonia water revealed a significant two-fold nuciferine yield relative to that obtained from non-basified leaves [ 51] .

      Table 2.  Extraction efficiency of alkaloids from lotus flower.

      Extraction method Content (mg/g dry weight) a Total
      1 2 3 4 5 6 7 8 9 10
      Methanol, reflux 1.76 (100) 1.75 (100) 0.07 (100) 0.63 (100) 0.69 (100) 0.83 (100) 1.45 (100) 5.73 (100) 1.30 (100) 0.75 (100) 14.96 (100)
      50% Methanol, reflux 1.09 (62) 1.35 (77) 0.05 (71) 0.50 (79) 0.61 (88) 0.78 (94) 1.35 (93) 3.79 (66) 0.94 (73) 0.56 (75) 11.02 (74)
      H 2O, reflux 0.24 (14) 0.35 (20) nd. b 0.21 (33) 0.18 (26) 0.38 (45) 0.78 (54) 2.57 (45) 0.66 (51) 0.29 (38) 5.66 (38)
      Methanol, sonication 0.88 (50) 1.11 (64) 0.03 (44) 0.39 (62) 0.33 (48) 0.47 (56) 0.97 (67) 2.77 (48) 0.70 (54) 0.42 (56) 8.07 (54)
      50% Methanol, sonication 0.98 (56) 1.27 (73) 0.04 (58) 0.49 (78) 0.47 (69) 0.80 (96) 1.38 (95) 3.93 (69) 0.97 (75) 0.59 (79) 10.92 (73)
      H 2O, sonication 0.14 (8) 0.21 (12) nd. 0.12 (20) 0.08 (11) 0.25 (30) 0.53 (37) 1.91 (33) 0.48 (37) 0.19 (26) 3.91 (26)
      1. nuciferine; 2. N-nornuciferine; 3. N-methylasimilobine; 4. Asimilobine; 5. Pronuciferine; 6. Armepavine; 7. norarmepavine; 8. N-methylcoclaurine; 9. coclaurine; 10. Norjuziphine. a. relative value (%) against the content obtained by methanol under reflux is given in parentheses. b. less than the quantitation limit.

      The conventional solvent extraction method is typically time consuming, requires large volume of organic solvents, and high cost waste solvent treatment. Consequently, modern or greener extraction methods, such as supercritical fluid extraction (SFC), high-speed counter-current chromatography (HSCCC), and microwave assisted extraction (MAE) techniques have been developed and applied in BIAs extraction. SFE utilizes supercritical fluid as the extraction solvent, which has high solubility as liquid and high diffusivity as gas. SFE is almost pollution-free, and can efficiently extract alkaloids with no residual organic solvents [ 52] . The solvating properties of supercritical fluid can dramatically rise when the pressure and temperature are near their critical points. The supercritical carbon dioxide (S-CO 2) is one of the most widely used supercritical fluids due to its low critical temperature (31 °C), low cost, non-toxicity, chemical inertness, and non-flammability properties [ 46, 5354] . However, due to the relatively low polarity of S-CO 2, polar agents, such as methanol and water are used as modifiers to raise its polarity and enhance its efficiency for BIAs extraction from lotus tissues. A previous study showed that the highest nuciferine yield of 325.54 μg/g could be achieved with extraction conditions set at 70 °C, 30 MPa, flow rate 0.2 ml/min, 2 h extraction time, and with 10% (v/v) diethylamine and 1% (v/v) water in methanol as the polarity modifier [ 51] .

      The HSCCC is a liquid-liquid partition chromatography technique that uses no solid support matrix. This method permits the introduction of crude samples directly into the hollow column, and eliminates the adsorbing effects on stationary phase material as well as artifact formation, thus offering maximum sample recovery capacity and a wide range of solvent system selection [ 55] . Ma et al. used a simple two-phase solvent system comprising of n-hexane-ethyl acetate-methanol-acetonitrile-water (5:3:3:2.5:5, v/v/v/v/v) to successfully purify four main aporphine alkaloids, including 2-hydroxy-1-methoxyaporphine, pronuciferine, nuciferine, and roemerine from a crude extract of lotus leaves [ 56] .

      In addition to the above-mentioned methods, several other extraction methods, such as maceration, percolation, soxhlet, and pressurized liquid extraction are available for selection. Modern and green methods are preferred mostly due to their energy-efficiency, speed, and less solvent requirement, and high extraction yield. However, traditional techniques are still widely used at the industrial level due to their low investment cost. Overall, the selection of suitable extraction techniques depends not only on the physical, chemical, and stability of the target alkaloids, but also on time and cost of the extraction methods.

    • Following extraction, the resulting crude BIA extracts are mixtures of a variety of natural products, such as phenolics, carotenes, glycosides, and terpenes, which require further separation and purification. The separation of BIAs depends largely on their physical and chemical characteristics, such as adsorption properties, partition coefficients, molecular sizes, solubility, and ionic strengths [ 47] . BIAs are mostly water-insoluble but soluble in organic solvents, while their acid salts are water-soluble or soluble in dilute acids, thus, these compounds can be precipitated by adding solvents (e.g., sodium bicarbonate, ammonia, or tartaric acid) that alter the reaction pH levels. Similar theory has been used to develop pH-zone-refining counter current chromatography techniques for lotus BIAs isolation [ 35, 57] .

