Antioxidant Activity in Extracts from Zingiberaceae Family: Cardamom, Turmeric, and Ginger
Abstract
:1. Introduction
Plants | Dose | Phytochemicals | Bioactive Compounds |
---|---|---|---|
Cardamom | 1–3 g/d | Terpenes | 1,8-cineole, α-terpinyl acetate, nerolidol, sabinene, g-terpinene, α-pinene, methyl linoleate, α-terpineol, β-pinene, n-hexadecanoic acid, and limonene |
Phenolic compounds | Curcumin I (1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) | ||
Turmeric | 0.5–2 g/d | Curcuminoid (polyphenolic compounds) | Curcumin II (demethoxycurcumin, 1-(4-hydroxy-3-methoxyphenyl)-7-(4-hydroxyphenyl)-1,6-heptadiene-3,5-dione, and curcumin III (bisdemethoxycurcumin, 1,7-bis(4-hydroxyphenyl)-1,6-heptadiene-3,5-dione) |
Non-curcuminoids (terpenoids and phenolic compounds) | Turmerones, elemene, bisacurone, curdione, cyclocurcumin, germacrone, furanodiene, curcumol, and calebin A | ||
Ginger | 1–3 g/d | Phenolics compounds | Gingerols, shogaols, paradols, quercetin, zingerone, and gingerenone-A |
Terpene components | β-bisabolene, α-curcumene, zingiberene, α-farnesene, β-sesquiphellandrene, and zingiberene |
2. Relationship between Oxidative Stress and Health
Free Radicals | No Radicals |
---|---|
Reactive Oxygen Species | |
Superoxide anion radical (O2¯˙) | Hydrogen peroxide (H2O2) |
Hydroxyl radical (OH˙) | Singlet molecular oxygen (O21 ∆g) |
Peroxyl radical (ROO˙) | |
Reactive Nitrogen Species | |
Nitric oxide (NO˙) | Nitrite (NO2¯) |
Nitrogen dioxide (NO2˙) | Peroxynitrite (ONOO¯) |
Endogenous Sources | Exogenous Sources |
---|---|
Inflammation | Exposure to contaminants |
Ischemia | Exposure to heavy metals (Hg, Cd, Pb, Fe, and As) |
Infection | Chemical solvents |
Cancer | Food cooking (smoked meat and used oil) |
Activation of immune cells | Medications (cyclosporine, tacrolimus, bleomycin, and gentamicin) |
Excessive exercise | Cigarette smoke |
Mental stress | Alcohol |
Aging | Radiation |
3. Antioxidant Activity of Zingiberaceae Family
3.1. Antioxidant Activity of Ginger
Extract | Function | Mechanism | References |
---|---|---|---|
Ginger | Antioxidant | Decreased AKT-phosphorylation | [70] |
Decreased ERS markers | [71] | ||
Decreased MDA and increased GPx levels | [72,73] | ||
Inhibition of XO | [21] | ||
Inhibition of NO production and reduction in iNOS | [75,76] | ||
Anti-inflammatory | Increased Beclin1 expression and obstruction of signaling in the PI3K/AKT/mTOR pathway | [78] | |
Inhibition of NF-κB activation and decrease in the level of IL-1β | [79] | ||
Inhibition of the production of PGE2 and pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, and COX-2) | [80] | ||
Anticancer | Decrease in tumor volume and tumor burden by increasing p21 CKDIs | [81,82,83] | |
Antiemetic | Inhibition of 5-HT3 receptors in the central and peripheral nervous systems | [84,85,86] | |
Antimicrobial | Inhibition of bacterial protein synthesis, DNA replication and cell division. | [94] | |
Cardiovascular disease prevention | Reducing effect of PCSK9 through a modulatory mechanism of lipid levels | [88] | |
Neurodegenerative disease prevention | Inhibition of TNF-α, NO, COX-2, and iNOS | [89] | |
Inhibition of glial cell activation | [90] | ||
Protective effect against amyloid-β | [91] |
3.2. Antioxidant Activity of Turmeric
Extract | Function | Mechanism | References |
---|---|---|---|
Turmeric | Antioxidant | Chelates of pro-oxidant metals | [99] |
Strongly inhibits advanced glycation end products and lipid peroxidation | [100] | ||
Reduces lipid peroxidation | |||
Acts as an activator of SIRT1 and SIRT3 | [100,101,102,103] | ||
Reduces MDA levels and CRP | [104] | ||
Improves serum SOD and CAT activities | [100,105] | ||
Enhances TAC | [105] | ||
Anti-inflammatory | Inhibits the production of inflammatory mediators | [111,112] | |
Inhibits the production of inflammatory molecules, such as TNF-α and IL-6 | |||
Inhibits COX-2 and 5-LOX | |||
Anticancer | Reduces the production of ROS that damage DNA and other cellular structures | [116,117] | |
Has anti-inflammatory action | |||
Induces apoptosis | |||
Inhibits angiogenesis | |||
Antimicrobial, antiviral and antifungal activity | Increases the efficacy of antimicrobial treatments | [118,119] | |
Inhibitions FtsZ assembly on the Z-ring and the suppression of bacterial cells | |||
Inhibits HIV replication | |||
Acidifies the intracellular environment | |||
Cardiovascular disease prevention | Has ability to prevent endothelial dysfunction and smooth muscle cell proliferation and migration | [114,115] | |
Inhibits Toll-like receptor 4 expression | |||
Has ability to suppress cholesterol accumulation in macrophage foam cells and atherosclerosis | |||
Neurodegenerative disease prevention | Inhibits the formation and promotes the disaggregation of amyloid-β plaques | [106,107,108] | |
Attenuates the hyperphosphorylation of tau and enhances its clearance | |||
Modifies microglial activity | |||
Inhibits acetylcholinesterase | |||
Binds to redox active metals (copper and iron) | |||
Suppresses the differentiation of Th17 cells | [5] | ||
Exhibits antioxidant activity | [111,112] |
3.3. Antioxidant Activity of Cardamom
Extract | Function | Mechanism of Action | References |
---|---|---|---|
Cardamom | Antioxidant | Decreased NO, APOP, and MDA | [8,122,123,126] |
