Structure-activity relationship, bioactivities, molecular mechanisms, and clinical application of nuciferine on inflammation-related diseases

Pharmacological Research - Tập 193 - Trang 106820 - 2023
Tong Zhao1, Yuchen Zhu1, Rui Zhao1, Shiyi Xiong1, Jing Sun1, Juntao Zhang1, Daidi Fan2, Jianjun Deng3,2, Haixia Yang1
1College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
2Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi’an, China
3State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China

Tài liệu tham khảo

Guo, 2015, Inflammasomes: mechanism of action, role in disease, and therapeutics, Nat. Med, 21, 677, 10.1038/nm.3893 Broderick, 2015, The inflammasomes and autoinflammatory syndromes, Annu Rev. Pathol., 10, 395, 10.1146/annurev-pathol-012414-040431 Markiewski, 2007, The role of complement in inflammatory diseases from behind the scenes into the spotlight, Am. J. Pathol., 171, 715, 10.2353/ajpath.2007.070166 Ferguson, 2010, Chronic inflammation and mutagenesis, Mutat. Res, 690, 3, 10.1016/j.mrfmmm.2010.03.007 GBD, 2017, Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017, Lancet 2018;392:1736–88 Strowig, 2012, Inflammasomes in health and disease, Nature, 481, 278, 10.1038/nature10759 Bordoni, 2017, Dairy products and inflammation: a review of the clinical evidence, Crit. Rev. Food Sci. Nutr., 57, 2497, 10.1080/10408398.2014.967385 Liu, 2018, Monomeric catechin and dimeric procyanidin B2 against human norovirus surrogates and their physicochemical interactions, Food Microbiol., 76, 346, 10.1016/j.fm.2018.06.009 Qu, 2019, Kiwifruit seed oil ameliorates inflammation and hepatic fat metabolism in high-fat diet-induced obese mice, J. Funct. Foods, 52, 715, 10.1016/j.jff.2018.12.003 Ren Z., Yang H., Zhu C., Fan D., Deng J. Dietary phytochemicals: As a potential natural source for treatment of alzheimer’s disease. Alzheimer’s Disease 2023. Bharathi Priya, 2021, An updated review on pharmacological properties of neferine-A bisbenzylisoquinoline alkaloid from Nelumbo nucifera, J. Food Biochem, 45, 10.1111/jfbc.13986 Ling, 2005, Isolation, characterization, and determination of antioxidative activity of oligomeric procyanidins from the seedpod of Nelumbo nucifera Gaertn, J. Agric. Food Chem., 53, 2441, 10.1021/jf040325p Sharma, 2017, A comprehensive review on chemical profiling of Nelumbo nucifera: Potential for drug development: phytochemical profiling of lotus, Phytother. Res, 31, 3, 10.1002/ptr.5732 Kashiwada, 2005, Anti-HIV benzylisoquinoline alkaloids and flavonoids from the leaves of Nelumbo nucifera, and structure–activity correlations with related alkaloids, Bioorg. Med. Chem., 13, 443, 10.1016/j.bmc.2004.10.020 Bai, 2022, Nuciferine inhibits TMEM16A in dietary adjuvant therapy for lung cancer, J. Agric. Food Chem., 70, 3687, 10.1021/acs.jafc.1c08375 Sun, 2022, Nuciferine protects against high-fat diet-induced hepatic steatosis via modulation of gut microbiota and bile acid metabolism in rats, J. Agric. Food Chem., 70, 12014, 10.1021/acs.jafc.2c04817 Huang, 2022, Chemistry and biology of nuciferine, Ind. Crops Prod., 179, 10.1016/j.indcrop.2022.114694 Yu, 2021, Akkermansia muciniphila: a potential novel mechanism of nuciferine to improve hyperlipidemia, Biomed. Pharm., 133, 10.1016/j.biopha.2020.111014 Wan, 2022, Nuciferine, an active ingredient derived from lotus leaf, lights up the way for the potential treatment of obesity and obesity-related diseases, Pharm. Res, 175, 10.1016/j.phrs.2021.106002 Boustie, 1998, Antipoliovirus structure-activity relationships of some aporphine alkaloids, J. Nat. Prod., 61, 480, 10.1021/np970382v