      Isolation of pure lotus BIAs using conventional methods, such as column chromatography and thin-layer chromatography (TLC) is extremely difficult due to their high structural similarity. The structurally similar BIAs can instead be rapidly separated by counter-current chromatography (CCC) according to their different partition coefficients in two immiscible solvents. The CCC is a liquid-liquid partition chromatography technique with no solid support matrixes, and has been widely used for the preparative isolation of lotus BIAs. Accurate selection of the solvent system is highly crucial when using the CCC method. Generally, the two-phase solvent system should have: no decomposition or denaturation effects on the target compounds, sufficient sample solubility and suitable partition coefficient values, and satisfactory retention capacity of the stationary phase [ 58] . For efficient separation, the partition coefficient ( K) value of the target alkaloids and the separation factor between two BIAs ( K2/ K1) should be close to 1 and greater than 1.5, respectively [ 55] . Other factors, such as the rotary speed, the mobile phase flow rate, and the column temperature also affect the target alkaloid separation.

      Preparative separation of liensinine and its analogues from lotus plumules using CCC was first reported by Wu et al. [ 59] . The authors used two-phase solvent systems of light petroleum (b.p. 60–90 °C)–ethyl acetate–tetrachloromethane–chloroform–methanol–water (1:1:4:4:6:2, v/v) and ethyl acetate–tetrachloromethane–methanol–water (1:6:4:1, v/v) ( Table 3) to successfully isolate bis-BIAs from lotus plumules in small- and large-scale, respectively. To limit the application of deleterious tetrachloromethane and chloroform solvents, another simplified HSCCC solvent system of n-hexane–ethyl acetate–methanol–water (5:8:4:5, v/v, containing 0.5% NH 4OH) was developed and successfully used to separate liensinine, isoliensinine, and neferine from crude lotus extract [ 60] . Later, several pH-zone-refining CCC solvent systems, which generally use 10 mM triethylamine in the upper organic phase and 5 mM HCl in the aqueous phase for adjustment of pH and K values of target compounds were developed [ 35, 57, 61, 62] . These two phase solvent systems were used to successfully isolate over 98% pure BIA singletons from both plumules and leaves. Other than the CCC technique, a preparative liquid chromatography has also been developed for high purity laboratory-scale isolation of liensinine, isoliensinine, and neferine [ 63] .

      Table 3.  Major two-phase solvent systems developed for preparative separation of lotus BIAs through counter-current chromatography (CCC) techniques.

      Solvent systems Mix ratios (v/v) Target BIAs Reference
      Light petroleum (60–90 °C)–ethyl acetate–tetrachloromethane–chloroform–methanol–water 1:1:4:4:6:2 Small scale
      Plumule BIAs
      [ 59]
      Ethyl acetate–tetrachloromethane–chloroform–methanol–water 1:6:4:1 Small scale
      Plumule BIAs
      [ 59]
      n-hexane–ethyl acetate–methanol–water 5:8:4:5
      0.5% NH 4OH
      Plumule BIAs [ 60]
      n-hexane–ethyl acetate–methanol–water 5:5:2:8
      10 mM triethylamine
      5 mM HCl
      Plumule BIAs [ 57]
      Diethyl ether – Na 2HPO 4/NaH 2PO 4 (pH = 7.2 – 7.5) 1:1 Plumule BIAs [ 62]
      Petroleum ether (60–90 °C)–ethyl acetate–methanol–water 5:5:2:8
      10 mM triethylamine
      5 mM HCl
      Leaf BIAs [ 61]
      n-hexane-ethyl acetate-methanol-water-[C 4mim][PF 6] 5:2:2:8:0.1
      10 mM triethylamine
      3 mM HCl
      Whole plant BIAs [ 35]

      After extraction and purification, lotus BIAs are subjected to further identification and quantification processes. High-performance liquid chromatography (HPLC) techniques, particularly the reversed-phase LC methods, were predominantly employed [ 45] . Identification of known BIA compounds is normally accomplished by HPLC separation and UV detection, followed by electrospray ionization (ESI), and tandem mass spectrometry (MS/MS). The reversed-phase C 18 columns are most ideal, and the application of aqueous acetonitrile containing 0.1% triethylamine as mobile phase has been shown to effectively separate both lotus leaf and plumule BIAs with strong peak signals and fine peak shapes [ 23, 63, 64] . Other optimized HPLC parameters for lotus BIAs characterization include: column temperature 30 C; 270–280 nm UV/photodiode array (DAD) detection wavelength; the gradient elution mode, 40%–80% acetonitrile at 0–15 min, 80% acetonitrile at 15–18 min, 80%–40% acetonitrile at 18–19 min, 40% acetonitrile at 19–25 min; and a flow rate 0.8 ml/min. The MS detection of lotus BIAs are preferably conducted in the positive mode, with cone voltage of 20 V and a nebulizer gas temperature/pressure of 150 °C/21 psi. The needle, shield, and capillary voltage parameters are normally used with default settings. Such HPLC-DAD-ESI-MS/MS methods have been used to identify the major leaf BIAs, including nuciferine, N-nornuciferine, O-nornuciferine, anonaine, and roemerine, as well as plumule BIAs, such as liensine, isoliensinine, and neferine ( Table 4; Fig. 6; Fig. 7).

