3,184
Views
0
CrossRef citations to date
0
Altmetric
Review Article

An updated comprehensive review of the therapeutic properties of Chamomile (Matricaria chamomilla L.)

, , , , , , , & show all
Pages 133-164 | Received 23 Aug 2023, Accepted 07 Dec 2023, Published online: 21 Dec 2023

References

  • El Joumaa, M. M.; Borjac, J. M. Matricaria Chamomilla: A Valuable Insight into Recent Advances in Medicinal Uses and Pharmacological Activities. Phytochem. Rev. 2022, 21(6), 1913–1940. DOI: 10.1007/s11101-022-09817-0.
  • Marković, M.; Pljevljakušić, D.; Kojičić, K.; Cupara, S. Ethnopharmacological Application of Chamomile (Matricaria Chamomilla L.) in the Pirot County of Southeastern Serbia. Arh. Farm. (Belgr). 2020, 70(4), 238–247. DOI: 10.5937/arhfarm2004238M.
  • Singh, O.; Khanam, Z.; Misra, N.; Srivastava, M. Chamomile (Matricaria chamomilla L.): An overview. Pharmacogn. Rev. 2011, 5(9), 82. DOI: 10.4103/0973-7847.79103.
  • Sharifi-Rad, M.; Nazaruk, J.; Polito, L.; Morais-Braga, M. F. B.; Rocha, J. E.; Coutinho, H. D. M.; Salehi, B.; Tabanelli, G.; Montanari, C.; Del Mar Contreras, M., et al. Matricaria Genus as a Source of Antimicrobial Agents: From Farm to Pharmacy and Food Applications. Microbiol. Res. 2018, 215, 76–88. DOI: 10.1016/j.micres.2018.06.010.
  • Chauhan, R.; Singh, S.; Kumar, V.; Kumar, A.; Kumari, A.; Rathore, S.; Kumar, R.; Singh, S. A Comprehensive Review on Biology, Genetic Improvement, Agro and Process Technology of German Chamomile (Matricaria Chamomilla L.). Plants. 2021, 11(1), 29. DOI: 10.3390/plants11010029.
  • Srivastava, J. K.; Shankar, E.; Gupta, S. Chamomile: A Herbal Medicine of the Past with a Bright Future (Review). Mol. Med. Rep. 2010, 3(6), 895–901. DOI: 10.3892/mmr.2010.377.
  • Kyokong, O.; Charuluxananan, S.; Muangmingsuk, V.; Rodanant, O.; Subornsug, K.; Punyasang, W. Efficacy of Chamomile-Extract Spray for Prevention of Post-Operative Sore Throat. J. Med. Assoc. Thai. 2002, 85(1), S180–5.
  • Togar, B.; Turkez, H.; Hacimuftuoglu, A.; Tatar, A.; Geyikoglu, F. Guaiazulene: biochemical activity and cytotoxic and genotoxic effects on rat neuron and N2a neuroblastom cells. J. Intercult. Ethnopharmacol. 2015, 4(1), 29. DOI: 10.5455/jice.20141124062203.
  • Attia, M.; Kim, S.-U.; Ro, D.-K. Molecular Cloning and Characterization of (+)-Epi-α-Bisabolol Synthase, Catalyzing the First Step in the Biosynthesis of the Natural Sweetener, Hernandulcin, in Lippia Dulcis. Arch. Biochem. Biophys. 2012, 527(1), 37–44. DOI: 10.1016/j.abb.2012.07.010.
  • Eddin, L. B.; Jha, N. K.; Goyal, S. N.; Agrawal, Y. O.; Subramanya, S. B.; Bastaki, S. M. A.; Ojha, S. Health Benefits, Pharmacological Effects, Molecular Mechanisms, and Therapeutic Potential of α-Bisabolol. Nutrients. 2022, 14(7), 1370. DOI: 10.3390/nu14071370.
  • Farias, K. S.; Kato, N. N.; Boaretto, A. G.; Weber, J. I.; Brust, F. R.; Alves, F. M.; Tasca, T.; Macedo, A. J.; Silva, D. B.; Carollo, C. A. Nectandra as a Renewable Source for (+)-α-Bisabolol, an Antibiofilm and Anti-Trichomonas Vaginalis Compound. Fitoterapia. 2019, 136, 104179. DOI: 10.1016/j.fitote.2019.104179.
  • Kamatou, G. P. P.; Viljoen, A. M. A Review of the Application and Pharmacological Properties of α -Bisabolol and α -Bisabolol-Rich Oils. J. Am. Oil Chem. Soc. 2010, 87(1), 1–7. DOI: 10.1007/s11746-009-1483-3.