Yang, 2012, An aporphine alkaloid from Nelumbo nucifera as an acetylcholinesterase inhibitor and the primary investigation for structure–activity correlations, Nat. Prod. Res., 26, 387, 10.1080/14786419.2010.487188 Yang, 2014, Synthesis and structure–activity relationship of nuciferine derivatives as potential acetylcholinesterase inhibitors, Med Chem. Res, 23, 3178, 10.1007/s00044-013-0905-9 Deng, 2020, π-π stacking interactions: non-negligible forces for stabilizing porous supramolecular frameworks, Sci. Adv., 6 Nakamura, 2013, Alkaloid constituents from flower buds and leaves of sacred lotus (Nelumbo nucifera, Nymphaeaceae) with melanogenesis inhibitory activity in B16 melanoma cells, Bioorg. Med Chem., 21, 779, 10.1016/j.bmc.2012.11.038 Yang, 2022, Design, synthesis and structure-activity relationship optimization of phenanthridine derivatives as new anti-vitiligo compounds, Bioorg. Chem., 119, 10.1016/j.bioorg.2021.105582 Lin, 2014, Anthelmintic activities of aporphine from Nelumbo nucifera Gaertn. cv. Rosa-plena against hymenolepis nana, IJMS, 15, 3624, 10.3390/ijms15033624 Agnihotri, 2008, Constituents of Nelumbo nucifera leaves and their antimalarial and antifungal activity, Phytochem Lett., 1, 89, 10.1016/j.phytol.2008.03.003 Heng, 2018, Synthesis and evaluation of nuciferine and roemerine enantiomers as 5-HT 2 and α 1 receptor antagonists, Med Chem. Commun., 9, 576, 10.1039/C7MD00629B Munusamy, 2013, Structure-based identification of aporphines with selective 5-HT(2A) receptor-binding activity, Chem. Biol. Drug Des., 81, 250, 10.1111/cbdd.12069 Vu, 2022, Structures and antiosteoclastogenic activity of compounds isolated from edible lotus (Nelumbo nucifera Gaertn.) leaves and stems, Fitoterapia, 162, 10.1016/j.fitote.2022.105294 Takashima, 2022, Structure-activity relationship study of 4,5-didehydroguadiscine, an aporphine alkaloid showing potent melanogenesis-inhibitory activity in B16 melanoma cells, Bioorg. Med Chem. Lett., 78, 10.1016/j.bmcl.2022.129034 Gregor, 2011, Inflammatory mechanisms in obesity, Annu Rev. Immunol., 29, 415, 10.1146/annurev-immunol-031210-101322 Ohashi, 2014, Role of anti-inflammatory adipokines in obesity-related diseases, Trends Endocrinol. Metab., 25, 348, 10.1016/j.tem.2014.03.009 Pellegrinelli, 2016, Adipose tissue plasticity: How fat depots respond differently to pathophysiological cues, Diabetologia, 59, 1075, 10.1007/s00125-016-3933-4 Klöting, 2010, Insulin-sensitive obesity, Am. J. Physiol. Endocrinol. Metab., 299, E506, 10.1152/ajpendo.00586.2009 Bassaganya-Riera, 2009, PPAR gamma is highly expressed in F4/80(hi) adipose tissue macrophages and dampens adipose-tissue inflammation, Cell Immunol., 258, 138, 10.1016/j.cellimm.2009.04.003 Guo, 2013, Nuciferine prevents hepatic steatosis and injury induced by a high-fat diet in hamsters, PLoS ONE, 8 Zhang, 2015, Nuciferine downregulates Per-Arnt-Sim kinase expression during its alleviation of lipogenesis and inflammation on oleic acid-induced hepatic steatosis in HepG2 cells, Front Pharm., 6, 238, 10.3389/fphar.2015.00238 Wang, 2020, Nuciferine modulates the gut microbiota and prevents obesity in high-fat diet-fed rats, Exp. Mol. Med, 52, 1959, 10.1038/s12276-020-00534-2 Bursill, 2006, Modulation of cholesterol metabolism by the green tea polyphenol (-)-epigallocatechin gallate in cultured human liver (HepG2) cells, J. Agric. Food Chem., 54, 1621, 10.1021/jf051736o Campia, 2009, Digoxin and ouabain increase the synthesis of cholesterol in human liver cells, Cell Mol. Life Sci., 66, 1580, 10.1007/s00018-009-9018-5 Kong, 