      Table 4.  Identification of major lotus leaf and plumule alkaloids and their HPLC-MS/MS ion characteristics.

      Peaks T R a (min) Molecular weight m/ z
      [M + H] +
      Major fragment ions Alkaloids
      1* 7.56 281 282 251/219 N-nornuciferine
      2* 9.03 281 282 265/250 O-nornuciferine
      3* 10.17 265 266 249/219 Anonaine
      4* 12.43 295 296 265/250 Nuciferine
      5* 13.39 279 280 249 Roemerine
      6 6.61 610 611 503/283/206 Liensinine
      7 9.47 610 611 489/297/192 Isoliensinine
      8 17.72 624 625 503/297/206 Neferine
      * The retention time (T R a (min) of peaks 1–5 as obtained by Chen et al. [ 23] .
      ∆ The retention time of peaks 6–9 as reported by Chen et al. [ 63] .

      Figure 6. 

      Analytical mass spectra of lotus leaf BIAs and their fragmentation pathways. The chemical structures and the corresponding peak numbers 1 to 5 represent signals for N-nornuciferine, O-nornuciferine, anonaine, nuciferine, and Roemerine, respectively. Figures are modified from Luo et al. [ 65 ] and Chen et al. [ 23 ].

      Figure 7. 

      Analytical mass spectra of lotus leaf BIAs and their fragmentation pathways. The chemical structures and the corresponding peak numbers 6 to 8 represent signals for liensinine, isoliensinine, and neferine, respectively. Figures are modified from Chen et al. [ 64 ], Deng et al. [ 11 ], and Lai et al. [ 66 ].

      Currently, lotus BIAs are predominantly identified using the ultra-performance liquid chromatography technique equipped with quadrupole time-of-flight mass spectrometry (UPLC-QTof-MS). In contrast to traditional HPLC-MS methods, the UPLC-Q-Tof-MS technique exhibits higher sensitivity and resolution, and mass measurement accuracy, making it a powerful and reliable analytical technique for plant metabolite identification [ 65, 66] . Under optimized UPLC-Q-Tof-MS conditions, over 20 BIAs were identified from lotus leaves and plumules, based on their chromatographic characteristics, UV spectra, exact mass, and MS fragments [ 31, 65, 67] . Moreover, these studies further verified previous observations that liensinine, isoliensinine, neferine, and lotusine are the major BIAs in lotus plumules, while those of aporphines, including nuciferine, roemerine, Anonaine, N- and O-nornuciferine are the major BIAs in lotus leaves.

    • In addition to their nutritional roles, lotus is also a key source of herbal traditional Chinese medicine. Lotus was originally used to treat various diseases, such as pharyngitis, chest pain, cough, vomiting blood, fever, and heat stroke [ 68, 69] . To date, extensive studies have identified a wide range of lotus bioactive ingredients, their pharmacological efficiencies, and health benefits. As key bioactive lotus constituents, BIA alkaloids have nowadays received significant attention. In the following sections, we present an overview of pharmacological activities of lotus BIAs from various tissues.

    • As introduced above, lotus leaves accumulate primarily aporphine-type BIAs, with Nuciferine, N- and O-nornuciferine as the richest components. Diversified pharmacological activities of lotus leaves therefore attribute predominantly to these three BIAs. The nuciferine, for example, has shown anti-obesity, anti-virus, antioxidant, anti-cardiovascular, antimicrobial, and anti-cancer activities.

    • Lipids are maintained through dynamic balances within the cell as Triglyceride (TG) by a series of synthesis and catabolism related transcription factors (TFs) and enzymes. Lotus alkaloids can exert lipid-regulating effects in several ways, including mainly inhibition of lipid synthesis and absorption, inhibition of cell proliferation and differentiation, and interaction with proteins. A study on the effects of nuciferine on blood lipids in male golden hamsters, feeding with normal diet, high-fat diet (HFD), or HFD supplemented with nuciferine (10 and 15 mg/kg·BW/day), revealed a reduction in total cholesterol (TC), TG, low-density lipoprotein and free fatty acids in hamsters treated with different doses of nuciferine after eight weeks [ 70] . In addition, nuciferine could alleviate dyslipidemia, and liver steatosis by inhibiting the expression of hepatic genes related to lipid metabolism in hamsters fed with a high-fat diet [ 70, 71] . Similarly, a recent study demonstrated that lotus leaf extracts could inhibit adipogenesis in 3T3-L1 preadipocytes and suppress obesity in high-fat diet-induced obese rats [ 72] . Overall, these pharmacological effects demonstrate the potential of lotus leaf BIAs as natural lipid-lowering agents.

