Andrographolide, A Natural Antioxidant: An Update
Abstract
:1. Introduction
2. Andrographolide Bioavailability
3. Antioxidant Proprieties of Andrographolide
3.1. Reactive Oxygen Species (ROS) Scavenging
3.2. Protective Effects of Andrographolide on Mitochondria
3.3. Inhibition of Free Radical-Producing Enzymes by Andrographolide
3.3.1. NADPH Oxidase
3.3.2. Xanthine Oxidase
3.4. Antioxidant Protective Proprieties of Andrographolide in Activation of Antioxidant System
3.4.1. SOD, CAT, and GPx
3.4.2. In Vivo Studies
3.4.3. In Vitro Studies
3.5. Nrf2 Signaling Pathway: Andrographolide Regulation
3.5.1. In Vitro Studies
3.5.2. In Vivo Studies
- SOD activity increased in the liver, kidney, heart, and red blood cells;
- CAT activity increased in the heart;
- GSH peroxidase activity increased in the kidney;
- GSH reductase increased in the kidney, heart, and red blood cells;
- GSH S-transferase increased in the liver;
- GSH protein increased in the heart;
- antioxidant proteins (SOD1, GST Ya, GST Yb, HO-1, GCLC, and GCLM) increased in the liver, kidney, and heart;
- The mRNA of GCLC, GCLM, GST Ya/Yb, SOD1, and HO-1 increased in the liver and kidney.
4. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Lim, J.C.W.; Chan, T.K.; Ng, D.S.W.; Sagineedu, S.R.; Stanslas, J.; Wong, W.S.F. Andrographolide and its analogues: Versatile bioactive molecules for combating inflammation and cancer. Clin. Exp. Pharmacol. Physiol. 2012, 39, 300–310. [Google Scholar] [CrossRef] [PubMed]
- Okhuarobo, A.; Falodun, J.E.; Erharuyi, O.; Imieje, V.; Falodun, A.; Langer, P. Harnessing the medicinal properties of Andrographis paniculata for diseases and beyond: A review of its phytochemistry and pharmacology. Asian Pac. J. Trop. Dis. 2014, 4, 213–222. [Google Scholar] [CrossRef]
- Abu-Ghefreh, A.A.; Canatan, H.; Ezeamuzie, C.I. In vitro and in vivo anti-inflammatory effects of andrographolide. Int. Immunopharmacol. 2009, 9, 313–318. [Google Scholar] [CrossRef]
- Madav, S.; Tripathi, H.C.; Mishra, S.K. Analgesic, Antipyretic and Antiulcerogenic Effects of Andrographolide. Indian J. Pharm. Sci. 1995, 57, 121. [Google Scholar]
- Pokala, N.; Alasyam, N.; Rasamal, K. Evaluation and comparison of antipyretic activity of aqueous leaf extracts of Vitex negundo and Andrographis paniculata in rabbits. Natl. J. Physiol. Pharm. Pharmacol. 2019, 9, 556–561. [Google Scholar] [CrossRef]
- Pan, C.-W.; Yang, S.-X.; Pan, Z.-Z.; Zheng, B.; Wang, J.-Z.; Lu, G.-R.; Xue, Z.-X.; Xu, C.-L. Andrographolide ameliorates d-galactosamine/lipopolysaccharide-induced acute liver injury by activating Nrf2 signaling pathway. Oncotarget 2017, 8, 41202–41210. [Google Scholar] [CrossRef]
- Shukla, B.; Visen, P.K.; Patnaik, G.K.; Dhawan, B.N. Choleretic effect of andrographolide in rats and guinea pigs. Planta Med. 1992, 58, 146–149. [Google Scholar] [CrossRef]
- Pongtuluran, O.B.; Rofaani, E. Tarwadi Antiviral and Immunostimulant Activities of Andrographis paniculata. Hayati J. Biosci. 2015, 22, 67–72. [Google Scholar]
- Puri, A.; Saxena, R.; Saxena, R.P.; Saxena, K.C.; Srivastava, V.; Tandon, J.S. Immunostimulant agents from Andrographis paniculata. J. Nat. Prod. 1993, 56, 995–999. [Google Scholar] [CrossRef]
- Akbar, S. Andrographis paniculata: A review of pharmacological activities and clinical effects. Altern Med. Rev. 2011, 16, 66–77. [Google Scholar]
- Chakravarti, R.N.; Chakravarti, D. Andrographolide, the Active Constituent of Andrographis Paniculata Nees. A Preliminary Communication. Ind. Med. Gaz. 1951, 86, 96–97. [Google Scholar] [PubMed]
- Manjula, S.; Kalaiarasi, C.; Pavan, M.S.; Hathwar, V.R.; Kumaradhas, P. Charge density and electrostatic potential of hepatitis C anti-viral agent andrographolide: An experimental and theoretical study. Acta Cryst. B 2018, 74, 693–704. [Google Scholar] [CrossRef]