2004, Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins, Nat. Med, 10, 1344, 10.1038/nm1135 Liu, 2020, Inhibitory kinetics and bioactivities of Nuciferine and methyl ganoderate on mucor miehei lipase and 3T3-L1 preadipocytes, Int. J. Biol. Macromol., 10 Zhou, 2020, Nuciferine reduced fat deposition by controlling triglyceride and cholesterol concentration in broiler chickens, Poult. Sci., 99, 7101, 10.1016/j.psj.2020.09.013 Xiong, 2021, [Effect of nuciferine on gut microbiota and inflammatory response in obese model mice], Zhongguo Zhong Yao Za Zhi, 46, 2104 Shi, 2021, Nuciferine improves high-fat diet-induced obesity via reducing intestinal permeability by increasing autophagy and remodeling the gut microbiota, Food Funct., 12, 5850, 10.1039/D1FO00367D Ying, 2020, The role of macrophages in obesity-associated islet inflammation and β-cell abnormalities, Nat. Rev. Endocrinol., 16, 81, 10.1038/s41574-019-0286-3 Lee, 2018, An integrated view of immunometabolism, Cell, 172, 22, 10.1016/j.cell.2017.12.025 Eguchi, 2017, Islet inflammation in type 2 diabetes and physiology, J. Clin. Invest, 127, 14, 10.1172/JCI88877 Sun, 2021, Expression of miRNA-29 in pancreatic β cells promotes inflammation and diabetes via TRAF3, Cell Rep., 34, 10.1016/j.celrep.2020.108576 Donath, 2011, Type 2 diabetes as an inflammatory disease, Nat. Rev. Immunol., 11, 98, 10.1038/nri2925 He, 2021, Mitophagy-mediated adipose inflammation contributes to type 2 diabetes with hepatic insulin resistance, J. Exp. Med, 218, 10.1084/jem.20201416 Li, 2018, Identify super quality markers from prototype-based pharmacokinetic markers of Tangzhiqing tablet (TZQ) based on in vitro dissolution/ permeation and in vivo absorption correlations, Phytomedicine, 45, 59, 10.1016/j.phymed.2018.04.001 Chen, 2022, Integrated pyroptosis measurement and metabolomics to elucidate the effect and mechanism of tangzhiqing on atherosclerosis, Front Physiol., 13, 10.3389/fphys.2022.937737 Li, 2018, Nuciferine and paeoniflorin can be quality markers of Tangzhiqing tablet, a Chinese traditional patent medicine, based on the qualitative, quantitative and dose-exposure-response analysis, Phytomedicine, 44, 155, 10.1016/j.phymed.2018.02.006 Li, 2020, Altering the inhibitory kinetics and molecular conformation of maltase by Tangzhiqing (TZQ), a natural α-glucosidase inhibitor, BMC Complement Med Ther., 20, 350, 10.1186/s12906-020-03156-3 Li, 2020, Alleviating the hydrolysis of carbohydrates, Tangzhiqing (TZQ) decreased the postprandial glycemia in healthy volunteers: an eight-period crossover study, Evid. Based Complement Altern. Med, 2020, 8138195 Hao, 2020, Docking studies on potential mechanisms for decreasing insulin resistance by the Tangzhiqing herbal formula, Evid. Based Complement Altern. Med, 2020, 1057648, 10.1155/2020/1057648 Liu, 2018, Metabolomics-based clinical efficacy and effect on the endogenous metabolites of Tangzhiqing tablet, a Chinese patent medicine for type 2 diabetes mellitus with hypertriglyceridemia, Evid. Based Complement Altern. Med, 2018, 5490491, 10.1155/2018/5490491 Yan, 2017, Lotus leaf aqueous extract reduces visceral fat mass and ameliorates insulin resistance in HFD-induced obese rats by regulating PPARγ2 expression, Front Pharm., 8, 409, 10.3389/fphar.2017.00409 Zhang, 2018, Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through a PPARα/PPARγ coactivator-1α pathway, Br. J. Pharm., 175, 4218, 10.1111/bph.14482 Zhou, 2009, Hypoglycemic hypolipidemic Eff. flavonoids lotus (Nelumbo nuficera Gaertn) leaf Diabet. mice, 4 Kim, 2013, Lotus leaf alleviates hyperglycemia and dyslipidemia in animal model of diabetes