Increased activity of CAT, SOD, and GSH | [8,122,123] | ||
Increased Sirt1 | [122,132] | ||
Anti- inflammatory | Decreased serum CRP | [126] | |
Decreased hs-CRP:IL-6 ratio | |||
Inhibited the production of inflammatory markers (IL-6, TNF-α, ALT, and fatty liver status) | [131] | ||
Anticancer * | Enhanced the hepatic activities of oxidation-preventing hormones (such as glutathione-S-transferases, SOD, GPx, and CAT) | [137] | |
Antiemetic | Volatile compounds from essential oil were significantly efficient to reduce nausea level | [139,140] | |
Antimicrobial | Enhanced NK cell activity and anti-MRSA biofilm activity and avoided MRSA adhesion ability; cardamom may downregulate MRSA biofilm-forming genes | [122,123,124,141] | |
Cardiovascular disease prevention | Decreased systolic, diastolic, and mean blood pressure, and increased fibrinolytic activity | [136] | |
Neurodegenerative disease prevention | Exhibited anti-cholinesterase activity, induced endogenous antioxidants, such as glutathione and superoxide dismutase, impeded the production of reactive hydroxyl radicals, prevented the formation of Aβ42 deposits, and protected cells from iron-induced death | [142] |
4. Conclusions and Future Research Lines
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Lakhan, S.E.; Ford, C.T.; Tepper, D. Zingiberaceae extracts for pain: A systematic review and meta-analysis. Nutr. J. 2015, 14, 50. [Google Scholar] [CrossRef] [PubMed]
- Peng, W.; Li, P.; Ling, R.; Wang, Z.; Feng, X.; Liu, J.; Yang, Q.; Yan, J. Diversity of Volatile Compounds in Ten Varieties of Zingiberaceae. Molecules 2022, 27, 565. [Google Scholar] [CrossRef]
- Rachkeeree, A.; Kantadoung, K.; Suksathan, R.; Puangpradab, R.; Page, P.A.; Sommano, S.R. Nutritional Compositions and Phytochemical Properties of the Edible Flowers from Selected Zingiberaceae Found in Thailand. Front. Nutr. 2018, 5, 3. [Google Scholar] [CrossRef] [PubMed]
- Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Roy, N.K.; Prasad, S.; Aggarwal, B.B. Curcumin, the golden nutraceutical: Multitargeting for multiple chronic diseases. Br. J. Pharmacol. 2017, 174, 1325–1348. [Google Scholar] [CrossRef] [PubMed]
- Kocaadam, B.; Şanlier, N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef] [PubMed]
- Abdullah; Ahmad, N.; Tian, W.; Zengliu, S.; Zou, Y.; Farooq, S.; Huang, Q.; Xiao, J. Recent advances in the extraction, chemical composition, therapeutic potential, and delivery of cardamom phytochemicals. Front. Nutr. 2022, 9, 1024820. [Google Scholar] [CrossRef]
- Kandikattu, H.K.; Rachitha, P.; Jayashree, G.V.; Krupashree, K.; Sukhith, M.; Majid, A.; Amruta, N.; Khanum, F. Anti-inflammatory and anti-oxidant effects of Cardamom (Elettaria repens (Sonn.) Baill) and its phytochemical analysis by 4D GCXGC TOF-MS. Biomed. Pharmacother. 2017, 91, 191–201. [Google Scholar] [CrossRef]
- Rahman, M.M.; Alam, M.N.; Ulla, A.; Sumi, F.A.; Subhan, N.; Khan, T.; Sikder, B.; Hossain, H.; Reza, H.M.; Alam, M.A. Cardamom powder supplementation prevents obesity, improves glucose intolerance, inflammation and oxidative stress in liver of high carbohydrate high fat diet induced obese rats. Lipids Health Dis. 2017, 16, 151. [Google Scholar] [CrossRef]
- Aghasi, M.; Koohdani, F.; Qorbani, M.; Nasli-Esfahani, E.; Ghazi-Zahedi, S.; Khoshamal, H.; Keshavarz, A.; Sotoudeh, G. Beneficial effects of green cardamom on serum SIRT1, glycemic indices and triglyceride levels in patients with type 2 diabetes mellitus: A randomized double-blind placebo controlled clinical trial. J. Sci. Food Agric. 2019, 99, 3933–3940. [Google Scholar] [CrossRef]
- Nair, A.; Amalraj, A.; Jacob, J.; Kunnumakkara, A.B.; Gopi, S. Non-Curcuminoids from Turmeric and Their Potential in Cancer Therapy and Anticancer Drug Delivery Formulations. Biomolecules 2019, 9, 13. [Google Scholar] [CrossRef]
- Aggarwal, B.B.; Yuan, W.; Li, S.; Gupta, S.C. Curcumin-free turmeric exhibits anti-inflammatory and anticancer activities: Identification of novel components of turmeric. Mol. Nutr. Food Res. 2013, 57, 1529–1542. [Google Scholar] [CrossRef] [PubMed]
- Abrahams, S.; Haylett, W.L.; Johnson, G.; Carr, J.A.; Bardien, S. Antioxidant effects of curcumin in models of neurodegeneration, aging, oxidative and nitrosative stress: A review. Neuroscience 2019, 406, 1–21. [Google Scholar] [CrossRef]
- Garza, G.R.C.; Luévano, J.H.E.; Rodríguez, A.F.B.; Montes, A.C.; Hernández, R.A.P.; Delgado, A.J.M.; Villarreal, S.M.L.; Rodríguez, J.R.; Casas, R.M.S.; Velázquez, U.C.; et al. Benefits of Cardamom (Elettaria cardamomum (L.) Maton) and Turmeric (Curcuma longa L.) Extracts for Their Applications as Natural Anti-Inflammatory Adjuvants. Plants 2021, 10, 1908. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.A.; Kitts, D.D. Turmeric and its bioactive constituents trigger cell signaling mechanisms that protect against diabetes and cardiovascular diseases. Mol. Cell. Biochem. 2021, 476, 3785–3814. [Google Scholar] [CrossRef] [PubMed]
- Ming, T.; Tao, Q.; Tang, S.; Zhao, H.; Yang, H.; Liu, M.; Ren, S.; Xu, H. Curcumin: An epigenetic regulator and its application in cancer. Biomed. Pharmacother. 2022, 156, 113956. [Google Scholar] [CrossRef] [PubMed]