- Paemanee, A.; Hitakarun, A.; Wintachai, P.; Roytrakul, S.; Smith, D.R. A proteomic analysis of the anti-dengue virus activity of andrographolide. Biomed. Pharmacother. 2019, 109, 322–332. [Google Scholar] [CrossRef] [PubMed]
- Wintachai, P.; Kaur, P.; Lee, R.C.H.; Ramphan, S.; Kuadkitkan, A.; Wikan, N.; Ubol, S.; Roytrakul, S.; Chu, J.J.H.; Smith, D.R. Activity of andrographolide against chikungunya virus infection. Sci. Rep. 2015, 5, 14179. [Google Scholar] [CrossRef] [PubMed]
- Lu, W.J.; Lin, K.H.; Hsu, M.J.; Chou, D.S.; Hsiao, G.; Sheu, J.R. Suppression of NF-κB signaling by andrographolide with a novel mechanism in human platelets: Regulatory roles of the p38 MAPK-hydroxyl radical-ERK2 cascade. Biochem. Pharmacol. 2012, 84, 914–924. [Google Scholar] [CrossRef] [PubMed]
- Mussbacher, M.; Salzmann, M.; Brostjan, C.; Hoesel, B.; Schoergenhofer, C.; Datler, H.; Hohensinner, P.; Basílio, J.; Petzelbauer, P.; Assinger, A.; et al. Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front. Immunol. 2019, 10, 85. [Google Scholar] [CrossRef]
- Chua, L.S. Review on Liver Inflammation and Antiinflammatory Activity of Andrographis paniculata for Hepatoprotection. Phytother. Res. 2014, 28, 1589–1598. [Google Scholar] [CrossRef]
- Singha, P.K.; Roy, S.; Dey, S. Protective activity of andrographolide and arabinogalactan proteins from Andrographis paniculata Nees. against ethanol-induced toxicity in mice. J. Ethnopharmacol. 2007, 111, 13–21. [Google Scholar] [CrossRef]
- Khan, I.; Khan, F.; Farooqui, A.; Ansari, I.A. Andrographolide Exhibits Anticancer Potential Against Human Colon Cancer Cells by Inducing Cell Cycle Arrest and Programmed Cell Death via Augmentation of Intracellular Reactive Oxygen Species Level. Nutr. Cancer 2018, 70, 787–803. [Google Scholar] [CrossRef]
- Liao, H.-C.; Chou, Y.-J.; Lin, C.-C.; Liu, S.-H.; Oswita, A.; Huang, Y.-L.; Wang, Y.-L.; Syu, J.-L.; Sun, C.-M.; Leu, C.-M.; et al. Andrographolide and its potent derivative exhibit anticancer effects against imatinib-resistant chronic myeloid leukemia cells by downregulating the Bcr-Abl oncoprotein. Biochem. Pharmacol. 2019, 163, 308–320. [Google Scholar] [CrossRef]
- Tan, W.S.D.; Liao, W.; Zhou, S.; Wong, W.S.F. Is there a future for andrographolide to be an anti-inflammatory drug? Deciphering its major mechanisms of action. Biochem. Pharmacol. 2017, 139, 71–81. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Tang, D.; Wang, J.; Wei, H.; Gao, J. Neuroprotection of Andrographolide Against Microglia-Mediated Inflammatory Injury and Oxidative Damage in PC12 Neurons. Neurochem. Res. 2019. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Cao, F.; Zhu, L.-L.; Liu, P.; Shang, Y.-R.; Liu, W.-H.; Dong, X.; Bao, H.-D.; Gong, P.; Wang, Z.-Y. Andrographolide impairs alpha-naphthylisothiocyanate-induced cholestatic liver injury in vivo. J. Nat. Med. 2019, 73, 388–396. [Google Scholar] [CrossRef] [PubMed]
- Scalbert, A.; Williamson, G. Dietary Intake and Bioavailability of Polyphenols. J. Nutr. 2000, 130, S2073–S2085. [Google Scholar] [CrossRef]
- Chen, H.-W.; Huang, C.-S.; Li, C.-C.; Lin, A.-H.; Huang, Y.-J.; Wang, T.-S.; Yao, H.-T.; Lii, C.-K. Bioavailability of andrographolide and protection against carbon tetrachloride-induced oxidative damage in rats. Toxicol. Appl. Pharmacol. 2014, 280, 1–9. [Google Scholar] [CrossRef]
- Bera, R.; Ahmed, S.K.M.; Sarkar, L.; Sen, T.; Karmakar, S. Pharmacokinetic analysis and tissue distribution of andrographolide in rat by a validated LC-MS/MS method. Pharm. Biol. 2014, 52, 321–329. [Google Scholar] [CrossRef]
- Xu, L.; Xiao, D.; Lou, S.; Zou, J.; Zhu, Y.; Fan, H.; Wang, G. A simple and sensitive HPLC-ESI-MS/MS method for the determination of andrographolide in human plasma. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2009, 877, 502–506. [Google Scholar] [CrossRef]