    • Currently, approximately 90% of global type 2 diabetic patients are characterized by hyperinsulinemia and insulin resistance. Therefore, controlling the blood glucose levels of patients is crucial for diabetes treatment [ 73] . Huperzine has been reported to lower blood sugar via various mechanism in the body. Conversely, nuciferine can increase the glucose uptake by fat and muscle cells, thereby promoting insulin secretion by pancreatic β-cells [ 56] . Nuciferine is also directly involved in the process of insulin secretion and has been shown to augment insulin secretion in the pancreatic β-cells by shutting down or stimulating the amplification of adenosine triphosphate pathway [ 74] .

    • Free radicals or oxidants that break down cells and tissues can affect metabolic function and cause different health challenges. Antioxidants are substances that effectively inhibit oxidation reaction of free radicals at low concentrations [ 75] . The antioxidant capacity of aporphine alkaloids isolated from the lotus leaves, including ( R)- N-methylasimilobine, lysicamine, and ( R)-nuciferine have been screened using antiradical scavenging, metal chelating, and ferric reducing power assays [ 38] . In addition, a previous study demonstrated that lotus leaf-fermented broth could exhibit 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging activity, ferric ion reducing power, superoxide dismutase-like activity, tyrosinase inhibition, and nitrite scavenging activity, which provided scientific basis for the application of sugar-fermented lotus leaves as an antioxidant condiment [ 76] . A recent study reported that, to enhance, achieve, and maintain higher scavenging activity of DPPH radicals and hydroxyl radicals for lotus leaves, the optimal treatment should include oven drying the leaves at ~55–65 °C to maintain alkaloids and amino acids content [ 77] .

    • Bacteria is known to be one of the leading causes of numerous diseases. Over exposure or misuse of antibiotics can result in antibacterial resistance. Chinese herbal medicine has been demonstrated to be an excellent alternative for the treatment of such bacterial infections to avoid antibiotic resistance [ 78] . The n-butanol compound lotus leaves extracts can effectively inhibit peridontitis-causing bacteria, such as Actinobacillus actinomycetemcomitans Y4, Actinomyces viscosus 19246, Porphyromonas gingivalis 33277, Fusobacterium nucleatum 25586, and Actinomyces naeslundii wvl 45 [ 79] . Moreover, the lotus leaf alkaloids not only have anti-mitogenic effects, but also exhibit a stronger antibacterial effect on Escherichia coli. The lotus leaf alkaloids and flavonoids are considered as key bioactive components responsible for the antibacterial activity.

    • A key characteristic of cancer is the indefinite cell proliferation, and lotus leaf alkaloids have been shown to exhibit antiproliferative properties against several cancer cells. For example, in the presence of nicotine, nuciferine could significantly inhibit the proliferation of non-small cell lung cancer cells, suppress the activity of Wnt/β-catenin signaling, enhance Axin stability, and induce apoptosis. In addition, nuciferine could also down-regulate the expression levels of β-catenin and its downstream targets, including c-myc, cyclin D, and VEGF-A [ 80] . Methanolic lotus leaves extract, containing nuciferine, N-methylasimilobine, (-)-lirinidine, 2-hydroxy-1-methoxy-6a, and 7-dehydroaporphine, significantly inhibited melanogenesis in B16 melanoma cells [ 19] . Furthermore, the inhibitory potential of nuciferine against the growth of human breast cancer cells has been reported [ 81] . The inhibitory effect of nuciferine is likely due to its ability to block cancer cell division cycle and enhance cancer cell apoptosis.

    • A recent study reported that 40 mg/kg nuciferine could not only significantly reduce aortic lesion and vascular plaque in ApoE(-/-) mice fed with a high-fat diet, but also attenuate migration and proliferation of vascular smooth muscle cells in vivo and in vitro through the Calm4/MMP12/AKT signaling pathway [ 82] . Nuciferine has also been associated with reduced vascular wall inflammation and significant down-regulation of vascular inflammatory factors, such as IL-1β, TNF-α, MCP-1, and NF- κB, with the latter being a major regulator during all phases of the inflammatory response [ 83] . Additionally, lotus leaf alkaloids are linked with vascular remodeling by down-regulating MMP-2 and 9 expression and inducing TIMP-2 expression. MMP is an important protease that degrades the extracellular matrix (ECM), while TIMP is a matrix metalloproteinase inhibitor, both of which contribute to atherosclerosis development [ 84] .

    • In contrast to lotus leaves, lotus plumules accumulate mainly bis-BIAs, with liensinine, isoliensinine, and neferine the most abundant components. The bioactivities of these bis-BIAs contribute largely to the pharmacokinetic properties of lotus plumules.