- Gupta, N.; Verma, K.; Nalla, S.; Kulshreshtha, A.; Lall, R.; Prasad, S. Free Radicals as a Double-Edged Sword: The Cancer Preventive and Therapeutic Roles of Curcumin. Molecules 2020, 25, 5390. [Google Scholar] [CrossRef]
- Jabczyk, M.; Nowak, J.; Hudzik, B.; Zubelewicz-Szkodzińska, B. Curcumin in Metabolic Health and Disease. Nutrients 2021, 13, 4440. [Google Scholar] [CrossRef]
- Mao, Q.-Q.; Xu, X.-Y.; Cao, S.-Y.; Gan, R.-Y.; Corke, H.; Beta, T.; Li, H.-B. Bioactive Compounds and Bioactivities of Ginger (Zingiber officinale Roscoe). Foods 2019, 8, 185. [Google Scholar] [CrossRef]
- Haniadka, R.; Saldanha, E.; Sunita, V.; Palatty, P.L.; Fayad, R.; Baliga, M.S. A review of the gastroprotective effects of ginger (Zingiber officinale Roscoe). Food Funct. 2013, 4, 845–855. [Google Scholar] [CrossRef]
- Marx, W.; Ried, K.; McCarthy, A.L.; Vitetta, L.; Sali, A.; McKavanagh, D.; Isenring, L. Ginger-Mechanism of action in chemotherapy-induced nausea and vomiting: A review. Crit. Rev. Food Sci. Nutr. 2017, 57, 141–146. [Google Scholar] [CrossRef]
- Rondanelli, M.; Fossari, F.; Vecchio, V.; Gasparri, C.; Peroni, G.; Spadaccini, D.; Riva, A.; Petrangolini, G.; Iannello, G.; Nichetti, M.; et al. Clinical trials on pain lowering effect of ginger: A narrative review. Physiol. Res. 2020, 34, 2843–2856. [Google Scholar] [CrossRef] [PubMed]
- Semwal, R.B.; Semwal, D.K.; Combrinck, S.; Viljoen, A.M. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry 2015, 117, 554–568. [Google Scholar] [CrossRef] [PubMed]
- Ajanaku, C.O.; Ademosun, O.T.; Atohengbe, P.O.; Ajayi, S.O.; Obafemi, Y.D.; Owolabi, O.A.; Akinduti, P.A.; Ajanaku, K.O. Functional bioactive compounds in ginger, turmeric, and garlic. Front. Nutr. 2022, 9, 1012023. [Google Scholar] [CrossRef] [PubMed]
- Jiang, T.A. Health Benefits of Culinary Herbs and Spices. J. AOAC Int. 2019, 102, 395–411. [Google Scholar] [CrossRef] [PubMed]
- Li, R.; Wang, Y.; Liu, Y.; Chen, Q.; Fu, W.; Wang, H.; Cai, H.; Peng, W.; Zhang, X. Curcumin Inhibits Transforming Growth Factor-β1-Induced EMT via PPARγ Pathway, Not Smad Pathway in Renal Tubular Epithelial Cells. PLoS ONE 2013, 8, e58848. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Dimango, E.; Zhu, Y.; Saroya, T.K.; Emala, C.W.; Sang, S. Pharmacokinetics of Gingerols, Shogaols, and Their Metabolites in Asthma Patients. J. Agric. Food Chem. 2022, 70, 9674–9683. [Google Scholar] [CrossRef]
- Racz, L.Z.; Racz, C.P.; Pop, L.C.; Tomoaia, G.; Mocanu, A.; Barbu, I.; Sárközi, M.; Roman, I.; Avram, A.; Tomoaia-Cotisel, M.; et al. Strategies for Improving Bioavailability, Bioactivity, and Physical-Chemical Behavior of Curcumin. Molecules 2022, 27, 6854. [Google Scholar] [CrossRef]
- Bi, X.; Yuan, Z.; Qu, B.; Zhou, H.; Liu, Z.; Xie, Y. Piperine enhances the bioavailability of silybin via inhibition of efflux transporters BCRP and MRP2. Phytomedicine 2019, 54, 98–108. [Google Scholar] [CrossRef]
- Sohn, S.-I.; Priya, A.; Balasubramaniam, B.; Muthuramalingam, P.; Sivasankar, C.; Selvaraj, A.; Valliammai, A.; Jothi, R.; Pandian, S. Biomedical Applications and Bioavailability of Curcumin-An Updated Overview. Pharmaceutics 2021, 13, 2102. [Google Scholar] [CrossRef]
- Taghipour, Y.D.; Bahramsoltani, R.; Marques, A.M.; Naseri, R.; Rahimi, R.; Haratipour, P.; Panah, A.I.; Farzaei, M.H.; Abdollahi, M. A systematic review of nano formulation of natural products for the treatment of inflammatory bowel disease: Drug delivery and pharmacological targets. DARU J. Pharm. Sci. 2018, 26, 229–239. [Google Scholar] [CrossRef]
- Wang, Q.; Yang, Q.; Cao, X.; Wei, Q.; Firempong, C.K.; Guo, M.; Shi, F.; Xu, X.; Deng, W.; Yu, J. Enhanced oral bioavailability and anti-gout activity of [6]-shogaol-loaded solid lipid nanoparticles. Int. J. Pharm. 2018, 550, 24–34. [Google Scholar] [CrossRef]
- Yücel, Ç.; Karatoprak, G.Ş.; Açıkara, Ö.B.; Akkol, E.K.; Barak, T.H.; Sobarzo-Sánchez, E.; Aschner, M.; Shirooie, S. Immunomodulatory and anti-inflammatory therapeutic potential of gingerols and their nanoformulations. Front. Pharmacol. 2022, 13, 902551. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Chen, X.; Mo, Q.; Liu, J.; Yao, X.; Di, X.; Qin, Z.; He, L.; Yao, Z. Cytochrome P450 metabolism studies of [6]-gingerol, [8]-gingerol, and [10]-gingerol by liver microsomes of humans and different species combined with expressed CYP enzymes. RSC Adv. 2023, 13, 5804–5812. [Google Scholar] [CrossRef] [PubMed]
- Ashokkumar, K.; Murugan, M.; Dhanya, M.K.; Warkentin, T.D. Botany, traditional uses, phytochemistry and biological activities of cardamom [Elettaria cardamomum (L.) Maton]—A critical review. J. Ethnopharmacol. 2020, 246, 112244. [Google Scholar] [CrossRef] [PubMed]
- Frijhoff, J.; Winyard, P.G.; Zarkovic, N.; Davies, S.S.; Stocker, R.; Cheng, D.; Knight, A.R.; Taylor, E.L.; Oettrich, J.; Ruskovska, T.; et al. Clinical Relevance of Biomarkers of Oxidative Stress. Antioxid. Redox Signal. 2015, 23, 1144–1170. [Google Scholar] [CrossRef] [PubMed]
- Scalbert, A.; Manach, C.; Morand, C.; Rémésy, C.; Jiménez, L. Dietary polyphenols and the prevention of diseases. Crit. Rev. Food Sci. Nutr. 2005, 45, 287–306. [Google Scholar] [CrossRef]
- Ďuračková, Z. Some current insights into oxidative stress. Physiol. Res. 2010, 59, 459–469. [Google Scholar] [CrossRef]
- Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
- Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.B.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxid. Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef]
- Del Río, L.A. ROS and RNS in plant physiology: An overview. J. Exp. Bot. 2015, 66, 2827–2837. [Google Scholar] [CrossRef]
- Giles, G.I.; Tasker, K.M.; Jacob, C. Hypothesis: The role of reactive sulfur species in oxidative stress. Free Radic. Biol. Med. 2001, 31, 1279–1283. [Google Scholar] [CrossRef] [PubMed]
- DeLeon, E.R.; Gao, Y.; Huang, E.; Arif, M.; Arora, N.; Divietro, A.; Patel, S.; Olson, K.R. A case of mistaken identity: Are reactive oxygen species actually reactive sulfide species? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 310, R549–R560. [Google Scholar] [CrossRef]
- Labunskyy, V.M.; Hatfield, D.L.; Gladyshev, V.N. Selenoproteins: Molecular pathways and physiological roles. Physiol. Rev. 2014, 94, 739–777. [Google Scholar] [CrossRef] [PubMed]
- Carvajal Carvajal, C. Especies reactivas del oxígeno: Formación, funcion y estrés oxidativo. Med. Leg. Costa Rica 2019, 36, 91–100. [Google Scholar]
- Kumar, S.; Pandey, A.K. Free radicals: Health implications and their mitigation by herbals. Br. J. Med. Med. Res. 2015, 7, 438–457. [Google Scholar] [CrossRef]
- Wu, J.Q.; Kosten, T.R.; Zhang, X.Y. Free radicals, antioxidant defense systems, and schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 46, 200–206. [Google Scholar] [CrossRef]
- Taniyama, Y.; Griendling, K.K. Reactive oxygen species in the vasculature: Molecular and cellular mechanisms. Hypertension 2003, 42, 1075–1081. [Google Scholar] [CrossRef]
- Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
- Frei, B. Reactive oxygen species and antioxidant vitamins: Mechanisms of action. Am. J. Med. 1994, 97, S5–S13. [Google Scholar] [CrossRef]
- Nishida, N.; Arizumi, T.; Takita, M.; Kitai, S.; Yada, N.; Hagiwara, S.; Inoue, T.; Minami, Y.; Ueshima, K.; Sakurai, T.; et al. Reactive oxygen species induce epigenetic instability through the formation of 8-hydroxydeoxyguanosine in human hepatocarcinogenesis. Dig. Dis. 2013, 31, 459–466. [Google Scholar] [CrossRef]
- Yasui, M.; Kanemaru, Y.; Kamoshita, N.; Suzuki, T.; Arakawa, T.; Honma, M. Tracing the fates of site-specifically introduced DNA adducts in the human genome. DNA Repair 2014, 15, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Valko, M.; Izakovic, M.; Mazur, M.; Rhodes, C.J.; Telser, J. Role of oxygen radicals in DNA damage and cancer incidence. Mol. Cell. Biochem. 2004, 266, 37–56. [Google Scholar] [CrossRef]
- Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [PubMed]
- Halliwell, B. Role of free radicals in the neurodegenerative diseases: Therapeutic implications for antioxidant treatment. Drugs Aging 2001, 18, 685–716. [Google Scholar] [CrossRef] [PubMed]
- Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef] [PubMed]
- Galle, J. Oxidative stress in chronic renal failure. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2001, 16, 2135–2137. [Google Scholar] [CrossRef]
- MacNee, W. Oxidative stress and lung inflammation in airways disease. Eur. J. Pharmacol. 2001, 429, 195–207. [Google Scholar] [CrossRef]
- Interdonato, M.; Pizzino, G.; Bitto, A.; Galfo, F.; Irrera, N.; Mecchio, A.; Pallio, G.; Ramistella, V.; De Luca, F.; Santamaria, A.; et al. Cadmium delays puberty onset and testis growth in adolescents. Clin. Endocrinol. 2015, 83, 357–362. [Google Scholar] [CrossRef]
- Rotariu, D.; Babes, E.E.; Tit, D.M.; Moisi, M.; Bustea, C.; Stoicescu, M.; Radu, A.-F.; Vesa, C.M.; Behl, T.; Bungau, A.F.; et al. Oxidative stress—Complex pathological issues concerning the hallmark of cardiovascular and metabolic disorders. Biomed. Pharmacother. 2022, 152, 113238. [Google Scholar] [CrossRef]
- Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev. 2010, 4, 118–126. [Google Scholar] [CrossRef]
- Alkadi, H. A Review on Free Radicals and Antioxidants. Infect. Disord. Drug Targets 2020, 20, 16–26. [Google Scholar] [CrossRef] [PubMed]
- Von Dentz, K.E.; Silva, B.S.; Queiroz, E.A.I.F.; Bomfim, G.F.; Nascimento, A.F.; Sugizaki, M.M.; Luvizotto, R.A.M. Hibiscus sabdariffa ethanolic extract modulates adipokine levels, decreases visceral fat and improves glycemic profile in high-fat/sugar diet-induced obese rats. Nutr. Food Sci. 2021, 51, 222–233. [Google Scholar] [CrossRef]
- Rahal, A.; Kumar, A.; Singh, V.; Yadav, B.; Tiwari, R.; Chakraborty, S.; Dhama, K. Oxidative stress, prooxidants, and antioxidants: The interplay. Biomed. Res. Int. 2014, 2014, 761264. [Google Scholar] [CrossRef] [PubMed]
- Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019, 25, 101084. [Google Scholar] [CrossRef] [PubMed]
- Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef]
- Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative stress: A key modulator in neurodegenerative diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef]
- Senoner, T.; Dichtl, W. Oxidative Stress in Cardiovascular Diseases: Still a Therapeutic Target? Nutrients 2019, 11, 2090. [Google Scholar] [CrossRef]
- Alolga, R.N.; Wang, F.; Zhang, X.; Li, J.; Tran, L.S.P.; Yin, X. Bioactive Compounds from the Zingiberaceae Family with Known Antioxidant Activities for Possible Therapeutic Uses. Antioxidants 2022, 11, 1281. [Google Scholar] [CrossRef]