- Sareer, O.; Ahmad, S.; Umar, S. Andrographis paniculata: A critical appraisal of extraction, isolation and quantification of andrographolide and other active constituents. Nat. Prod. Res. 2014, 28, 2081–2101. [Google Scholar] [CrossRef]
- Casamonti, M.; Risaliti, L.; Vanti, G.; Piazzini, V.; Bergonzi, M.C.; Bilia, A.R. Andrographolide Loaded in Micro- and Nano-Formulations: Improved Bioavailability, Target-Tissue Distribution, and Efficacy of the “King of Bitters”. Engineering 2019, 5, 69–75. [Google Scholar] [CrossRef]
- Yang, T.; Sheng, H.-H.; Feng, N.-P.; Wei, H.; Wang, Z.-T.; Wang, C.-H. Preparation of andrographolide-loaded solid lipid nanoparticles and their in vitro and in vivo evaluations: Characteristics, release, absorption, transports, pharmacokinetics, and antihyperlipidemic activity. J. Pharm. Sci. 2013, 102, 4414–4425. [Google Scholar] [CrossRef]
- Li, B.; Jiang, T.; Liu, H.; Miao, Z.; Fang, D.; Zheng, L.; Zhao, J. Andrographolide protects chondrocytes from oxidative stress injury by activation of the Keap1-Nrf2-Are signaling pathway. J. Cell. Physiol. 2018, 234, 561–571. [Google Scholar] [CrossRef] [PubMed]
- Tan, W.S.D.; Liao, W.; Peh, H.Y.; Vila, M.; Dong, J.; Shen, H.-M.; Wong, W.S.F. Andrographolide simultaneously augments Nrf2 antioxidant defense and facilitates autophagic flux blockade in cigarette smoke-exposed human bronchial epithelial cells. Toxicol. Appl. Pharmacol. 2018, 360, 120–130. [Google Scholar] [CrossRef] [PubMed]
- Yan, H.; Huang, Z.; Bai, Q.; Sheng, Y.; Hao, Z.; Wang, Z.; Ji, L. Natural product andrographolide alleviated APAP-induced liver fibrosis by activating Nrf2 antioxidant pathway. Toxicology 2018, 396–397, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. Oxidative stress: oxidants and antioxidants. Exp. Physiol. 1997, 82, 291–295. [Google Scholar] [CrossRef]
- Piconi, L.; Quagliaro, L.; Ceriello, A. Oxidative stress in diabetes. Clin. Chem. Lab. Med. 2003, 41, 1144–1149. [Google Scholar] [CrossRef]
- Singh, U.; Devaraj, S.; Jialal, I. Vitamin E, oxidative stress, and inflammation. Annu. Rev. Nutr. 2005, 25, 151–174. [Google Scholar] [CrossRef]
- Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [Green Version]
- Sosa, V.; Moliné, T.; Somoza, R.; Paciucci, R.; Kondoh, H.; LLeonart, M.E. Oxidative stress and cancer: An overview. Ageing Res. Rev. 2013, 12, 376–390. [Google Scholar] [CrossRef]
- Krithika, R.; Verma, R.J.; Shrivastav, P.S. Antioxidative and cytoprotective effects of andrographolide against CCl4-induced hepatotoxicity in HepG2 cells. Hum. Exp. Toxicol. 2013, 32, 530–543. [Google Scholar] [CrossRef]
- Zhao, Y.; Kao, C.-P.; Wu, K.-C.; Liao, C.-R.; Ho, Y.-L.; Chang, Y.-S. Chemical compositions, chromatographic fingerprints and antioxidant activities of Andrographis Herba. Molecules 2014, 19, 18332–18350. [Google Scholar] [CrossRef] [Green Version]
- Lin, F.L.; Wu, S.J.; Lee, S.C.; Ng, L.T. Antioxidant, antioedema and analgesic activities of Andrographis paniculata extracts and their active constituent andrographolide. Phytother. Res. 2009, 23, 958–964. [Google Scholar] [CrossRef] [PubMed]
- Peng, S.; Gao, J.; Liu, W.; Jiang, C.; Yang, X.; Sun, Y.; Guo, W.; Xu, Q. Andrographolide ameliorates OVA-induced lung injury in mice by suppressing ROS-mediated NF-κB signaling and NLRP3 inflammasome activation. Oncotarget 2016, 7, 80262–80274. [Google Scholar] [CrossRef] [PubMed]
- Sheeja, K.; Shihab, P.K.; Kuttan, G. Antioxidant and anti-inflammatory activities of the plant Andrographis paniculata Nees. Immunopharmacol. Immunotoxicol. 2006, 28, 129–140. [Google Scholar] [CrossRef] [PubMed]
- Shen, Y.-C.; Chen, C.-F.; Chiou, W.-F. Andrographolide prevents oxygen radical production by human neutrophils: Possible mechanism(s) involved in its anti-inflammatory effect. Br. J. Pharmacol. 2002, 135, 399–406. [Google Scholar] [CrossRef] [Green Version]