  • Aziz, Z. A. A.; Ahmad, A.; Setapar, S. H. M.; Karakucuk, A.; Azim, M. M.; Lokhat, D.; Rafatullah, M.; Ganash, M.; Kamal, M. A.; Ashraf, G. M. Essential Oils: Extraction Techniques, Pharmaceutical And Therapeutic Potential - A Review. Curr. Drug Metab. 2018, 19(13), 1100–1110. DOI: 10.2174/1389200219666180723144850.
  • Deligeorgopoulou, A.; Allemann, R. K. Evidence for Differential Folding of Farnesyl Pyrophosphate in the Active Site of Aristolochene Synthase: A Single-Point Mutation Converts Aristolochene Synthase into an (E)-β-Farnesene Synthase. Biochemistry. 2003, 42(25), 7741–7747. DOI: 10.1021/bi034410m.
  • Amara, U.; Mashwani, Z. U. R.; Khan, A.; Laraib, S.; Wali, R.; Sarwar, U.; Ain, Q. T.; Shakeel, S.; Rahimullah, S. Conservation Status and Therapeutic Potential of Saussurea lappa: An Overview. AJPS. 2017, 08(3), 602–614. DOI: 10.4236/ajps.2017.83041.
  • Telascrea, M.; de Araújo, C. C.; Marques, M. O. M.; Facanali, R.; de Moraes, P. L. R.; Cavalheiro, A. J. Essential Oil from Leaves of Cryptocarya Mandioccana Meisner (Lauraceae): Composition and Intraspecific Chemical Variability. Biochem. Syst. Ecol. 2007, 35(4), 222–232. DOI: 10.1016/j.bse.2006.09.015.
  • Fahlbusch, K.; Hammerschmidt, F.; Panten, J.; Pickenhagen, W.; Schatkowski, D.; Bauer, K.; Garbe, D.; Surburg, H. Flavors and Fragrances. In Ullmann’s Encyclopedia of Industrial Chemistry; Wiley, 2003; pp. 1131–1145. DOI:10.1002/14356007.a11_141.
  • Mahdavi, B.; Ghezi, S.; Maleki, B. Chemical Composition and Bioactivity of Essential Oil and Extracts of Salvia Limbata C.A. Mey (Lamiaceae). Curr. Bioact. Compd. 2020, 16(9), 1299–1305. DOI: 10.2174/1573407216666200218111906.
  • Salehi, A.; Hazrati, S. How Essential Oil Content and Composition Fluctuate in German Chamomile Flowers During the Day? J. Essent. Oil Bear. Plants. 2017, 20(3), 622–631. DOI: 10.1080/0972060X.2017.1351895.
  • Flemming, M.; Kraus, B.; Rascle, A.; Jürgenliemk, G.; Fuchs, S.; Fürst, R.; Heilmann, J. Revisited Anti-Inflammatory Activity of Matricine in Vitro: Comparison with Chamazulene. Fitoterapia. 2015, 106, 122–128. DOI: 10.1016/j.fitote.2015.08.010.
  • Héthelyi, É.; Dános, B.; Tétényi, P. Phytochemical Studies on the Essential Oils of Species Belonging to theAchillea Genus by Gas Chromatography/Mass Spectrometry. Biol. Mass Spectrom. 1989, 18(8), 629–636. DOI: 10.1002/bms.1200180821.
  • Stanojevic, L. P.; Marjanovic-Balaban, Z. R.; Kalaba, V. D.; Stanojevic, J. S.; Cvetkovic, D. J. Chemical Composition, Antioxidant and Antimicrobial Activity of Chamomile Flowers Essential Oil (Matricaria Chamomilla L.). J. Essent. Oil Bear. Plants. 2016, 19(8), 2017–2028. DOI: 10.1080/0972060X.2016.1224689.
  • Rauter, A. P.; Ennis, M.; Hellwich, K.-H.; Herold, B. J.; Horton, D.; Moss, G. P.; Schomburg, I. Nomenclature of flavonoids (IUPAC Recommendations 2017). Pure Appl. Chem. 2018, 90(9), 1429–1486. DOI: 10.1515/pac-2013-0919.
  • Redaelli, C.; Formentini, L.; Santaniello, E. Apigenin 7-Glucoside and Its 2″- and 6″-Acetates from Ligulate Flowers of Matricaria Chamomilla. Phytochemistry. 1980, 19(5), 985–986. DOI: 10.1016/0031-9422(80)85160-0.
  • Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.; Novellino, E., et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019, 20(6), 1305. DOI: 10.3390/ijms20061305.
  • Chen, W.; Viljoen, A. M. Geraniol — a Review of a Commercially Important Fragrance Material. South African J. Bot. 2010, 76(4), 643–651. DOI: 10.1016/j.sajb.2010.05.008.
  • Farkoosh, S. S.; Ardakani, M. R. R.; Darzi, F.; Faregh, M. T. Effect of Mycorrhizal Symbiosis and Bacillus Coagolance on Qualitative and Quantitative Traits of Matricaria Chamomilla Under Different Levels of Phosphorus. Middle-East J. Sci. Res. 2011, 8, 01–09.
  • Yonzon, M.; Lee, D. J.; Yokochi, T.; Kawano, Y.; Nakahara, T. Antimicrobial Activities of Essential Oils of Nepal. J. Essent. Oil Res. 2005, 17(1), 107–111. DOI: 10.1080/10412905.2005.9698846.
  • Ekiert, H.; Świątkowska, J.; Knut, E.; Klin, P.; Rzepiela, A.; Tomczyk, M.; Szopa, A. Artemisia Dracunculus (Tarragon): A Review of Its Traditional Uses, Phytochemistry and Pharmacology. Front. Pharmacol. 2021, 12, 383–426. DOI: 10.3389/fphar.2021.653993.
  • Molnar, M.; Mendešević, N.; Šubarić, D.; Banjari, I.; Jokić, S. Comparison of Various Techniques for the Extraction of Umbelliferone and Herniarin in Matricaria Chamomilla Processing Fractions. Chem. Cent. J. 2017, 11(1), 78. DOI: 10.1186/s13065-017-0308-y.
  • Mazimba, O. Umbelliferone: Sources, Chemistry and Bioactivities Review. Bull. Fac. Pharmacy, Cairo Univ. 2017, 55(2), 223–232. DOI: 10.1016/j.bfopcu.2017.05.001.
  • Ionita, R.; Postu, P. A.; Mihasan, M.; Gorgan, D. L.; Hancianu, M.; Cioanca, O.; Hritcu, L. Ameliorative Effects of Matricaria Chamomilla L. Hydroalcoholic Extract on Scopolamine-Induced Memory Impairment in Rats: A Behavioral and Molecular Study. Phytomedicine. 2018, 47, 113–120. DOI: 10.1016/j.phymed.2018.04.049.
  • Nováková, L.; Vildová, A.; Mateus, J. P.; Gonçalves, T.; Solich, P. Development and Application of UHPLC–MS/MS Method for the Determination of Phenolic Compounds in Chamomile Flowers and Chamomile Tea Extracts. Talanta. 2010, 82(4), 1271–1280. DOI: 10.1016/j.talanta.2010.06.057.
  • Plazas, M.; Andújar, I.; Vilanova, S.; Hurtado, M.; Gramazio, P.; Herraiz, F. J.; Prohens, J. Breeding for Chlorogenic Acid Content in Eggplant: Interest and Prospects. Not. Bot. Horti Agrobot. Cluj-Napoca. 2013, 41(1), 26. DOI: 10.15835/nbha4119036.
  • Espíndola, K. M. M.; Ferreira, R. G.; Narvaez, L. E. M.; Silva Rosario, A. C. R.; da Silva, A. H. M.; Silva, A. G. B.; Vieira, A. P. O.; Monteiro, M. C. Chemical and Pharmacological Aspects of Caffeic Acid and Its Activity in Hepatocarcinoma. Front. Oncol. 2019, 9, 67–77. DOI: 10.3389/fonc.2019.00541.
  • Colunga Biancatelli, R. M. L.; Berrill, M.; Catravas, J. D.; Marik, P. E. Quercetin and Vitamin C: An Experimental, Synergistic Therapy for the Prevention and Treatment of SARS-CoV-2 Related Disease (COVID-19). Front. Immunol. 2020, 11, 231–255. DOI: 10.3389/fimmu.2020.01451.
  • Jucá, M. M.; Cysne Filho, F. M. S.; de Almeida, J. C.; Mesquita, D. D. S.; Barriga, J. R. D. M.; Dias, K. C. F.; Barbosa, T. M.; Vasconcelos, L. C.; Leal, L. K. A. M.; Ribeiro, J. E., et al. Flavonoids: biological activities and therapeutic potential. Nat. Prod. Res. 2020, 34(5), 692–705. DOI: 10.1080/14786419.2018.1493588.