- Kang, Q.; Yang, C. Oxidative stress and diabetic retinopathy: Molecular mechanisms, pathogenetic role and therapeutic implications. Redox Biol. 2020, 37, 101799. [Google Scholar] [CrossRef]
- Romero, A.; Forero, M.; Sequeda-Castañeda, L.G.; Grismaldo, A.; Iglesias, J.; Celis-Zambrano, C.A.; Schuler, I.; Morales, L. Effect of ginger extract on membrane potential changes and AKT activation on a peroxide-induced oxidative stress cell model. J. King Saud Univ. Sci. 2018, 30, 263–269. [Google Scholar] [CrossRef]
- Carnuta, M.G.; Deleanu, M.; Barbalata, T.; Toma, L.; Raileanu, M.; Sima, A.V.; Stancu, C.S. Zingiber officinale extract administration diminishes steroyl-CoA desaturase gene expression and activity in hyperlipidemic hamster liver by reducing the oxidative and endoplasmic reticulum stress. Phytomedicine 2018, 48, 62–69. [Google Scholar] [CrossRef] [PubMed]
- Jalali, M.; Mahmoodi, M.; Moosavian, S.P.; Jalali, R.; Ferns, G.; Mosallanezhad, A.; Imanieh, M.H.; Mosallanezhad, Z. The effects of ginger supplementation on markers of inflammatory and oxidative stress: A systematic review and meta-analysis of clinical trials. Phyther. Res. 2020, 34, 1723–1733. [Google Scholar] [CrossRef] [PubMed]
- Sheikhhossein, F.; Borazjani, M.; Jafari, A.; Askari, M.; Vataniyan, E.; Gholami, F.; Amini, M.R. Effects of ginger supplementation on biomarkers of oxidative stress: A systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. ESPEN 2021, 45, 111–119. [Google Scholar] [CrossRef] [PubMed]
- Cinelli, M.A.; Do, H.T.; Miley, G.P.; Silverman, R.B. Inducible nitric oxide synthase: Regulation, structure, and inhibition. Med. Res. Rev. 2020, 40, 158–189. [Google Scholar] [CrossRef] [PubMed]
- Ippoushi, K.; Azuma, K.; Ito, H.; Horie, H.; Higashio, H. [6]-Gingerol inhibits nitric oxide synthesis in activated J774.1 mouse macrophages and prevents peroxynitrite-induced oxidation and nitration reactions. Life Sci. 2003, 73, 3427–3437. [Google Scholar] [CrossRef]
- Eun, M.K.; Hye, J.K.; Kim, S.; Woo, H.C.; Yeon, H.C.; Shi, Y.R.; Young, S.K.; Woo, S.K.; Park, S.Y. Modulation of macrophage functions by compounds isolated from Zingiber officinale. Planta Med. 2009, 75, 148–151. [Google Scholar] [CrossRef]
- Ballester, P.; Cerdá, B.; Arcusa, R.; Marhuenda, J.; Yamedjeu, K.; Zafrilla, P. Effect of Ginger on Inflammatory Diseases. Molecules 2022, 27, 7223. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Sun, X.; Jiang, L.; Liu, X.; Chen, M.; Yao, X.; Sun, Q.; Yang, G. 6-Gingerol induces autophagy to protect HUVECs survival from apoptosis. Chem. Biol. Interact. 2016, 256, 249–256. [Google Scholar] [CrossRef]
- Hsiang, C.Y.; Lo, H.Y.; Huang, H.C.; Li, C.C.; Wu, S.L.; Ho, T.Y. Ginger extract and zingerone ameliorated trinitrobenzene sulphonic acid-induced colitis in mice via modulation of nuclear factor-κB activity and interleukin-1β signalling pathway. Food Chem. 2013, 136, 170–177. [Google Scholar] [CrossRef]
- Han, S.J.; Kim, M.; D’Agati, V.D.; Lee, H.T. 6-Shogaol protects against ischemic acute kidney injury by modulating NF-κB and heme oxygenase-1 pathways. Am. J. Physiol. Renal Physiol. 2019, 317, F743–F756. [Google Scholar] [CrossRef]
- Mansingh, D.P.; Sunanda, O.J.; Sali, V.K.; Vasanthi, H.R. [6]-Gingerol-induced cell cycle arrest, reactive oxygen species generation, and disruption of mitochondrial membrane potential are associated with apoptosis in human gastric cancer (AGS) cells. J. Biochem. Mol. Toxicol. 2018, 32, e22206. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.J.; Jeon, Y.; Kim, T.; Lim, W.C.; Ham, J.; Park, Y.N.; Kim, T.J.; Ko, H. Combined treatment with zingerone and its novel derivative synergistically inhibits TGF-β1 induced epithelial-mesenchymal transition, migration and invasion of human hepatocellular carcinoma cells. Bioorg. Med. Chem. Lett. 2017, 27, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
- Zadorozhna, M.; Mangieri, D. Molecular Sciences Mechanisms of Chemopreventive and Therapeutic Proprieties of Ginger Extracts in Cancer. Int. J. Mol. Sci. 2021, 22, 6599. [Google Scholar] [CrossRef]
- Kim, S.D.; Kwag, E.B.; Yang, M.X.; Yoo, H.S. Efficacy and Safety of Ginger on the Side Effects of Chemotherapy in Breast Cancer Patients: Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2022, 23, 11267. [Google Scholar] [CrossRef] [PubMed]
- Maroufi, S.S.; Moradimajd, P.; Moosavi, S.A.A.; Imani, F.; Samaee, H.; Oguz, M. Dose Ginger Have Preventative Effects on PONV-Related Eye Surgery? A Clinical Trial. Anesthesiol. Pain Med. 2019, 9, e92072. [Google Scholar] [CrossRef]
- Sharifzadeh, F.; Kashanian, M.; Koohpayehzadeh, J.; Rezaian, F.; Sheikhansari, N.; Eshraghi, N. A comparison between the effects of ginger, pyridoxine (vitamin B6) and placebo for the treatment of the first trimester nausea and vomiting of pregnancy (NVP). J. Matern. Fetal. Neonatal Med. 2018, 31, 2509–2514. [Google Scholar] [CrossRef] [PubMed]
- Roudsari, N.M.; Lashgari, N.A.; Momtaz, S.; Roufogalis, B.; Abdolghaffari, A.H.; Sahebkar, A. Ginger: A complementary approach for management of cardiovascular diseases. Biofactors 2021, 47, 933–951. [Google Scholar] [CrossRef] [PubMed]