- Geng, J.; Liu, W.; Xiong, Y.; Ding, H.; Jiang, C.; Yang, X.; Li, X.; Elgehama, A.; Sun, Y.; Xu, Q.; et al. Andrographolide sulfonate improves Alzheimer-associated phenotypes and mitochondrial dysfunction in APP/PS1 transgenic mice. Biomed. Pharmacother. 2018, 97, 1032–1039. [Google Scholar] [CrossRef]
- Das, S.; Gautam, N.; Dey, S.K.; Maiti, T.; Roy, S. Oxidative stress in the brain of nicotine-induced toxicity: Protective role of Andrographis paniculata Nees and vitamin E. Appl. Physiol. Nutr. Metab. 2009, 34, 124–135. [Google Scholar] [CrossRef]
- Roy, D.N.; Sen, G.; Chowdhury, K.D.; Biswas, T. Combination therapy with andrographolide and d-penicillamine enhanced therapeutic advantage over monotherapy with d-penicillamine in attenuating fibrogenic response and cell death in the periportal zone of liver in rats during copper toxicosis. Toxicol. Appl. Pharmacol. 2011, 250, 54–68. [Google Scholar] [CrossRef]
- Batkhuu, J.; Hattori, K.; Takano, F.; Fushiya, S.; Oshiman, K.; Fujimiya, Y. Suppression of NO production in activated macrophages in vitro and ex vivo by neoandrographolide isolated from Andrographis paniculata. Biol. Pharm. Bull. 2002, 25, 1169–1174. [Google Scholar] [CrossRef] [Green Version]
- Das, S.; Neogy, S.; Gautam, N.; Roy, S. In vitro nicotine induced superoxide mediated DNA fragmentation in lymphocytes: Protective role of Andrographis paniculata Nees. Toxicol. In Vitro 2009, 23, 90–98. [Google Scholar] [CrossRef]
- Alzaharna, M.; Alqouqa, I.; Cheung, H.-Y. Taxifolin synergizes Andrographolide-induced cell death by attenuation of autophagy and augmentation of caspase dependent and independent cell death in HeLa cells. PLoS ONE 2017, 12, e0171325. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Guo, W.; Li, L.; Fu, Z.; Liu, W.; Gao, J.; Shu, Y.; Xu, Q.; Sun, Y.; Gu, Y. Andrographolide reversed 5-FU resistance in human colorectal cancer by elevating BAX expression. Biochem. Pharmacol. 2016, 121, 8–17. [Google Scholar] [CrossRef]
- Chen, W.; Feng, L.; Nie, H.; Zheng, X. Andrographolide induces autophagic cell death in human liver cancer cells through cyclophilin D-mediated mitochondrial permeability transition pore. Carcinogenesis 2012, 33, 2190–2198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chern, C.-M.; Liou, K.-T.; Wang, Y.-H.; Liao, J.-F.; Yen, J.-C.; Shen, Y.-C. Andrographolide inhibits PI3K/AKT-dependent NOX2 and iNOS expression protecting mice against hypoxia/ischemia-induced oxidative brain injury. Planta Med. 2011, 77, 1669–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, E.; Liu, X.; Du, Z.; Yang, R.; Zhao, Y. Andrographolide Ameliorates Diabetic Cardiomyopathy in Mice by Blockage of Oxidative Damage and NF-κB-Mediated Inflammation. Oxid. Med. Cell Longev. 2018, 2018, 9086747. [Google Scholar] [CrossRef] [Green Version]
- Yang, N.; Liu, Y.-Y.; Pan, C.-S.; Sun, K.; Wei, X.-H.; Mao, X.-W.; Lin, F.; Li, X.-J.; Fan, J.-Y.; Han, J.-Y. Pretreatment with andrographolide pills(®) attenuates lipopolysaccharide-induced pulmonary microcirculatory disturbance and acute lung injury in rats. Microcirculation 2014, 21, 703–716. [Google Scholar] [CrossRef] [PubMed]
- Lu, C.-Y.; Yang, Y.-C.; Li, C.-C.; Liu, K.-L.; Lii, C.-K.; Chen, H.-W. Andrographolide inhibits TNFα-induced ICAM-1 expression via suppression of NADPH oxidase activation and induction of HO-1 and GCLM expression through the PI3K/Akt/Nrf2 and PI3K/Akt/AP-1 pathways in human endothelial cells. Biochem. Pharmacol. 2014, 91, 40–50. [Google Scholar] [CrossRef]
- Yuan, M.; Meng, W.; Liao, W.; Lian, S. Andrographolide Antagonizes TNF-α-Induced IL-8 via Inhibition of NADPH Oxidase/ROS/NF-κB and Src/MAPKs/AP-1 Axis in Human Colorectal Cancer HCT116 Cells. J. Agric. Food Chem. 2018, 66, 5139–5148. [Google Scholar] [CrossRef]
- Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [Green Version]
- Yan, G.-R.; Zhou, H.-H.; Wang, Y.; Zhong, Y.; Tan, Z.-L.; Wang, Y.; He, Q.-Y. Protective effects of andrographolide analogue AL-1 on ROS-induced RIN-mβ cell death by inducing ROS generation. PLoS ONE 2013, 8, e63656. [Google Scholar] [CrossRef] [Green Version]