  • Ganeshpurkar, A.; Saluja, A. K. The Pharmacological Potential of Rutin. Saudi Pharm. J. 2017, 25(2), 149–164. DOI: 10.1016/j.jsps.2016.04.025.
  • Ma, Y.; Li, W.; Mai, J.; Wang, J.; Wei, Y.; Ledesma-Amaro, R.; Ji, X.-J. Engineering Yarrowia Lipolytica for Sustainable Production of the Chamomile Sesquiterpene (−)-α-Bisabolol. Green Chem. 2021, 23(2), 780–787. DOI: 10.1039/D0GC03180A.
  • Hajaji, S.; Sifaoui, I.; López-Arencibia, A.; Reyes-Batlle, M.; Jiménez, I. A.; Bazzocchi, I. L.; Valladares, B.; Akkari, H.; Lorenzo-Morales, J.; Piñero, J. E. Leishmanicidal Activity of α-Bisabolol from Tunisian Chamomile Essential Oil. Parasitol. Res. 2018, 117(9), 2855–2867. DOI: 10.1007/s00436-018-5975-7.
  • Shanmuganathan, B.; Suryanarayanan, V.; Sathya, S.; Narenkumar, M.; Singh, S. K.; Ruckmani, K.; Pandima Devi, K. Anti-amyloidogenic and anti-apoptotic effect of α-bisabolol against Aβ induced neurotoxicity in PC12 cells. Eur. J. Med. Chem. 2018, 143, 1196–1207. DOI: 10.1016/j.ejmech.2017.10.017.
  • Nagoor Meeran, M. F.; Azimullah, S.; Laham, F.; Tariq, S.; Goyal, S. N.; Adeghate, E.; Ojha, S. α-Bisabolol Protects Against β-Adrenergic Agonist-Induced Myocardial Infarction in Rats by Attenuating Inflammation, Lysosomal Dysfunction, NLRP3 Inflammasome Activation and Modulating Autophagic Flux. Food Funct. 2020, 11(1), 965–976. DOI: 10.1039/C9FO00530G.
  • Baylac, S. Inhibition of 5-Lipoxygenase by Essential Oils and Other Natural Fragrant Extracts. Int. J. Aromather. 2003, 13(2–3), 138–142. DOI: 10.1016/S0962-4562(03)00083-3.
  • Shi, T.; Li, Y.; Zhu, L.; Tong, Y.; Yang, J.; Fang, Y.; Wang, M.; Zhang, J.; Jiang, Y.; Yang, S. Engineering the Oleaginous Yeast Yarrowia Lipolytica for β‐Farnesene Overproduction. Biotechnol. J. 2021, 16(7), 63. DOI: 10.1002/biot.202100097.
  • Brantner, A. H.; Pfeifhofer, H. W.; Ercegovac, O.; Males, Z.; Plazibat, M. Essential Oil Composition and Antioxidant Activity ofThymus Bracteosus Vis. Ex Benth. Flavour Fragr. J. 2005, 20(6), 596–600. DOI: 10.1002/ffj.1494.
  • Qadir, M.; Maurya, A. K.; Agnihotri, V. K.; Shah, W. A. Volatile Composition, Antibacterial and Antioxidant Activities of Artemisia Tournefortiana Reichb. from Kashmir, India. Nat. Prod. Res. 2021, 35(1), 152–156. DOI: 10.1080/14786419.2019.1613990.
  • Oliveira, A.; França, H.; Kuster, R.; Teixeira, L.; Rocha, L. Chemical Composition and Antibacterial Activity of Brazilian Propolis Essential Oil. J. Venom. Anim. Toxins Incl. Trop. Dis. 2010, 16(1), 121–130. DOI: 10.1590/S1678-91992010005000007.
  • Qin, Y.; Yang, Z.; Song, D.; Wang, Q.; Gu, S.; Li, W.; Duan, H.; Zhou, J.; Yang, X. Bioactivities of Synthetic Salicylate-Substituted Carboxyl (E)-β-Farnesene Derivatives as Ecofriendly Agrochemicals and Their Binding Mechanism with Potential Targets in Aphid Olfactory System. Pest Manag. Sci. 2020, 76(7), 2465–2472. DOI: 10.1002/ps.5787.
  • Zhang, J.-P.; Qin, Y.-G.; Dong, Y.-W.; Song, D.-L.; Duan, H.-X.; Yang, X.-L. Synthesis and Biological Activities of (E)- β -Farnesene Analogues Containing 1,2,3-Thiadiazole. Chinese Chem. Lett. 2017, 28(2), 372–376. DOI: 10.1016/j.cclet.2016.10.030.