mellitus, Nutr. Res Pr., 7, 166, 10.4162/nrp.2013.7.3.166 Zheng, 2018, Global aetiology and epidemiology of type 2 diabetes mellitus and its complications, Nat. Rev. Endocrinol., 14, 88, 10.1038/nrendo.2017.151 Tang, 2021, Nuciferine administration in C57BL/6J mice with gestational diabetes mellitus induced by a high-fat diet: the improvement of glycolipid disorders and intestinal dysbacteriosis, Food Funct., 12, 11174, 10.1039/D1FO02714J Wang, 2020, PPARs as metabolic regulators in the liver: lessons from liver-specific PPAR-null mice, Int J. Mol. Sci., 21 Li, 2018, Co-administration of nuciferine reduces the concentration of metformin in liver via differential inhibition of hepatic drug transporter OCT1 and MATE1: nuciferine differentially inhibits hepatic OCT1 and MATE1, Biopharm. Drug Dispos., 39, 411, 10.1002/bdd.2158 Kuper, 2000, Infections as a major preventable cause of human cancer, J. Intern Med, 248, 171, 10.1046/j.1365-2796.2000.00742.x Coussens, 2002, Inflammation and cancer, Nature, 420, 860, 10.1038/nature01322 Zheng, 2022, The pharmacological mechanisms of Xiaochaihutang in treating breast cancer based on network pharmacology, Contrast Media Mol. Imaging, 2022, 3900636, 10.1155/2022/3900636 Shao, 2021, Xiao-Chai-Hu-Tang ameliorates tumor growth in cancer comorbid depressive symptoms via modulating gut microbiota-mediated TLR4/MyD88/NF-κB signaling pathway, Phytomedicine, 88, 10.1016/j.phymed.2021.153606 Zhou, 2019, YAP inhibition by nuciferine via AMPK-mediated downregulation of HMGCR sensitizes pancreatic cancer cells to gemcitabine, Biomolecules, 9, 10.3390/biom9100620 Liu, 2020, A multiple-targets alkaloid nuciferine overcomes paclitaxel-induced drug resistance in vitro and in vivo, Phytomedicine, 79, 10.1016/j.phymed.2020.153342 Liu, 2015, Nuciferine, extracted from Nelumbo nucifera Gaertn, inhibits tumor-promoting effect of nicotine involving Wnt/β-catenin signaling in non-small cell lung cancer, J. Ethnopharmacol., 165, 83, 10.1016/j.jep.2015.02.015 Qi, 2016, Identification of the anti-tumor activity and mechanisms of nuciferine through a network pharmacology approach, Acta Pharm. Sin., 37, 963, 10.1038/aps.2016.53 Xu, 2020, Nuciferine inhibits skin cutaneous melanoma cell growth by suppressing TLR4/NF-κB signaling, Anticancer Agents Med Chem., 20, 2099, 10.2174/1871520620666200811114607 Wu, 2015, 7-Hydroxydehydronuciferine induces human melanoma death via triggering autophagy and apoptosis, Exp. Dermatol., 24, 930, 10.1111/exd.12805 Wu, 2018, Isolation and purification of alkaloids from lotus leaves by ionic-liquid-modified high-speed countercurrent chromatography, J. Sep Sci., 41, 571, 10.1002/jssc.201700851 Li, 2021, Nuciferine protects against folic acid-induced acute kidney injury by inhibiting ferroptosis, Br. J. Pharm., 178, 1182, 10.1111/bph.15364 Kang, 2017, Liensinine and nuciferine, bioactive components of Nelumbo nucifera, inhibit the growth of breast cancer cells and breast cancer-associated bone loss, Evid. Based Complement Altern. Med, 2017, 1583185, 10.1155/2017/1583185 Abdelmalek, 2021, Nonalcoholic fatty liver disease: another leap forward, Nat. Rev. Gastroenterol. Hepatol., 18, 85, 10.1038/s41575-020-00406-0 Angulo, 2015, Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease, Gastroenterology, 149 Younossi, 2019, The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis, J. Hepatol., 71, 793, 10.1016/j.jhep.2019.06.021 Yin, 2014, Hugan Qingzhi medication ameliorates hepatic steatosis by activating AMPK and PPARα pathways in L02 cells and HepG2 cells, J. Ethnopharmacol., 154, 229, 10.1016/j.jep.2014.04.011 