- Ives, A.; Nomura, J.; Martinon, F.; Roger, T.; LeRoy, D.; Miner, J.N.; Simon, G.; Busso, N.; So, A. Xanthine oxidoreductase regulates macrophage IL1β secretion upon NLRP3 inflammasome activation. Nat. Commun. 2015, 6, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Kelley, E.E. Diminishing Inflammation by Reducing Oxidant Generation: Nitrated Fatty Acid-Mediated Inactivation of Xanthine Oxidoreductase. Adv. Exp. Med. Biol. 2019, 1127, 59–65. [Google Scholar] [PubMed]
- Thangathirupathi, A.; Ali, N.; Natarajan, P.; Ramesh Kumar, D. Molecular docking studies of andrographolide with xanthine oxidase. Asian J. Pharm. Clin. Res. 2013, 6, 295–297. [Google Scholar]
- Fridovich, I. Superoxide dismutases. Annu. Rev. Biochem. 1975, 44, 147–159. [Google Scholar] [CrossRef] [PubMed]
- Aebi, H. Catalase in vitro. In Methods in Enzymology; Oxygen Radicals in Biological Systems; Academic Press: Cambridge, MA, USA, 1984; Volume 105, pp. 121–126. [Google Scholar]
- Flohe, L.; Günzler, W.A.; Schock, H.H. Glutathione peroxidase: A selenoenzyme. FEBS Lett. 1973, 32, 132–134. [Google Scholar] [CrossRef] [Green Version]
- Rotruck, J.T.; Pope, A.L.; Ganther, H.E.; Swanson, A.B.; Hafeman, D.G.; Hoekstra, W.G. Selenium: Biochemical role as a component of glutathione peroxidase. Science 1973, 179, 588–590. [Google Scholar] [CrossRef]
- Zitka, O.; Skalickova, S.; Gumulec, J.; Masarik, M.; Adam, V.; Hubalek, J.; Trnkova, L.; Kruseova, J.; Eckschlager, T.; Kizek, R. Redox status expressed as GSH:GSSG ratio as a marker for oxidative stress in paediatric tumour patients. Oncol. Lett. 2012, 4, 1247–1253. [Google Scholar] [CrossRef]
- Trivedi, N.P.; Rawal, U.M.; Patel, B.P. Hepatoprotective effect of andrographolide against hexachlorocyclohexane-induced oxidative injury. Integr. Cancer Ther. 2007, 6, 271–280. [Google Scholar] [CrossRef]
- Akowuah, G.A.; Zhari, I.; Mariam, A.; Yam, M.F. Absorption of andrographolides from Andrographis paniculata and its effect on CCl(4)-induced oxidative stress in rats. Food Chem. Toxicol. 2009, 47, 2321–2326. [Google Scholar] [CrossRef]
- Al Batran, R.; Al-Bayaty, F.; Al-Obaidi, M.M.J.; Abdulla, M.A. Acute toxicity and the effect of andrographolide on Porphyromonas gingivalis-induced hyperlipidemia in rats. Biomed. Res. Int. 2013, 2013, 594012. [Google Scholar] [CrossRef]
- Al Batran, R.; Al-Bayaty, F.; Al-Obaidi, M.M.J.; Ashrafi, A. Insights into the antiatherogenic molecular mechanisms of andrographolide against Porphyromonas gingivalis-induced atherosclerosis in rabbits. Naunyn Schmiedebergs Arch. Pharmacol. 2014, 387, 1141–1152. [Google Scholar] [CrossRef]
- Liu, Y.-H.; Zhang, Z.-B.; Zheng, Y.-F.; Chen, H.-M.; Yu, X.-T.; Chen, X.-Y.; Zhang, X.; Xie, J.-H.; Su, Z.-Q.; Feng, X.-X.; et al. Gastroprotective effect of andrographolide sodium bisulfite against indomethacin-induced gastric ulceration in rats. Int. Immunopharmacol. 2015, 26, 384–391. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Pradhan, G.K.; Das, S.; Nath, D.; Das Saha, K. Enhanced protective activity of nano formulated andrographolide against arsenic induced liver damage. Chem. Biol. Interact. 2015, 242, 281–289. [Google Scholar] [CrossRef] [PubMed]
- Thakur, A.K.; Rai, G.; Chatterjee, S.S.; Kumar, V. Beneficial effects of an Andrographis paniculata extract and andrographolide on cognitive functions in streptozotocin-induced diabetic rats. Pharm. Biol. 2016, 54, 1528–1538. [Google Scholar] [CrossRef] [Green Version]
- Naik, R.R.; Munipally, P.K.; Nagaraju, T. Andrographolide reorganise hyperglycaemia and distorted antioxidant profile in streptozotocin-induced diabetic rats. Cardiovasc. Hematol. Agents Med. Chem. 2017. [Google Scholar] [CrossRef] [PubMed]