- El-Seweidy, M.M.; sarhan Amin, R.; Husseini Atteia, H.; El-Zeiky, R.R.; Al-gabri, N.A. Dyslipidemia induced inflammatory status, platelet activation and endothelial dysfunction in rabbits: Protective role of 10-Dehydrogingerdione. Biomed. Pharmacother. 2019, 110, 456–464. [Google Scholar] [CrossRef]
- Angelopoulou, E.; Paudel, Y.N.; Papageorgiou, S.G.; Piperi, C. Elucidating the Beneficial Effects of Ginger (Zingiber officinale Roscoe) in Parkinson’s Disease. ACS Pharmacol. Transl. Sci. 2022, 5, 838–848. [Google Scholar] [CrossRef]
- Moon, M.; Kim, H.G.; Choi, J.G.; Oh, H.; Lee, P.K.; Ha, S.K.; Kim, S.Y.; Park, Y.; Huh, Y.; Oh, M.S. 6-Shogaol, an active constituent of ginger, attenuates neuroinflammation and cognitive deficits in animal models of dementia. Biochem. Biophys. Res. Commun. 2014, 449, 8–13. [Google Scholar] [CrossRef]
- Reddy, P.H.; Manczak, M.; Yin, X.; Grady, M.C.; Mitchell, A.; Tonk, S.; Kuruva, C.S.; Bhatti, J.S.; Kandimalla, R.; Vijayan, M.; et al. Protective Effects of Indian Spice Curcumin Against Amyloid-β in Alzheimer’s Disease. J. Alzheimers. Dis. 2018, 61, 843–866. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, N.; Karobari, M.I.; Yousaf, A.; Mohamed, R.N.; Arshad, S.; Basheer, S.N.; Peeran, S.W.; Noorani, T.Y.; Assiry, A.A.; Alharbi, A.S.; et al. The Antimicrobial Efficacy Against Selective Oral Microbes, Antioxidant Activity and Preliminary Phytochemical Screening of Zingiber officinale. Infect. Drug Resist. 2022, 15, 2773. [Google Scholar] [CrossRef]
- Park, M.; Bae, J.; Lee, D.S. Antibacterial activity of [10]-gingerol and [12]-gingerol isolated from ginger rhizome against periodontal bacteria. Phyther. Res. 2008, 22, 1446–1449. [Google Scholar] [CrossRef] [PubMed]
- Zorofchian Moghadamtousi, S.; Abdul Kadir, H.; Hassandarvish, P.; Tajik, H.; Abubakar, S.; Zandi, K. A review on antibacterial, antiviral, and antifungal activity of curcumin. Biomed Res. Int. 2014, 2014, 186864. [Google Scholar] [CrossRef] [PubMed]
- Karuppiah, P.; Rajaram, S. Antibacterial effect of Allium sativum cloves and Zingiber officinale rhizomes against multiple-drug resistant clinical pathogens. Asian Pac. J. Trop. Biomed. 2012, 2, 597. [Google Scholar] [CrossRef]
- Gaschler, M.M.; Stockwell, B.R. Lipid peroxidation in cell death. Biochem. Biophys. Res. Commun. 2017, 482, 419–425. [Google Scholar] [CrossRef]
- Tan, B.L.; Norhaizan, M.E.; Liew, W.P.P.; Rahman, H.S. Antioxidant and oxidative stress: A mutual interplay in age-related diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef]
- Espinosa-Diez, C.; Miguel, V.; Mennerich, D.; Kietzmann, T.; Sánchez-Pérez, P.; Cadenas, S.; Lamas, S. Antioxidant responses and cellular adjustments to oxidative stress. Redox Biol. 2015, 6, 183–197. [Google Scholar] [CrossRef]
- Jakubczyk, K.; Drużga, A.; Katarzyna, J.; Skonieczna-Żydecka, K. Antioxidant Potential of Curcumin-A Meta-Analysis of Randomized Clinical Trials. Antioxidants 2020, 9, 1092. [Google Scholar] [CrossRef]
- Alizadeh, M.; Kheirouri, S. Curcumin reduces malondialdehyde and improves antioxidants in humans with diseased conditions: A comprehensive meta-analysis of randomized controlled trials. BioMedicine 2019, 9, 23. [Google Scholar] [CrossRef]
- Lee, J.H.; Song, M.Y.; Song, E.K.; Kim, E.K.; Moon, W.S.; Han, M.K.; Park, J.W.; Kwon, K.B.; Park, B.H. Overexpression of SIRT1 protects pancreatic β-cells against cytokine toxicity by suppressing the nuclear factor-κB signaling pathway. Diabetes 2009, 58, 344–351. [Google Scholar] [CrossRef] [PubMed]
- Lai, L.; Yan, L.; Gao, S.; Hu, C.L.; Ge, H.; Davidow, A.; Park, M.; Bravo, C.; Iwatsubo, K.; Ishikawa, Y.; et al. Type 5 adenylyl cyclase increases oxidative stress by transcriptional regulation of manganese superoxide dismutase via the SIRT1/FoxO3a pathway. Circulation 2013, 127, 1692–1701. [Google Scholar] [CrossRef] [PubMed]
- Qiu, X.; Brown, K.; Hirschey, M.D.; Verdin, E.; Chen, D. Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab. 2010, 12, 662–667. [Google Scholar] [CrossRef] [PubMed]
- Dehzad, M.J.; Ghalandari, H.; Nouri, M.; Askarpour, M. Antioxidant and anti-inflammatory effects of curcumin/turmeric supplementation in adults: A GRADE-assessed systematic review and dose–response meta-analysis of randomized controlled trials. Cytokine 2023, 164, 156144. [Google Scholar] [CrossRef]
- Panahi, Y.; Hosseini, M.S.; Khalili, N.; Naimi, E.; Majeed, M.; Sahebkar, A. Antioxidant and anti-inflammatory effects of curcuminoid-piperine combination in subjects with metabolic syndrome: A randomized controlled trial and an updated meta-analysis. Clin. Nutr. 2015, 34, 1101–1108. [Google Scholar] [CrossRef]
- Spagnuolo, C.; Napolitano, M.; Tedesco, I.; Moccia, S.; Milito, A.; Russo, G.L. Neuroprotective Role of Natural Polyphenols. Curr. Top. Med. Chem. 2016, 16, 1943–1950. [Google Scholar] [CrossRef]
- Tang, M.; Taghibiglou, C. The Mechanisms of Action of Curcumin in Alzheimer’s Disease. J. Alzheimers. Dis. 2017, 58, 1003–1016. [Google Scholar] [CrossRef]
- Belwal, T.; Nabavi, S.M.; Nabavi, S.F.; Dehpour, A.R.; Shirooie, S. Naturally Occurring Chemicals against Alzheimer’s Disease; Academic Press: Cambridge, MA, USA, 2020; ISBN 0128192135. [Google Scholar]
- He, Y.; Yue, Y.; Zheng, X.; Zhang, K.; Chen, S.; Du, Z. Curcumin, inflammation, and chronic diseases: How are they linked? Molecules 2015, 20, 9183–9213. [Google Scholar] [CrossRef]