  • Qin, Y.-G.; Yang, Z.-K.; Zhou, J.-J.; Zhang, S.-Y.; Pan, S.-X.; Liu, Y.; Gu, S.-H.; Duan, H.-X.; Yang, X.-L. Effects of Carboxyl and Acylamino Linkers in Synthetic Derivatives of Aphid Alarm Pheromone (E)-β-Farnesene on Repellent, Binding and Aphicidal Activity. J. Mol. Struct. 2022, 1268, 133658. DOI: 10.1016/j.molstruc.2022.133658.
  • Juteau, F.; Masotti, V.; Bessière, J. M.; Dherbomez, M.; Viano, J. Antibacterial and Antioxidant Activities of Artemisia Annua Essential Oil. Fitoterapia. 2002, 73(6), 532–535. DOI: 10.1016/S0367-326X(02)00175-2.
  • Montanari, R. M.; Barbosa, L. C. A.; Demuner, A. J.; Silva, C. J.; Carvalho, L. S.; Andrade, N. J. Chemical Composition and Antibacterial Activity of Essential Oils from Verbenaceae Species: Alternative Sources of (E)-Caryophyllene and Germacrene-D. Quim. Nova. 2011, 34(9), 1550–1555. DOI: 10.1590/S0100-40422011000900013.
  • Laouer, H.; Yabrir, B.; Djeridane, A.; Yousfi, M.; Beldovini, N.; Lamamra, M. Composition, Antioxidant and Antimicrobial Activities of the Essential Oil of Marrubium Deserti. Nat. Prod. Commun. 2009, 4(8), 1934578X0900400. DOI: 10.1177/1934578X0900400824.
  • Schepetkin, I.; Özek, G.; Özek, T.; Kirpotina, L.; Khlebnikov, A.; Quinn, M. Chemical Composition and Immunomodulatory Activity of Hypericum Perforatum Essential Oils. Biomolecules. 2020, 10(6), 916. DOI: 10.3390/biom10060916.
  • de Veras, B. O.; de Oliveira, J. R. S.; de Menezes Lima, V. L.; Do Amaral Ferraz Navarro, D. M.; de Oliveira Farias de Aguiar, J. C. R.; de Medeiros Moura, G. M.; da Silva, J. W.; de Assis, C. R. D.; Gorlach-Lira, K.; de Assis, P. A. C., et al. The Essential Oil of the Leaves of Verbesina Macrophylla (Cass.) S.F.Blake Has Antimicrobial, Anti-Inflammatory and Antipyretic Activities and is Toxicologically Safe. J. Ethnopharmacol. 2021, 265, 113248. DOI: 10.1016/j.jep.2020.113248.
  • Pripdeevech, P.; Saansoomchai, J. Antibacterial Activity and Chemical Composition of Essential Oil and Various Extracts of Fagraea Fragrans Roxb. Flowers. Chiang Mai J. Sci. 2013, 40, 214–223.
  • Solymosi, P. Herbicidal Activity of Chamazulene. Növényvédelem. 2000, 36, 119–123. DOI: 10.1016/B978-0-12-818188-1.00031-1.
  • Akram, W.; Tagde, P.; Ahmed, S.; Arora, S.; Emran, T. B.; Babalghith, A. O.; Sweilam, S. H.; Simal-Gandara, J. Guaiazulene and Related Compounds: A Review of Current Perspective on Biomedical Applications. Life. sci. 2023, 316, 121389. DOI: 10.1016/j.lfs.2023.121389.
  • Ornano, L.; Venditti, A.; Ballero, M.; Sanna, C.; Quassinti, L.; Bramucci, M.; Lupidi, G.; Papa, F.; Vittori, S.; Maggi, F.; et al. Chemopreventive and Antioxidant Activity of the Chamazulene‐Rich Essential Oil Obtained from Artemisia Arborescens L. Growing on the Isle of La Maddalena, Sardinia, Italy. Chem. Biodivers. 2013, 10(8), 1464–1474.
  • Capuzzo, A.; Occhipinti, A.; Maffei, M. E. Antioxidant and Radical Scavenging Activities of Chamazulene. Nat. Prod. Res. 2014, 28(24), 2321–2323. DOI: 10.1080/14786419.2014.931393.
  • Safayhi, H.; Sabieraj, J.; Sailer, E.-R.; Ammon, H. Chamazulene: An Antioxidant-Type Inhibitor of Leukotriene B 4 Formation. Planta. med. 1994, 60(5), 410–413. DOI: 10.1055/s-2006-959520.