Cui, 2020, Untargeted metabolomic analysis of the effects and mechanism of nuciferine treatment on rats with nonalcoholic fatty liver disease, Front Pharm., 11, 858, 10.3389/fphar.2020.00858 Kim, 2017, Hederagenin supplementation alleviates the pro-inflammatory and apoptotic response to alcohol in rats, Nutrients, 9 Lu, 2018, Alleviating acute alcoholic liver injury in mice with Bacillus subtilis co-expressing alcohol dehydrogenase and acetaldehyde dehydrogenase, J. Funct. Foods, 49, 342, 10.1016/j.jff.2018.09.006 Shu, 2019, Nuciferine alleviates acute alcohol-induced liver injury in mice: roles of suppressing hepatic oxidative stress and inflammation via modulating miR-144/Nrf2/HO-1 cascade, J. Funct. Foods, 58, 105, 10.1016/j.jff.2019.04.055 Brunner, 2005, Endothelial function and dysfunction. Part II: association with cardiovascular risk factors and diseases. A statement by the Working Group on Endothelins and Endothelial Factors of the European Society of Hypertension, J. Hypertens., 23, 233, 10.1097/00004872-200502000-00001 Harishkumar, 2021, Nuciferine attenuates doxorubicin-induced cardiotoxicity: an in vitro and in vivo study, Cardiovasc Toxicol., 21, 947, 10.1007/s12012-021-09689-4 Xiao, 2014, New paradigms in inflammatory signaling in vascular endothelial cells, Am. J. Physiol. Heart Circ. Physiol., 306, H317, 10.1152/ajpheart.00182.2013 Wang, 2015, Nuciferine relaxes rat mesenteric arteries through endothelium-dependent and -independent mechanisms, Br. J. Pharm., 172, 5609, 10.1111/bph.13021 Go, 2014, Heart disease and stroke statistics—2014 update: a report from the American heart association, Circulation, 129 Grabowska-Fudala, 2018, Depressive symptoms in stroke patients treated and non-treated with intravenous thrombolytic therapy: a 1-year follow-up study, J. Neurol., 265, 1891, 10.1007/s00415-018-8938-0 Wu, 2020, UPLC-Q-TOF/MS-based serum metabolomics reveals the anti-ischemic stroke mechanism of nuciferine in MCAO rats, ACS Omega, 5, 33433, 10.1021/acsomega.0c05388 Chen, 2022, Nuciferine attenuates acute ischemic stroke in a rat model: a metabolomic approach for the mechanistic study, Mol. Omics, 18, 765, 10.1039/D2MO00158F HarishKumar, 2022, Nuciferine from Nelumbo nucifera Gaertn. attenuates isoproterenol-induced myocardial infarction in Wistar rats, Biotechnol. Appl. Biochem, 69, 1176, 10.1002/bab.2194 Su, 2020, Research advances in the mechanisms of hyperuricemia-induced renal injury, Biomed. Res Int, 2020, 5817348, 10.1155/2020/5817348 Wang, 2015, Nuciferine restores potassium oxonate-induced hyperuricemia and kidney inflammation in mice, Eur. J. Pharm., 747, 59, 10.1016/j.ejphar.2014.11.035 Wang, 2016, Nuciferine alleviates renal injury by inhibiting inflammatory responses in fructose-fed rats, J. Agric. Food Chem., 64, 7899, 10.1021/acs.jafc.6b03031 Wang, 2020, 1H NMR and UHPLC/Q-Orbitrap-MS-based metabolomics combined with 16S rRNA gut microbiota analysis revealed the potential regulation mechanism of nuciferine in hyperuricemia rats, J. Agric. Food Chem., 68, 14059, 10.1021/acs.jafc.0c04985 Dixon, 2012, Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell, 149, 1060, 10.1016/j.cell.2012.03.042 Stockwell, 2017, Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease, Cell, 171, 273, 10.1016/j.cell.2017.09.021 Peng, 2020, Involvement of regulated necrosis in blinding diseases: focus on necroptosis and ferroptosis, Exp. Eye Res., 191, 10.1016/j.exer.2020.107922 Song, 2020, Nuciferine prevents bone loss by disrupting multinucleated osteoclast formation and promoting type H vessel formation, FASEB J., 34, 4798, 