    • Antioxidant compounds, including alkaloids, phenolics, and saponins could be obtained from lotus plumules using 50% ethanol solvent extraction, and DPPH as well as nitric oxide in-vitro assays to test the hydro alcoholic extract of lotus plumules (HANN) revealed excellent free radical scavenging activity [ 85] . Neferine not only exhibited scavenging activity against ABTS, DPPH, NO, ONOO and ${\text{O}^-_2} $ radicals, but also showed significant inhibitory properties against lipid peroxidation and protein nitration in the antioxidant assays (cell-free). In vivo assays also revealed that neferine could significantly reduce NFκB activation and moderately inhibit NO synthesis [ 86] . Neferine could also inhibit various diseases, such as vascular inflammation, hyperglycemia, and liver disease by reducing NO concentration in different cells and inhibiting the elevation of intracellular molecules, such as reactive oxygen species (ROS) and malondialdehyde (MDA) [ 87] . In addition, total alkaloid from Nelumbinis Plumula (NPA) and its main alkaloids, including liensinine, isoliensinine, and neferine showed significant cytoprotective effect on oxidative stress induced by tert-butyl hydroperoxide (t-BHP) in the human hepatocellular HepG2 cell line, and the protective effect could be associated with reduced ROS formation, thiobarbituric acid-reactive substance production, lactate dehydrogenase release, and elevated GSH levels [ 88] .

    • Neferine is a crucial skin anti-inflammatory and anti-aging agent. For example, a previous study demonstrated that neferine could reduce the phosphorylation level of the MAPK/NF-κB pathway, and inhibit mast cell degranulation and cytokine expression by suppressing elevated intracellular calcium in mast RBL-2H3 cells stimulated with A23187/phorbol ester (PMA) [ 89] . In lipopolysaccharide-induced human endothelial cells, neferine could significantly inhibit the formation of inflammatory mediators, such as NO, tumor necrosis factor-α, cyclooxygenase-2, inducible nitric oxide synthase, and interleukin 1β due to its ability to regulate mitogen-activated protein kinase and nuclear factor-κβ pathways [ 90] . In addition, neferine could inhibit lipopolysaccharide and dextran sulfate sodium-induced inflammation both in vitro and in vivo, alter protein expression of iNOS, COX-2, receptor-interacting protein 1 (RIP1), RIP3, mixed lineage kinase domain-like protein (MLKL), and increase the protein expression of caspase-8 in colon tissues, all of which affected the incidence and prevalence of ulcerative colitis (UC) [ 91] . As the combined effects of oxidative stress and inflammation leads to pathogenesis of numerous diseases, the antioxidant and anti-inflammatory mechanisms of lotus seed alkaloids need to be further explored.

    • Liensinine and isoliensinine are two main bis-BIAs found in the lotus seed plumules, and both have been reported to have inhibitory effects against cell proliferation. For example, liensinine could functionally regulate the transforming growth factor β1-induced proliferation, migration, and signaling pathways of human tenon fibroblast cells via the mitogen-activated protein kinase 7 gene to enable rapid patients recovery after glaucoma surgery [ 92] . Liensinine could also not only significantly inhibit the proliferation of GBC cells in vitro by suppressing their G2/M phase growth in a dose- and time-dependent manner, but also induce apoptosis in gallbladder cancer cells by inhibiting the Zinc finger X-chromosomal protein (ZFX)-induced PI3K/AKT pathway [ 93] . In addition, previous reports have shown that liensinine could inhibit over-proliferation of gastric cancer cells and osteosarcoma cells [ 94, 95] . Isoliensinine is a liensinine isoform, which has also been shown to exhibit inhibitory effects against angiotensin II (Ang II)-induced proliferation of porcine coronary artery smooth muscle cells (CASMCs) [ 96] , as well as HepG2, Huh-7, and H22 hepatocellular carcinoma (HCC) cells [ 97] . Moreover, BIA alkaloids mixtures, including neferine, liensinine, and isoliensinine extracted from lotus plumules could delay or inhibit the abnormal proliferation and migration of pulmonary artery smooth muscle cell (PASMCs) by regulating the expression of p-SRC and PIM1 [ 98] .

    • Extensive anti-cardiovascular activities of Neferine, liensinine, and isoliensinine, such as anti-arrhythmic, anti-thrombic, and anti-hypertensive have been documented. Wicha et al. [ 99] demonstrated that neferine not only had hypotensive effects on NG-nitro-L-arginine methyl ester(L-NAME)-induced rats, but also induced vascular relaxation via the endothelial nitric oxide synthase (ENOS)/nitric oxide (NO)/soluble guanylate cyclase (SGC) pathway. Neferine has shown potential effectiveness in preventing episodes of reentrant ventricular tachycardia and sudden cardiac death after myocardial ischemic injury, and was effective against ischemic arrhythmias. In addition, neferine was shown to significantly inhibit adenosine diphosphate (ADP), collagen, arachidonic acid (AA) and platelet-activating factor (PAF)-induced platelet aggregation in rabbits. A more recent study showed that, unlike neferine, isoliensinine could effectively scavenge early afterdepolarizations (EADs) and delayed afterdepolarizations (DADs) by inhibiting I NaL and I CaL in ventricular myocytes, thus, demonstrating its potential anti-arrhythmic effects [ 100] .