  • Ali, F.; Rahul, N.; Jyoti, F.; Siddique, S. Health Functionality of Apigenin: A Review. Int. J. Food. Prop. 2017, 20(6), 1197–1238. DOI: 10.1080/10942912.2016.1207188.
  • Lapchak, P. A.; Boitano, P. D. Effect of the Pleiotropic Drug CNB-001 on Tissue Plasminogen Activator (tPa) Protease Activity in Vitro: Support for Combination Therapy to Treat Acute Ischemic Stroke. J. Neurol. Neurophysiol. 2014, 5, 1–9. DOI: 10.1155/2014/525141.
  • Lee, J.-H.; Zhou, H. Y.; Cho, S. Y.; Kim, Y. S.; Lee, Y. S.; Jeong, C. S. Anti-Inflammatory Mechanisms of Apigenin: Inhibition of Cyclooxygenase-2 Expression, Adhesion of Monocytes to Human Umbilical Vein Endothelial Cells, and Expression of Cellular Adhesion Molecules. Arch. Pharm. Res. 2007, 30(10), 1318–1327. DOI: 10.1007/BF02980273.
  • Telange, D. R.; Patil, A. T.; Pethe, A. M.; Fegade, H.; Anand, S.; Dave, V. S. Formulation and Characterization of an Apigenin-Phospholipid Phytosome (APLC) for Improved Solubility, in vivo Bioavailability, and Antioxidant Potential. Eur. J. Pharm. Sci. 2017, 108, 36–49. DOI: 10.1016/j.ejps.2016.12.009.
  • Miguel, F. G.; Cavalheiro, A. H.; Spinola, N. F.; Ribeiro, D. L.; Barcelos, G. R. M.; Antunes, L. M. G.; Hori, J. I.; Marquele-Oliveira, F.; Rocha, B. A.; Berretta, A. A. Validation of a RP-HPLC-DAD Method for Chamomile (Matricaria Recutita) Preparations and Assessment of the Marker, Apigenin-7-Glucoside, Safety and Anti-Inflammatory Effect. Evidence-Based Complement. Altern. Med. 2015, 2015, 1–9. DOI: 10.1155/2015/828437.
  • Pamunuwa, G.; Karunaratne, D. N.; Waisundara, V. Y. Antidiabetic Properties, Bioactive Constituents, and Other Therapeutic Effects of Scoparia Dulcis. Evidence-Based Complement. Altern. Med. 2016, 2016, 1–11. DOI: 10.1155/2016/8243215.
  • Shay, J.; Elbaz, H. A.; Lee, I.; Zielske, S. P.; Malek, M. H.; Hüttemann, M. Molecular Mechanisms and Therapeutic Effects of (−)-Epicatechin and Other Polyphenols in Cancer, Inflammation, Diabetes, and Neurodegeneration. OXID. MED. CELL LONGEV. 2015, 2015, 1–13. DOI: 10.1155/2015/181260.
  • Bijani, S.; Dizaji, R.; Sharafi, A.; Hosseini, M.-J. Neuroprotective Effect of Apigenin on Depressive-Like Behavior: Mechanistic Approach. Neurochem. Res. 2022, 47(3), 644–655. DOI: 10.1007/s11064-021-03473-0.
  • Wang, Q.-Q.; Cheng, N.; Yi, W.-B.; Peng, S.-M.; Zou, X.-Q. Synthesis, Nitric Oxide Release, and α-Glucosidase Inhibition of Nitric Oxide Donating Apigenin and Chrysin Derivatives. Bioorg. Med. Chem. 2014, 22(5), 1515–1521. DOI: 10.1016/j.bmc.2014.01.038.
  • Iizumi, Y.; Oishi, M.; Taniguchi, T.; Goi, W.; Sowa, Y.; Sakai, T.; Fei, P. The Flavonoid Apigenin Downregulates CDK1 by Directly Targeting Ribosomal Protein S9. PLoS One. 2013, 8(8), e73219. DOI: 10.1371/journal.pone.0073219.
  • Maggioni, D.; Garavello, W.; Rigolio, R.; Pignataro, L.; Gaini, R.; Nicolini, G. Apigenin Impairs Oral Squamous Cell Carcinoma Growth in vitro Inducing Cell Cycle Arrest and Apoptosis. Int. J. Oncol. 2013, 43(5), 1675–1682. DOI: 10.3892/ijo.2013.2072.