- Peng, Y.; Ao, M.; Dong, B.; Jiang, Y.; Yu, L.; Chen, Z.; Hu, C.; Xu, R. Anti-Inflammatory Effects of Curcumin in the Inflammatory Diseases: Status, Limitations and Countermeasures. Drug Des. Devel. Ther. 2021, 15, 4503–4525. [Google Scholar] [CrossRef]
- Yousefian, M.; Shakour, N.; Hosseinzadeh, H.; Hayes, A.W.; Hadizadeh, F.; Karimi, G. The natural phenolic compounds as modulators of NADPH oxidases in hypertension. Phytomedicine 2019, 55, 200–213. [Google Scholar] [CrossRef]
- Lin, X.; Bai, D.; Wei, Z.; Zhang, Y.; Huang, Y.; Deng, H.; Huang, X. Curcumin attenuates oxidative stress in RAW264.7 cells by increasing the activity of antioxidant enzymes and activating the Nrf2-Keap1 pathway. PLoS ONE 2019, 14, e0216711. [Google Scholar] [CrossRef] [PubMed]
- Jamilian, M.; Foroozanfard, F.; Kavossian, E.; Aghadavod, E.; Shafabakhsh, R.; Hoseini, A.; Asemi, Z. Effects of curcumin on body weight, glycemic control and serum lipids in women with polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. ESPEN 2020, 36, 128–133. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Zou, J.; Li, P.; Zheng, X.; Feng, D. Curcumin Protects against Atherosclerosis in Apolipoprotein E-Knockout Mice by Inhibiting Toll-like Receptor 4 Expression. J. Agric. Food Chem. 2018, 66, 449–456. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.F.; Ching, L.C.; Huang, Y.C.; Chen, C.Y.; Chiang, A.N.; Kou, Y.R.; Shyue, S.K.; Lee, T.S. Molecular mechanism of curcumin on the suppression of cholesterol accumulation in macrophage foam cells and atherosclerosis. Mol. Nutr. Food Res. 2012, 56, 691–701. [Google Scholar] [CrossRef] [PubMed]
- Meiyanto, E.; Putri, H.; Arum Larasati, Y.; Yudi Utomo, R.; Istighfari Jenie, R.; Ikawati, M.; Lestari, B.; Yoneda-Kato, N.; Nakamae, I.; Kawaichi, M.; et al. Anti-proliferative and Anti-metastatic Potential of Curcumin Analogue, Pentagamavunon-1 (PGV-1), Toward Highly Metastatic Breast Cancer Cells in Correlation with ROS Generation. Adv. Pharm. Bull. 2019, 9, 445–452. [Google Scholar] [CrossRef]
- Mansouri, K.; Rasoulpoor, S.; Daneshkhah, A.; Abolfathi, S.; Salari, N.; Mohammadi, M.; Rasoulpoor, S.; Shabani, S. Clinical effects of curcumin in enhancing cancer therapy: A systematic review. BMC Cancer 2020, 20, 791. [Google Scholar] [CrossRef]
- Rai, D.; Singh, J.K.; Roy, N.; Panda, D. Curcumin inhibits FtsZ assembly: An attractive mechanism for its antibacterial activity. Biochem. J. 2008, 410, 147–155. [Google Scholar] [CrossRef]
- Sharma, M.; Manoharlal, R.; Puri, N.; Prasad, R. Antifungal curcumin induces reactive oxygen species and triggers an early apoptosis but prevents hyphae development by targeting the global repressor TUP1 in Candida albicans. Biosci. Rep. 2010, 30, 391–404. [Google Scholar] [CrossRef]
- Saeed, A.; Sultana, B.; Anwar, F.; Mushtaq, M.; Alkharfy, K.M.; Gilani, A.-H. Antioxidant and antimutagenic potential of seeds and pods of green cardamom (Elettaria cardamomum). Int. J. Pharmacol. 2014, 10, 461–469. [Google Scholar] [CrossRef]
- Bhaswant, M.; Poudyal, H.; Mathai, M.L.; Ward, L.C.; Mouatt, P.; Brown, L. Green and black cardamom in a diet-induced rat model of metabolic syndrome. Nutrients 2015, 7, 7691–7707. [Google Scholar] [CrossRef]
- Jamil, B.; Abbasi, R.; Abbasi, S.; Imran, M.; Khan, S.U.; Ihsan, A.; Javed, S.; Bokhari, H.; Imran, M. Encapsulation of Cardamom Essential Oil in Chitosan Nano-composites: In-vitro Efficacy on Antibiotic-Resistant Bacterial Pathogens and Cytotoxicity Studies. Front. Microbiol. 2016, 7, 1580. [Google Scholar] [CrossRef] [PubMed]
- Preedy, V.R.; Watson, R.R. Nuts and Seeds in Health and Disease Prevention; Academic Press: Cambridge, MA, USA, 2020; ISBN 0128216107. [Google Scholar]
- Cui, H.; Zhang, C.; Li, C.; Lin, L. Inhibition mechanism of cardamom essential oil on methicillin-resistant Staphylococcus aureus biofilm. LWT 2020, 122, 109057. [Google Scholar] [CrossRef]
- GiaccoF, B. Oxidativestressanddiabetic complications. Circ. Res. 2010, 107, 1058. [Google Scholar]
- Kazemi, S.; Yaghooblou, F.; Siassi, F.; Rahimi Foroushani, A.; Ghavipour, M.; Koohdani, F.; Sotoudeh, G. Cardamom supplementation improves inflammatory and oxidative stress biomarkers in hyperlipidemic, overweight, and obese pre-diabetic women: A randomized double-blind clinical trial. J. Sci. Food Agric. 2017, 97, 5296–5301. [Google Scholar] [CrossRef]
- Savini, I.; Catani, M.V.; Evangelista, D.; Gasperi, V.; Avigliano, L. Obesity-associated oxidative stress: Strategies finalized to improve redox state. Int. J. Mol. Sci. 2013, 14, 10497–10538. [Google Scholar] [CrossRef]
- Higuchi, M.; Dusting, G.J.; Peshavariya, H.; Jiang, F.; Hsiao, S.T.-F.; Chan, E.C.; Liu, G.-S. Differentiation of human adipose-derived stem cells into fat involves reactive oxygen species and Forkhead box O1 mediated upregulation of antioxidant enzymes. Stem Cells Dev. 2013, 22, 878–888. [Google Scholar] [CrossRef]
- Manna, P.; Jain, S.K. Obesity, oxidative stress, adipose tissue dysfunction, and the associated health risks: Causes and therapeutic strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef]