    • To date, numerous studies have demonstrated the roles of alkaloids from lotus plumules, especially neferine in the treatment of different types of cancer. Neferine is a common chemosensitizer of vincristine that enhances the anti-tumor effect of vincristine by inhibiting gastric cancer cell proliferation (SGC7901) leading to their apoptosis. Neferine has also been shown to inhibit the growth of lung adenocarcinoma A549 cells, arrest their cell cycle in G1 phase, and further induce apoptosis through lipid peroxidation, depletion of the mitochondrial membrane potential, MAPKs activation, DNA degredation, and intracellular calcium accumulation [ 101] . In addition, neferine could cure liver cancer cells (HepG2) and breast cancer cells (MCF-7) by inhibiting their cell proliferation and growth through different mechanisms [ 102] . Chang et al. [ 103] reported that in addition to neferine, liensinine could also act as an anti-tumor agent by inhibiting normal mitochondrial energy supply, impairing lysosomal function, and inhibiting the growth of non-small cell lung cancer both in vitro and in vivo. In addition, liensinine could inhibit the growth of gastric cancer cells by increasing ROS levels and inhibiting the PI3K/AKT pathway [ 94] . Similarly, isoliensinine could cause apoptosis in HepG2, Huh-7, and H22 hepatocellular carcinoma (HCC) cells by decreasing NF-κB activity and constitutively phosphorylating NF-κB p65 subunit at Ser536 in HCC cells [ 97] . Overall, lotus seeds are now widely used in various cancer treatments due to their broad antagonistic properties.

      In addition to the pharmacological effects mentioned above, lotus BIAs possess substantial functions in protection against photoaging related skin problems [ 104] , reduced clinical manifestation of Alzheimer disorders [ 105] , suppression of CCl 4-induced liver damage, and bleomycin-induced pulmonary fibrosis [ 106, 107] . Other minor BIAs isolated from lotus, such as members of the 1-benzylisoquinolins type BIAs have also shown notable anti-HIV, anti-inflammatory, anti-arrhythmic, and anti-thrombotic properties [ 17] . Thus, lotus BIAs have significant potential for the development of novel drugs in treating various life-threatening human diseases, such as microbial infection, inflammation, atherosclerosis, cancer, obesity, neurological disorders, and diabetes.

    • As mentioned above, lotus BIAs are predominantly aporphines and bis-BIAs, which accumulate mainly in lotus leaves and plumules [ 11, 18] . Notable, 1-benzylisoquinolines, which are mostly biosynthetic intermediates and derivatives of the aporphines and bis-BIAs, have also been detected in lotus tissues in trace amounts. The biosynthetic pathways of lotus BIAs have been proposed based on their identified structures [ 12, 22, 29, 108] . Noteworthily, lotus BIAs primarily occur as R-enantiomers, which is contrary to the predominant S-enantiomers occurrence in the Ranunculales.

      Similar to other plant BIAs, the biosynthesis of lotus BIAs starts also from the L-amino acid tyrosine. Tyrosine is first converted into L-dopamine (L-DOPA) and 4-hydroxyphenylacetaldehyde (4-HPAA), respectively by the catalyzation of tyrosine decarboxylase (TYDC) [ 109] . L-DOPA and 4-HPAA are subsequently condensed by norcoclaurine synthase (NCS) into ( R)-norcoclaurine, the first BIA scaffold in plants [ 110] ( Fig. 8). Two additional enzymatic steps, catalyzed by norcoclaurine 6-O-methyltransferase (6OMT) and ( R)-coclaurine N-methyltransferase (CNMT) respectively, yield the core intermediate ( R)- N-methylcoclaurine of almost all BIAs [ 15] . In other BIA accumulating species, N-methylcoclaurine normally undergoes two further 3'-hydoxylation and 4'-Omethylation reactions, catalyzed by ( S)- N-methylcoclaurine 3'-hydroxylase (CYP80B) and 3'-hydroxy- N-methylcoclaurine 4'- O-methyltransferase (4'OMT), respectively, to yield ( S)-reticuline, which is another central precursor and a major branch point of numerous BIA structures, including morphine and berberine. Interestingly, neither reticuline nor corytuberine have been isolated from lotus tissues to date. In addition, all identified aporphine-type BIAs in lotus lack the hydroxyl and methyl modifications at the C-4' and C-3' positions, suggesting that the two enzymatic steps catalyzed by CYP80B and 4’OMT are not necessary for aporphine biosynthesis in lotus [ 22, 110] . Consistently, a recent study showed that pronuciferine is the lotus aporphine biosynthesis precursor, and it is synthesized from the core intermediate ( R)- N-methylcoclaurine by the enzymatic actions of NnCYP80G and 4- O-methyltransferase (7OMT) [ 12] ( Fig. 8). According to the lotus BIA structures, production of nuciferine from the pronuciferine precursor may arise from an unknown dehydration reaction [ 18] . Additional modifications, such as N-demethylation, O-demethylation, and methylenedioxy-bridge formation, catalyzed by the N-demethlase (NDM), O-demethlase (NDM), and CYP719A, respectively [ 22, 111] , transforms nuciferine into other diverse aporphines ( Fig. 8).