  • Takagaki, N.; Sowa, Y.; Oki, T.; Nakanishi, R.; Yogosawa, S.; Sakai, T. Apigenin induces cell cycle arrest and p21/WAF1 expression in a p53-independent pathway. Int. J. Oncol. 2005, 25, 5257. DOI: 10.3892/ijo.26.1.185.
  • SEO, H.-S.; KU, J. M.; CHOI, H.-S.; WOO, J.-K.; JANG, B.-H.; GO, H.; SHIN, Y. C.; KO, S.-G. Apigenin Induces Caspase-Dependent Apoptosis by Inhibiting Signal Transducer and Activator of Transcription 3 Signaling in HER2-Overexpressing SKBR3 Breast Cancer Cells. Mol. Med. Rep. 2015, 12(2), 2977–2984. DOI: 10.3892/mmr.2015.3698.
  • Seo, H.-S.; Choi, H.-S.; Kim, S.-R.; Choi, Y. K.; Woo, S.-M.; Shin, I.; Woo, J.-K.; Park, S.-Y.; Shin, Y. C.; Ko, S.-K. Apigenin Induces Apoptosis via Extrinsic Pathway, Inducing p53 and Inhibiting STAT3 and Nfκb Signaling in HER2-Overexpressing Breast Cancer Cells. Mol. Cell. Biochem. 2012, 366(1–2), 319–334. DOI: 10.1007/s11010-012-1310-2.
  • Chiang, L.-C.; Ng, L. T.; Lin, I.-C.; Kuo, P.-L.; Lin, C.-C. Anti-Proliferative Effect of Apigenin and Its Apoptotic Induction in Human Hep G2 Cells. Cancer Lett. 2006, 237(2), 207–214. DOI: 10.1016/j.canlet.2005.06.002.
  • Avallone, R.; Zanoli, P.; Puia, G.; Kleinschnitz, M.; Schreier, P.; Baraldi, M. Pharmacological Profile of Apigenin, a Flavonoid Isolated from Matricaria Chamomilla. Biochem. Pharmacol. 2000, 59(11), 1387–1394. DOI: 10.1016/S0006-2952(00)00264-1.
  • Zanoli, P.; Avallone, R.; Baraldi, M. Behavioral Characterisation of the Flavonoids Apigenin and Chrysin. Fitoterapia. 2000, 71, S117–S123. DOI: 10.1016/S0367-326X(00)00186-6.
  • Izumi, S.; Takashima, O.; Hirata, T. Geraniol is a Potent Inducer of Apoptosis-Like Cell Death in the Cultured Shoot Primordia of Matricaria Chamomilla. Biochem. Biophys. Res. Commun. 1999, 259(3), 519–522. DOI: 10.1006/bbrc.1999.0813.
  • Lei, Y.; Fu, P.; Jun, X.; Cheng, P. Pharmacological Properties of Geraniol – a Review. Planta. med. 2019, 85(1), 48–55. DOI: 10.1055/a-0750-6907.
  • Chaudhary, S. C.; Siddiqui, M. S.; Athar, M.; Alam, M. S. Geraniol Inhibits Murine Skin Tumorigenesis by Modulating COX‐2 Expression, Ras‐ERK1/2 Signaling Pathway and Apoptosis. J. Appl. Toxicol. 2013, 33(8), 828–837. DOI: 10.1002/jat.2739.
  • Qi, F.; Yan, Q.; Zheng, Z.; Liu, J.; Chen, Y.; Zhang, G. Geraniol and Geranyl Acetate Induce Potent Anticancer Effects in Colon Cancer Colo-205 Cells by Inducing Apoptosis, DNA Damage and Cell Cycle Arrest. J. Buon. 2018, 23(2), 346–352. DOI: 10.1177/1934578X1701200519.
  • Khan, A. Q.; Khan, R.; Qamar, W.; Lateef, A.; Rehman, M. U.; Tahir, M.; Ali, F.; Hamiza, O. O.; Hasan, S. K.; Sultana, S. Geraniol Attenuates 12-O-Tetradecanoylphorbol-13-Acetate (TPA)-Induced Oxidative Stress and Inflammation in Mouse Skin: Possible Role of p38 MAP Kinase and NF-Κb. Exp. Mol. Pathol. 2013, 94(3), 419–429. DOI: 10.1016/j.yexmp.2013.01.006.
  • Wittig, C.; Scheuer, C.; Parakenings, J.; Menger, M. D.; Laschke, M. W.; Addison, C. L. Geraniol Suppresses Angiogenesis by Downregulating Vascular Endothelial Growth Factor (VEGF)/VEGFR-2 Signaling. PLoS One. 2015, 10(7), e0131946. DOI: 10.1371/journal.pone.0131946.