10.1096/fj.201902551R Wang, 2023, Network pharmacology and pharmacological evaluation for deciphering novel indication of Sishen Wan in insomnia treatment, Phytomedicine, 108, 10.1016/j.phymed.2022.154500 Su, 2013, Curative effect of warming kidney and fortifying spleen recipe on diarrhea-predominant irritable bowel syndrome, J. Tradit. Chin. Med., 33, 615, 10.1016/S0254-6272(14)60030-3 Xu, 2021, Simultaneous determination of ten bioactive components from Shenling Baizhu San in rat plasma by UHPLC-MS/MS: application to a comparative pharmacokinetic study in normal and two models of ulcerative colitis rats, Evid. Based Complement Altern. Med., 2021, 3518241, 10.1155/2021/3518241 Wang, 2022, Herbal formula Shenling Baizhu San for chronic diarrhea in adults: a systematic review and meta-analysis, Integr. Cancer Ther., 21, 10.1177/15347354221081214 Jiao, 2022, Shenling Baizhu San ameliorates ulcerative colitis by regulating the gut microbiota and its tryptophan metabolites: a complementary medicine to mesalamine, J. Ethnopharmacol., 291, 10.1016/j.jep.2022.115145 Cai, 2018, The role of diallyl thiosulfinate associated with nuciferine and diosgenin in the treatment of premature ejaculation: a pilot study, Arch. Ital. Urol. Androl., 90, 59, 10.4081/aiua.2018.1.59 Plotti, 2023, Role of protopine and nuciferine in the management of nocturia, urgency, and dysuria in pre- and postmenopausal women: a prospective pilot study, Gynecol. Obstet. Invest, 88, 47, 10.1159/000528509 Zhang, 2021, Screening of hypolipidemic active components in Jiang-Zhi-Ning and its preliminary mechanism research based on “active contribution value” study, J. Ethnopharmacol., 272, 10.1016/j.jep.2021.113926 Chen, 2012, The active ingredients of Jiang-Zhi-Ning: study of the Nelumbo nucifera alkaloids and their main bioactive metabolites, Molecules, 17, 9855, 10.3390/molecules17089855 Chen, 2018, Nuciferine alleviates LPS-induced mastitis in mice via suppressing the TLR4-NF-κB signaling pathway, Inflamm. Res, 67, 903, 10.1007/s00011-018-1183-2 Wu, 2017, Nuciferine ameliorates inflammatory responses by inhibiting the TLR4-mediated pathway in lipopolysaccharide-induced acute lung injury, Front Pharm., 8, 939, 10.3389/fphar.2017.00939 Du, 2022, Nuciferine protects against high-fat diet-induced hepatic steatosis and insulin resistance via activating TFEB-mediated autophagy-lysosomal pathway, Acta Pharm. Sin. B, 12, 2869, 10.1016/j.apsb.2021.12.012 Wen, 2021, Nuciferine attenuates the progression of osteoarthritis by targeting PI3K/Akt/NF-κB signaling pathway, J. Funct. Foods, 86, 10.1016/j.jff.2021.104682 Fan, 2022, Nuciferine prevents hepatic steatosis associated with improving intestinal mucosal integrity, mucus-related microbiota and inhibiting TLR4/MyD88/NF-κB pathway in high-fat induced rats, J. Funct. Foods, 88, 10.1016/j.jff.2021.104859 Hemmings, 2015, The PI3K-PKB/Akt pathway, Cold Spring Harb. Perspect. Biol., 7, a026609, 10.1101/cshperspect.a026609 Brazil, 2004, Advances in protein kinase B signalling: AKTion on multiple fronts, Trends Biochem Sci., 29, 233, 10.1016/j.tibs.2004.03.006 Missiroli, 2009, Nuclear translocation of active AKT is required for erythroid differentiation in erythropoietin treated K562 erythroleukemia cells, Int J. Biochem Cell Biol., 41, 570, 10.1016/j.biocel.2008.07.002 Xiao, 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, 10.1016/j.phymed.2022.154536 Kuriakose, 2016, ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways, Sci. Immunol., 1 Kanneganti, 2006, Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3, Nature, 440, 233, 10.1038/nature04517 