      Figure 8. 

      Proposed BIAs biosynthetic pathway in lotus. Steps marked with red, green, and purple background represent the common reactions for the biosynthesis of (R)-N-Methylcoclaurine, the lotus aporphine branch, and the bis-BIA biosynthesis branch, respectively. All biosynthetic enzymes are shown in red. The (R)-N-Methylcoclaurine is the branch point for aporphine and bis-BIA biosynthesis in lotus. Dotted arrows indicate multiple enzymatic or unknown steps.

      The lotus bis-BIAs can also directly be synthesized from the ( R)- N-methylcoclaurine precursor ( Fig. 8). The 7-O3' coupling reaction, catalyzed by CYP80A enzyme, transfers two molecules of ( R)- N-methylcoclaurine into Nelumboferine, which is the first bis-BIAs in the bis-BIA biosynthesis pathway [ 12, 108] . Two further enzymatic steps, catalyzed by the 4'OMT and 7OMT enzymes respectively, yield the three major bis-BIAs, including liensinine, isoliensinine, and neferine. Other enzymatic modifications, including N-demethylation, double 4'-Omethylation, 8-O3' and 3'-3' intermolecular couplings, result in the production of other diverse bis-BIA structures ( Fig. 4).

    • The currently advanced next generation sequencing techniques have facilitated the availability of at least seven high quality lotus genome versions as references for isolation of BIAs biosynthetic genes [ 2, 112115] . The previously functionally characterized BIAs pathway genes in the Ranunculales [ 116] have been retrieved and used to query and predict most of their corresponding homologs from the lotus genomic and transcriptomic data based on sequence similarities. As a result, at least 5, 4, and 1 TYDC, NCS, and CNMT candidate gene copies, respectively have been predicted in the early lotus BIA biosynthesis pathway [ 22] . Similarly, seven OMT genes have been identified, with four and three of which forming clusters with the 6OMT/4’OMT and 7OMT clades, respectively. In addition, downstream lotus BIA biosynthetic gene sequences, including CYP80A, CYP80G, CYP719A, NDM, and ODM have been isolated [ 22, 29] .

      The lack of stable lotus transformation system has hindered functional characterization of most its BIA pathway enzymes. Consequently, functional studies have mostly involved correlation analysis between gene expression levels and spatial temporal BIA accumulation patterns in different lotus tissues or developmental stages [ 22, 29, 117, 118] . In vitro enzymatic assays were recently used to functionally characterize four lotus OMTs, with NnOMT1 exhibiting 6OMT activity and accepting both S- and R-substrates, while NnOMT5 mainly showing 7- O-methyltransferase activity with strong S-enantiomer stereospecific preference [ 119] . In contrast, Nn6OMT and Nn7OMT have both been shown to display 6-O and 7-O methyltransferase activities [ 108, 120] . Notably, none of these NnOMTs accepted aporphine substrates, indicating that O-methylation reactions proceed primarily from 1-benzylisoquinoline intermediates.

      The core intermediate ( R)- N-Methylcoclaurine is the lotus BIA biosynthesis branch point, from which intermolecular C-O phenol coupling reaction catalyzed by NnCYP80A produces diverse bis-BIAs, while the intramolecular C-C phenol coupling reaction catalyzed by NnCYP80G yields aporphines ( Fig. 8). Two recent studies [ 12, 108] have functionally characterized the enzymes responsible for the lotus CYP80A and CYP80G members. The expression of NnCYP80G in yeast could efficiently convert the ( R)- N-Methylcoclaurine substrate into a pronuciferine glaziovine in lotus, while in the presence of NnOMT5, glaziovine could efficiently be transferred into pronuciferine [ 12] . Similarly, the expression of NnCYP80A augmented with CPR and CYB5 in yeast could catalyze the production of nelumboferine from ( R)- N-Methylcoclaurine substrate [ 108] . In addition, NnCYP719A, which is the third P450 in lotus could efficiently convert caaverine and lirinidine substrates into anonaine and roemerine, respectively.

      Although the sequences of other lotus BIA biosynthetic genes, such as NnTYDCs, NnNCSs, NnCNMT, NnODMs, and NnNDMs have already been isolated based on similarities with homologs from other BIA producing species, they are still yet to be functionally validated. Since none of the isolated lotus NCSs exhibit NCS activity, a non-enzymatic Pictet-Spengler condensation of dopamine and 4-HPAA might yield norcoclaurine in lotus [ 108] . However, since both R and S enantiomers of norcoclaurine have been detected in lotus, it is unlikely to entirely rule out the possibility of additional enzyme requirement for the Pictet-Spengler condensation and the formation of ( S)-norcoclaurine in lotus, as well as a second enzyme for stereochemical inversion of ( S)-norcoclaurine to ( R)-norcoclaurine.