- Daneshi-Maskooni, M.; Keshavarz, S.A.; Mansouri, S.; Qorbani, M.; Alavian, S.M.; Badri-Fariman, M.; Jazayeri-Tehrani, S.A.; Sotoudeh, G. The effects of green cardamom on blood glucose indices, lipids, inflammatory factors, paraxonase-1, sirtuin-1, and irisin in patients with nonalcoholic fatty liver disease and obesity: Study protocol for a randomized controlled trial. Trials 2017, 18, 260. [Google Scholar] [CrossRef]
- Daneshi-Maskooni, M.; Keshavarz, S.A.; Qorbani, M.; Mansouri, S.; Alavian, S.M.; Badri-Fariman, M.; Jazayeri-Tehrani, S.A.; Sotoudeh, G. Green cardamom increases Sirtuin-1 and reduces inflammation in overweight or obese patients with non-alcoholic fatty liver disease: A double-blind randomized placebo-controlled clinical trial. Nutr. Metab. 2018, 15, 63. [Google Scholar] [CrossRef]
- Daneshi-Maskooni, M.; Keshavarz, S.A.; Qorbani, M.; Mansouri, S.; Alavian, S.M.; Badri-Fariman, M.; Jazayeri-Tehrani, S.A.; Sotoudeh, G. Green cardamom supplementation improves serum irisin, glucose indices, and lipid profiles in overweight or obese non-alcoholic fatty liver disease patients: A double-blind randomized placebo-controlled clinical trial. BMC Complement. Altern. Med. 2019, 19, 59. [Google Scholar] [CrossRef]
- Aghasi, M.; Ghazi-Zahedi, S.; Koohdani, F.; Siassi, F.; Nasli-Esfahani, E.; Keshavarz, A.; Qorbani, M.; Khoshamal, H.; Salari-Moghaddam, A.; Sotoudeh, G. The effects of green cardamom supplementation on blood glucose, lipids profile, oxidative stress, sirtuin-1 and irisin in type 2 diabetic patients: A study protocol for a randomized placebo-controlled clinical trial. BMC Complement. Altern. Med. 2018, 18, 18. [Google Scholar] [CrossRef] [PubMed]
- Cheshmeh, S.; Ghayyem, M.; Khamooshi, F.; Heydarzadeh, N.; Hojati, N.; Mosaieby, E.; Moradi, S.; Pasdar, Y. Green Cardamom Plus Low Calorie Diet Can Decrease Inflammatory Genes Expression among Obese Women with Poly Cystic Ovarian Syndrome, A Double Blind Randomized Clinical Trial. Eat Weight Disord 2021, 27, 821–830. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Ding, Y.; Ramprasath, T.; Zou, M.-H. Oxidative stress, GTPCH1, and endothelial nitric oxide synthase uncoupling in hypertension. Antioxid. Redox Signal. 2021, 34, 750–764. [Google Scholar] [CrossRef] [PubMed]
- Verma, S.K.; Jain, V.; Katewa, S.S. Blood pressure lowering, fibrinolysis enhancing and antioxidant activities of cardamom (Elettaria cardamomum). Indian J. Biochem. Biophys. 2009, 46, 503–506. [Google Scholar]
- Qiblawi, S.; Dhanarasu, S. Chemopreventive effect of cardamom (Elettaria cardamomum L.) against benzo (α) pyrene-induced forestomach papillomagenesis in swiss albino mice. J. Environ. Pathol. Toxicol. Oncol. 2015, 34, 95–104. [Google Scholar] [CrossRef]
- Bhat, J.; Damle, A.; Vaishnav, P.P.; Albers, R.; Joshi, M.; Banerjee, G. In vivo enhancement of natural killer cell activity through tea fortified with Ayurvedic herbs. Phyther. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2010, 24, 129–135. [Google Scholar]
- Hunt, R.; Dienemann, J.; Norton, H.J.; Hartley, W.; Hudgens, A.; Stern, T.; Divine, G. Aromatherapy as treatment for postoperative nausea: A randomized trial. Anesth. Analg. 2013, 117, 597–604. [Google Scholar] [CrossRef]
- Khatiban, M.; Mirzaie, M.; Fazeli, A.; Tapak, L.; Khalili, Z. Effect of Cardamom Inhalation Therapy on Intra-and Postoperative Nausea and Vomiting of Mothers Undergoing Spinal Anesthesia for Elective Cesarean Section. J. PeriAnesthesia Nurs. 2022, 37, 452–457. [Google Scholar] [CrossRef]
- Teneva, D.; Denkova, Z.; Denkova, R.; Atanasova, T.; Nenov, N.; Merdzhanov, P. Antimicrobial activity of essential oils and extracts from black pepper, cumin, coriander and cardamom against some pathogenic and saprophytic microorganisms. Bulg. J. Vet. Med. 2015, 18, 373–377. [Google Scholar] [CrossRef]
- Paul, K.; Ganguly, U.; Chakrabarti, S.; Bhattacharjee, P. Is 1,8-Cineole-Rich Extract of Small Cardamom Seeds More Effective in Preventing Alzheimer’s Disease than 1,8-Cineole Alone? Neuromol. Med. 2020, 22, 150–158. [Google Scholar] [CrossRef]
- Gomaa, A.A.; Makboul, R.M.; El-Mokhtar, M.A.; Abdel-Rahman, E.A.; Ahmed, I.A.; Nicola, M.A. Terpenoid-rich Elettaria cardamomum extract prevents Alzheimer-like alterations induced in diabetic rats via inhibition of GSK3β activity, oxidative stress and pro-inflammatory cytokines. Cytokine 2019, 113, 405–416. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ballester, P.; Cerdá, B.; Arcusa, R.; García-Muñoz, A.M.; Marhuenda, J.; Zafrilla, P. Antioxidant Activity in Extracts from Zingiberaceae Family: Cardamom, Turmeric, and Ginger. Molecules 2023, 28, 4024. https://doi.org/10.3390/molecules28104024
Ballester P, Cerdá B, Arcusa R, García-Muñoz AM, Marhuenda J, Zafrilla P. Antioxidant Activity in Extracts from Zingiberaceae Family: Cardamom, Turmeric, and Ginger. Molecules. 2023; 28(10):4024. https://doi.org/10.3390/molecules28104024
Chicago/Turabian StyleBallester, Pura, Begoña Cerdá, Raúl Arcusa, Ana María García-Muñoz, Javier Marhuenda, and Pilar Zafrilla. 2023. "Antioxidant Activity in Extracts from Zingiberaceae Family: Cardamom, Turmeric, and Ginger" Molecules 28, no. 10: 4024. https://doi.org/10.3390/molecules28104024
APA StyleBallester, P., Cerdá, B., Arcusa, R., García-Muñoz, A. M., Marhuenda, J., & Zafrilla, P. (2023). Antioxidant Activity in Extracts from Zingiberaceae Family: Cardamom, Turmeric, and Ginger. Molecules, 28(10), 4024. https://doi.org/10.3390/molecules28104024