- Guo, H.; Zhang, Z.; Su, Z.; Sun, C.; Zhang, X.; Zhao, X.; Lai, X.; Su, Z.; Li, Y.; Zhan, J.Y. Enhanced anti-tumor activity and reduced toxicity by combination andrographolide and bleomycin in ascitic tumor-bearing mice. Eur. J. Pharmacol. 2016, 776, 52–63. [Google Scholar] [CrossRef]
- Zhan, J.Y.-X.; Wang, X.-F.; Liu, Y.-H.; Zhang, Z.-B.; Wang, L.; Chen, J.-N.; Huang, S.; Zeng, H.-F.; Lai, X.-P. Andrographolide Sodium Bisulfate Prevents UV-Induced Skin Photoaging through Inhibiting Oxidative Stress and Inflammation. Mediat. Inflamm. 2016, 2016, 3271451. [Google Scholar] [CrossRef] [Green Version]
- Lu, H.; Zhang, X.; Zhou, Y.; Wen, X.; Zhu, L. Proteomic alterations in mouse kidney induced by andrographolide sodium bisulfite. Acta Pharmacol. Sin. 2011, 32, 888–894. [Google Scholar] [CrossRef] [Green Version]
- Yan, H.; Li, Y.; Yang, Y.; Zhang, Z.; Zhang, G.; Sun, Y.; Yu, P.; Wang, Y.; Xu, L. Protective effects of andrographolide derivative AL-1 on high glucose-induced oxidative stress in RIN-m cells. Curr. Pharm. Des. 2016, 22, 499–505. [Google Scholar] [CrossRef]
- Gu, L.-L.; Zhang, X.-Y.; Xing, W.-M.; Xu, J.-D.; Lu, H. Andrographolide-induced apoptosis in human renal tubular epithelial cells: Roles of endoplasmic reticulum stress and inflammatory response. Environ. Toxicol. Pharmacol. 2016, 45, 257–264. [Google Scholar] [CrossRef]
- Yu, A.-L.; Lu, C.-Y.; Wang, T.-S.; Tsai, C.-W.; Liu, K.-L.; Cheng, Y.-P.; Chang, H.C.; Lii, C.-K.; Chen, H.-W. Induction of heme oxygenase 1 and inhibition of tumor necrosis factor alpha-induced intercellular adhesion molecule expression by andrographolide in EA.hy926 cells. J. Agric. Food Chem. 2010, 58, 7641–7648. [Google Scholar] [CrossRef]
- Lin, H.-C.; Su, S.-L.; Lu, C.-Y.; Lin, A.-H.; Lin, W.-C.; Liu, C.-S.; Yang, Y.-C.; Wang, H.-M.; Lii, C.-K.; Chen, H.-W. Andrographolide inhibits hypoxia-induced HIF-1α-driven endothelin 1 secretion by activating Nrf2/HO-1 and promoting the expression of prolyl hydroxylases 2/3 in human endothelial cells. Environ. Toxicol. 2017, 32, 918–930. [Google Scholar] [CrossRef] [PubMed]
- Guan, S.P.; Tee, W.; Ng, D.S.W.; Chan, T.K.; Peh, H.Y.; Ho, W.E.; Cheng, C.; Mak, J.C.; Wong, W.S.F. Andrographolide protects against cigarette smoke-induced oxidative lung injury via augmentation of Nrf2 activity. Br. J. Pharmacol. 2013, 168, 1707–1718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.-C.; Tseng, C.-K.; Young, K.-C.; Sun, H.-Y.; Wang, S.-W.; Chen, W.-C.; Lin, C.-K.; Wu, Y.-H. Andrographolide exerts anti-hepatitis C virus activity by up-regulating haeme oxygenase-1 via the p38 MAPK/Nrf2 pathway in human hepatoma cells. Br. J. Pharmacol. 2014, 171, 237–252. [Google Scholar] [CrossRef] [PubMed]
- Yen, T.-L.; Chen, R.-J.; Jayakumar, T.; Lu, W.-J.; Hsieh, C.-Y.; Hsu, M.-J.; Yang, C.-H.; Chang, C.-C.; Lin, Y.-K.; Lin, K.-H.; et al. Andrographolide stimulates p38 mitogen-activated protein kinase-nuclear factor erythroid-2-related factor 2-heme oxygenase 1 signaling in primary cerebral endothelial cells for definite protection against ischemic stroke in rats. Transl. Res. 2016, 170, 57–72. [Google Scholar] [CrossRef] [PubMed]
- Wong, S.Y.; Tan, M.G.K.; Wong, P.T.H.; Herr, D.R.; Lai, M.K.P. Andrographolide induces Nrf2 and heme oxygenase 1 in astrocytes by activating p38 MAPK and ERK. J. Neuroinflamm. 2016, 13, 251. [Google Scholar] [CrossRef] [Green Version]
- Seo, J.Y.; Pyo, E.; An, J.-P.; Kim, J.; Sung, S.H.; Oh, W.K. Andrographolide Activates Keap1/Nrf2/ARE/HO-1 Pathway in HT22 Cells and Suppresses Microglial Activation by Aβ42 through Nrf2-Related Inflammatory Response. Mediat. Inflamm. 2017, 2017, 5906189. [Google Scholar] [CrossRef] [Green Version]
- Gu, L.; Yu, Q.; Li, Q.; Zhang, L.; Lu, H.; Zhang, X. Andrographolide Protects PC12 Cells Against β-Amyloid-Induced Autophagy-Associated Cell Death Through Activation of the Nrf2-Mediated p62 Signaling Pathway. Int. J. Mol. Sci. 2018, 19, 2844. [Google Scholar] [CrossRef] [Green Version]