Christgen, 2020, Toward targeting inflammasomes: insights into their regulation and activation, Cell Res., 30, 315, 10.1038/s41422-020-0295-8 Mathur, 2018, Molecular mechanisms of inflammasome signaling, J. Leukoc. Biol., 103, 233, 10.1189/jlb.3MR0617-250R Martinon, 2006, Gout-associated uric acid crystals activate the NALP3 inflammasome, Nature, 440, 237, 10.1038/nature04516 Fan, 2015, PPARs and ERRs: molecular mediators of mitochondrial metabolism, Curr. Opin. Cell Biol., 33, 49, 10.1016/j.ceb.2014.11.002 Mansouri, 2008, Systemic and distal repercussions of liver-specific peroxisome proliferator-activated receptor-alpha control of the acute-phase response, Endocrinology, 149, 3215, 10.1210/en.2007-1339 Shan, 2008, Ligand activation of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) attenuates carbon tetrachloride hepatotoxicity by downregulating proinflammatory gene expression, Toxicol. Sci., 105, 418, 10.1093/toxsci/kfn142 Zhang, 2018, Nuciferine inhibits LPS-induced inflammatory response in BV2 cells by activating PPAR-γ, Int Immunopharmacol., 63, 9, 10.1016/j.intimp.2018.07.015 Herzig, 2018, AMPK: guardian of metabolism and mitochondrial homeostasis, Nat. Rev. Mol. Cell Biol., 19, 121, 10.1038/nrm.2017.95 Long, 2006, AMP-activated protein kinase signaling in metabolic regulation, J. Clin. Invest., 116, 1776, 10.1172/JCI29044 Viollet, 2009, AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives, Acta Physiol., 196, 81, 10.1111/j.1748-1716.2009.01970.x Ma, 2015, Pronuciferine and nuciferine inhibit lipogenesis in 3T3-L1 adipocytes by activating the AMPK signaling pathway, Life Sci., 136, 120, 10.1016/j.lfs.2015.07.001 Xu, 2022, Distinct AMPK-mediated FAS/HSL pathway is implicated in the alleviating effect of nuciferine on obesity and hepatic steatosis in HFD-fed mice, Nutrients, 14, 1898, 10.3390/nu14091898 Li, 2023, Nuciferine ameliorates nonesterified fatty acid-induced bovine mammary epithelial cell lipid accumulation, apoptosis, and impaired migration via activating LKB1/AMPK signaling pathway, J. Agric. Food Chem., 71, 443, 10.1021/acs.jafc.2c06133 Kulhari, 2023, Nuciferine alleviates intestinal inflammation by inhibiting MAPK/NF-κB and NLRP3/Caspase 1 pathways in vivo and in vitro, Int. Immunopharmacol., 115, 10.1016/j.intimp.2022.109613 Kim, 2022, Nuciferine attenuates lipopolysaccharide-stimulated inflammatory responses by inhibiting p38 MAPK/ATF2 signaling pathways, Inflammopharmacology, 10.1007/s10787-022-01075-y Qi, 2020, Changes of gut microbiota and its correlation with short chain fatty acids and bioamine in piglets at the early growth stage, Front Vet. Sci., 7 Zhang, 2017, Interactions between intestinal microbiota and host immune response in inflammatory bowel disease, Front Immunol., 8, 942, 10.3389/fimmu.2017.00942 Hansen, 2012, Microbiota of de-novo pediatric IBD: increased Faecalibacterium prausnitzii and reduced bacterial diversity in Crohn’s but not in ulcerative colitis, Am. J. Gastroenterol., 107, 1913, 10.1038/ajg.2012.335 Blutt, 2012, IgA is important for clearance and critical for protection from rotavirus infection, Mucosal Immunol., 5, 712, 10.1038/mi.2012.51 Glauben, 2014, Histone deacetylase inhibitors modulate interleukin 6-dependent CD4+ T cell polarization in vitro and in vivo, J. Biol. Chem., 289, 6142, 10.1074/jbc.M113.517599 Zenewicz, 2009, CD4 T-cell differentiation and inflammatory bowel disease, Trends Mol. Med., 15, 199, 10.1016/j.molmed.2009.03.002 Zhu, 2022, Nuciferine regulates immune function and gut microbiota in DSS-induced ulcerative colitis, Front Vet. Sci., 9, 10.3389/fvets.2022.939377