      Unlike their characterization, little knowledge is still available on the regulation of lotus BIA genes. A previous comparative transcriptomic analysis of high and low BIA accumulating lotus cultivars showed that the expression levels of most BIA biosynthetic genes were significantly higher in the former than the latter cultivar [ 22] , suggesting transcriptional regulation of lotus BIA biosynthesis. Correlation analysis between gene expression profiles and BIA contents revealed 16 candidate TFs, such as WRKYs, MYBs, ERFs, and bHLHs that potentially regulate lotus BIAs biosynthesis. As an important group of secondary metabolites, lotus BIA biosynthesis is regulated by mechanical wounding and jasmonate (JA) treatment [ 11, 121] . To date, the only two functionally characterized BIA regulator TFs are NnWRKY70a and NnWRKY70b, which both belong to the group III WRKY TFs and are JA responsive. Overexpression of NnWRKY70a and NnWRKY70b in the lotus petals significantly enhanced BIA biosynthesis. Functional validation assays showed that NnWRKY70a and NnWRKY70b could directly bind and activate one or more BIA biosynthetic gene promoters, while NnWRKY70b could physically interact with NnJAZ1 and two other WRKY TFs (NnWRKY53b and NnWRKY70a), thus suggesting its potential interaction with other WRKY TFs to regulate lotus BIA biosynthesis via the JA signaling pathway [ 122] .

    • Lotus contains a vast number of BIAs, most of which are R-conformers. Aporphines and bisbenzylisoquinoline alkaloids are the two major lotus BIAs types, and are predominantly accumulated in the leaves and plumules, respectively. The lotus BIAs are generally weak alkalescent compounds with poor solubility or insoluble properties in water, but soluble in organic solvents. The crude lotus BIAs are commonly extracted with ethanol or methanol solvents, assisted with reflux, sonication, and microwave techniques. Critical fluid extraction and high-speed counter-current chromatography are relatively greener, energy-efficient, time- and solvent-saving, as well as high yielding BIA extraction techniques. The UPLC-Tof-MS is currently the most popular method for BIA isolation, characterization, and quantification. Lotus BIAs harbors significant medicinal properties with potential application in the treatment of various life-threatening diseases, such as obesity, inflammation, cancer, HIV, and aniocardiopathy.

      The availability of high-quality lotus genome sequences have facilitated the isolation of most genes or enzymes involved in lotus BIA biosynthesis and regulation. However, functional characterization specifically through protein expression in engineered yeast strain and downstream enzyme activity assays have only been successfully conducted for NnOMTs, NnCYP80A, and NnCYP80G. Future intensive work on functional characterization of putative genes and enzymes is needed to fully elucidate the lotus BIA biosynthetic pathways. In addition, although the tissue specific accumulation of lotus BIAs has been clarified, the exact cell types involved in lotus biosynthesis and storage remain largely unknown. Combining cutting edge spatial metabolomics with single-cell RNA sequencing would not only give valuable clues on the localization of BIAs and their biosynthetic enzymes, but also facilitate the isolation of BIA biosynthesis regulators as well as possible intermediate transporters. The unique stereochemistry of BIAs in this basal eudicot species is equally worthy of research both at the molecular and biochemical levels.

      The ultimate aim studying lotus BIAs is of course to develop clinical drugs based on these molecules. There is no doubt that BIAs in lotus bear significant pharmacological activities. It should be noticed, however, unmodified lotus BIAs possess suboptimal efficiency in absorption, metabolism, excretion and toxicity properties. Research should be directed on total chemical synthesis, structural modifications, creation of BIA hybrids or new BIA analogs, with an aim to develop lotus BIA-based novel drugs for treatment of degenerative diseases, cancer, and the continuous threat of novel infections like COVID-19.

    • The authors confirm contribution to the paper as follows: study design: Li J, Deng X; wrote the manuscript: Wei X, Deng X; revised the manuscript: Zhang M, Ogutu C, Yang M. All authors reviewed the results and approved the final version of the manuscript.

    • The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

      • This project was supported by funds received from the National Natural Science Foundation of China (Grant no. 32070336 and 32370428) and the Natural Science Foundation of Shandong Province (Grant no. ZR2021MC163).

      • The authors declare that they have no conflict of interest.

      • Copyright: © 2024 by the author(s). Published by Maximum Academic Press, Fayetteville, GA. This article is an open access article distributed under Creative Commons Attribution License (CC BY 4.0), visit https://creativecommons.org/licenses/by/4.0/.
    Figure (8)  Table (4) References (122)
  • About this article
    Cite this article
    Wei X, Zhang M, Yang M, Ogutu C, Li J, et al. 2024. Lotus (Nelumbo nucifera) benzylisoquinoline alkaloids: advances in chemical profiling, extraction methods, pharmacological activities, and biosynthetic elucidation. Vegetable Research 4: e005 doi: 10.48130/vegres-0024-0004
    Wei X, Zhang M, Yang M, Ogutu C, Li J, et al. 2024. Lotus ( Nelumbo nucifera) benzylisoquinoline alkaloids: advances in chemical profiling, extraction methods, pharmacological activities, and biosynthetic elucidation. Vegetable Research 4: e005 doi: 10.48130/vegres-0024-0004

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return