- Wong, D.P.W.; Ng, M.Y.; Leung, J.Y.; Boh, B.K.; Lim, E.C.; Tan, S.H.; Lim, S.; Seah, W.H.; Hu, C.Z.; Ho, B.C.; et al. Regulation of the NRF2 transcription factor by andrographolide and organic extracts from plant endophytes. PLoS ONE 2018, 13, e0204853. [Google Scholar] [CrossRef]
- Khole, S.; Mittal, S.; Jagadish, N.; Ghosh, D.; Gadgil, V.; Sinkar, V.; Ghaskadbi, S. Andrographolide enhances redox status of liver cells by regulating microRNA expression. Free Radic. Biol. Med. 2019, 130, 397–407. [Google Scholar] [CrossRef]
- Zhang, D.D.; Hannink, M. Distinct cysteine residues in Keap1 are required for Keap1-dependent ubiquitination of Nrf2 and for stabilization of Nrf2 by chemopreventive agents and oxidative stress. Mol. Cell. Biol. 2003, 23, 8137–8151. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.-J.; Sun, Z.; Chen, W.; Li, Y.; Villeneuve, N.F.; Zhang, D.D. Activation of Nrf2 by arsenite and monomethylarsonous acid is independent of Keap1-C151: Enhanced Keap1-Cul3 interaction. Toxicol. Appl. Pharmacol. 2008, 230, 383–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, C.; Eggler, A.L.; Mesecar, A.D.; van Breemen, R.B. Modification of keap1 cysteine residues by sulforaphane. Chem. Res. Toxicol. 2011, 24, 515–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hossain, M.S.; Urbi, Z.; Sule, A.; Hafizur Rahman, K.M. Andrographis paniculata (Burm. f.) Wall. ex Nees: A review of ethnobotany, phytochemistry, and pharmacology. Sci. World J. 2014, 2014, 274905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, B.; Yan, L.; Zhou, P.; Dong, Z.; Feng, S.; Liu, K.; Gong, Z. CHP1002, a novel andrographolide derivative, inhibits pro-inflammatory inducible nitric oxide synthase and cyclooxygenase-2 expressions in RAW264.7 macrophages via up-regulation of heme oxygenase-1 expression. Int. Immunopharmacol. 2013, 15, 289–295. [Google Scholar] [CrossRef]
- Tan, W.S.D.; Peh, H.Y.; Liao, W.; Pang, C.H.; Chan, T.K.; Lau, S.H.; Chow, V.T.; Wong, W.S.F. Cigarette Smoke-Induced Lung Disease Predisposes to More Severe Infection with Nontypeable Haemophilus influenzae: Protective Effects of Andrographolide. J. Nat. Prod. 2016, 79, 1308–1315. [Google Scholar] [CrossRef]
- Sulaiman, I.; Tan, K.; Mohtarrudin, N.; Lim, J.C.W.; Stanslas, J. Andrographolide prevented toluene diisocyanate-induced occupational asthma and aberrant airway E-cadherin distribution via p38 MAPK-dependent Nrf2 induction. Pulm. Pharmacol. Ther. 2018, 53, 39–51. [Google Scholar] [CrossRef]
Treatment(s) | Nrf2 mRNA | Nrf2 Protein | Nrf2 Translocation | Nrf2 Phosphorylation | Nrf2 Binding Activity to ARE Sequence | Nrf2 Inhibitor (Keap-1) | Nrf2 Turnover and Ubiquitination | Nrf2 Targets | Ref. |
---|---|---|---|---|---|---|---|---|---|
2.5, 5, and 7.5 µM for 16 h followed by incubation with TNF-α (1 ng/mL) for an additional 6 h | - | - | ↗ | - | ↗ | - | - | HO-1 mRNA and protein ↗ | [81] |
7.5 µM andrographolide for 16 h | - | - | - | - | - | - | - | HO-1 protein ↗ | [82] |
30 µM andrographolide with 2% cigarette smoke extract | - | - | ↗ | - | ↗ | - | - | GSH ↗; expression of antioxidants GCLM, GCLC, GR, GPx-2, and HO-1 ↗ | [83] |
30 µM for 24 h | - | - | ↗ | - | ↗ | - | - | ↗ ARE-sensitive genes including HO-1, GCLC, GCLM, SRXN1, TXNRD1, and GSR but not NQ01 | [32] |
1, 5, 7.5, and 10 µM | - | ↗ | ↗ | - | ↗ | No effect | - | HO-1 promoter activity and mRNA ↗ | [84] |
7.5 µM andrographolide for 24 h | - | - | ↗ | - | - | - | - | HO-1 mRNA and protein ↗; GCLM mRNA and protein ↗ but not GCLC | [56] |
5 and 10 µM | - | - | ↗ | p-Nrf2 (ser40) ↗ | - | - | - | HO-1 mRNA and protein ↗ | [85] |
1, 5, 10, 30, and 50 µM | 50 µM for 24h ↗ | 1–50 µM for 1h ↗, and 50 µM for 30 min to 24 h ↗ | 50 µM for 30 min to 24 h ↗ | no effect on p-Nrf2 (ser40) | - | No effect | ↘ | HO-1 mRNA (2 h with 50 µM) and protein (4 h with 50 µM) ↗ | [86] |
1, 5, and 10 µM | - | ↗ | ↗ | - | ↗ | No effect | - | HO-1 mRNA and protein ↗ | [87] |
Glucose 25 mM and andrographolide 0.1, 1, 5, and 10 µM | - | ↗ | - | - | - | - | - | HO-1 protein ↗, Nox2, Nox4, and P47phox mRNA ↘ | [54] |
H2O2 0.1 mmol/L and andrographolide 0.625 and 2.5 µg/mL | - | ↗ | - | - | - | - | - | Activities of SOD and CAT ↗; SOD and CAT proteins ↗ | [31] |
β-amyloid (Aβ) peptide 10 µM and andrographoldie 20 µM for 1 h | ↗ | ↗ | - | - | - | - | - | - | [88] |
1, 7.5, 15, 30, 60, and 120 µM | - | ↗ | - | - | ↗ | At low concentration, function of the CUL3-RBX1-KEAP-1 E3 ubiquitin ligase via Cys151 ↘ at high concentration action via a mechanism independent of Cys151 in KEAP-1 | ↗ | - | [89] |
20 and 40 µM | ↗ | ↗ | ↗ | - | - | - | - | HO-1 protein ↗; GSH protein ↗; GCLC/GCLM/GS mRNAs and proteins ↗; GR mRNA, protein, and activity ↗ but GPx1 mRNA and total GPx activity ↘ | [93] |
Model(s) | Treatment(s) | Nrf2 mRNA | Nrf2 Protein | Nrf2 Translocation | Nrf2 Binding Activity to ARE Sequence | Nrf2 Inhibitor (Keap-1) | Nrf2 Targets | Ref. |
---|---|---|---|---|---|---|---|---|
Sprague–Dawley rats (liver, heart, and kidneys) | 30 or 50 mg/kg/day for 5 consecutive days, intragastrical | - | - | ↗ (liver) | ↗ (liver) | - | SOD activity ↗ (liver, kidney, heart, and red blood cells); CAT activity ↗ (heart); GSH peroxidase activity ↗ (kidney); GSH reductase ↗ (kidney, heart, and red blood cells); GSH S-transferase ↗ (liver); GSH protein ↗ (heart); antioxidant proteins (SOD1, GST Ya, GST Yb, HO-1, GCLC, and GCLM) ↗ (liver, kidney, and heart); mRNA (GCLC, GCLM, GST Ya/Yb, SOD1, and HO-1) ↗ (liver and kidney) | [25] |
Wistar rats | 0.1 mg/kg, intraperitoneal (6 h) | - | - | - | - | - | HO-1 protein ↗ (brains) | [85] |
BALB/c mice | 5 or 10 mg/kg, intraperitoneal (1 and 24 h) | - | - | ↗ (lung) | - | mRNA ↘ (lung) | HO-1, GR, GCLM, GPx-2, and NQO1 (mRNA) ↗ | [96] |
BALB/c mice | LPS/GalN (1 h) followed by andrographolide (2.5, 5, or 10 mg/kg), intraperitoneal (8 h) | - | ↗ (liver) | - | - | - | HO-1 protein ↗ (liver) | [6] |
C57BL/6 mice | Acetaminophen (orally) every day for 6 weeks followed by andrographolide (20 or 40 mg/kg, orally) treatment every day at 2 weeks after acetaminophen administration | - | - | ↗ (liver) | - | - | ANDRO reversed the decreased hepatic expression of GCLC, GCLM and HO-1 mRNA expression induced by acetaminophen. Co-treatment with ANDRO (40 mg/kg) ↗ NQO1 mRNA | [33] |
C57BL/6 mice | Streptozotocin (intraperitoneal injection) for 5 consecutive days followed by andrographolide (1, 10, or 20 mg/kg/day) for 12 weeks by intragastric gavage | ↗ (heart) | - | - | - | - | SOD activity ↗; MDA and 4-HNE ↘; Nox2, Nox-4, p47phox, Nrf2, and HO-1 mRNA ↘ | [54] |
Balb/c mice | Toluene diisocyanate treatment (dermally and intranasally) for asthma induction with andrographolide treatment (0.1, 0.5, or 1 mg/kg, prophylatic regimen) | - | ↗ (lung) | - | - | - | HO-1 protein ↗ (1 mg/kg, lung) | [97] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mussard, E.; Cesaro, A.; Lespessailles, E.; Legrain, B.; Berteina-Raboin, S.; Toumi, H. Andrographolide, A Natural Antioxidant: An Update. Antioxidants 2019, 8, 571. https://doi.org/10.3390/antiox8120571
Mussard E, Cesaro A, Lespessailles E, Legrain B, Berteina-Raboin S, Toumi H. Andrographolide, A Natural Antioxidant: An Update. Antioxidants. 2019; 8(12):571. https://doi.org/10.3390/antiox8120571
Chicago/Turabian StyleMussard, Eugenie, Annabelle Cesaro, Eric Lespessailles, Brigitte Legrain, Sabine Berteina-Raboin, and Hechmi Toumi. 2019. "Andrographolide, A Natural Antioxidant: An Update" Antioxidants 8, no. 12: 571. https://doi.org/10.3390/antiox8120571
APA StyleMussard, E., Cesaro, A., Lespessailles, E., Legrain, B., Berteina-Raboin, S., & Toumi, H. (2019). Andrographolide, A Natural Antioxidant: An Update. Antioxidants, 8(12), 571. https://doi.org/10.3390/antiox8120571