  • Murbach Teles Andrade, B. F.; Conti, B. J.; Santiago, K. B.; Fernandes, A.; Sforcin, J. M. C ymbopogon martinii essential oil and geraniol at noncytotoxic concentrations exerted immunomodulatory/anti-inflammatory effects in human monocytes. J. Pharm. Pharmacol. 2014, 66(10), 1491–1496. DOI: 10.1111/jphp.12278.
  • Su, Y.-W.; Chao, S.-H.; Lee, M.-H.; Ou, T.-Y.; Tsai, Y.-C. Inhibitory Effects of Citronellol and Geraniol on Nitric Oxide and Prostaglandin E 2 Production in Macrophages. Planta. med. 2010, 76(15), 1666–1671. DOI: 10.1055/s-0030-1249947.
  • El Azab, F.; Elguindy, E.; Yacout, N. M.; Elgamal, G. A. Hepatoprotective Impact of Geraniol Against CCl4-Induced Liver Fibrosis in Rats. Pakistan J. Biol. Sci. 2020, 23(12), 1650–1658. DOI: 10.3923/pjbs.2020.1650.1658.
  • Chen, J.; Fan, X.; Zhou, L.; Gao, X. Treatment with Geraniol Ameliorates Methionine‐Choline‐Deficient Diet‐Induced Non‐Alcoholic Steatohepatitis in Rats. J. Gastroenterol. Hepatol. 2016, 31(7), 1357–1365. DOI: 10.1111/jgh.13272.
  • Ibrahim, S. M.; El- Denshary, E. S.; Abdallah, D. M.; Hribal, M. L. Geraniol, Alone and in Combination with Pioglitazone, Ameliorates Fructose-Induced Metabolic Syndrome in Rats via the Modulation of Both Inflammatory and Oxidative Stress Status. PLoS One. 2015, 10(2), e0117516. DOI: 10.1371/journal.pone.0117516.
  • Galle, M.; Kladniew, B. R.; Castro, M. A.; Villegas, S. M.; Lacunza, E.; Polo, M.; de Bravo, M. G.; Crespo, R. Modulation by Geraniol of Gene Expression Involved in Lipid Metabolism Leading to a Reduction of Serum-Cholesterol and Triglyceride Levels. Phytomedicine. 2015, 22(7–8), 696–704. DOI: 10.1016/j.phymed.2015.04.005.
  • Singh, S.; Fatima, Z.; Ahmad, K.; Hameed, S.; Sturtevant, J. Fungicidal action of geraniol against Candida albicans is potentiated by abrogated CaCdr1p drug efflux and fluconazole synergism. PLoS One. 2018, 13(8), e0203079. DOI: 10.1371/journal.pone.0203079.
  • Singh, S.; Fatima, Z.; Hameed, S. Insights into the Mode of Action of Anticandidal Herbal Monoterpenoid Geraniol Reveal Disruption of Multiple MDR Mechanisms and Virulence Attributes in Candida albicans. Arch. Microbiol. 2016, 198(5), 459–472. DOI: 10.1007/s00203-016-1205-9.
  • Sharma, Y.; Khan, L. A.; Manzoor, N. Anti-Candida Activity of Geraniol Involves Disruption of Cell Membrane Integrity and Function. J. Med. Mycol. 2016, 26(3), 244–254. DOI: 10.1016/j.mycmed.2016.04.004.
  • Leite, M. C. A.; de Brito Bezerra, A. P.; de Sousa, J. P.; de Oliveira Lima, E. Investigating the Antifungal Activity and Mechanism(s) of Geraniol Against Candida albicans Strains. Med. Mycol. 2015, 53(3), 275–284. DOI: 10.1093/mmy/myu078.
  • Asgharzade, S.; Khorrami, M. B.; Forouzanfar, F. Neuroprotective Effect of Herniarin Following Transient Focal Cerebral Ischemia in Rats. Metab. Brain Dis. 2021, 36(8), 2505–2510. DOI: 10.1007/s11011-021-00841-1.
  • Shaibani, Z. The Effect of Herniarin on Oxidative Stress in the Hippocampus in Streptozotocin-Induced Diabetic Rats. J. Anim. Biol. 2021, 14, 55–63. DOI: 10.22034/ASCIJ.2021.684776.
  • Salehcheh, M.; Safari, O.; Khodayar, M. J.; Mojiri-Forushani, H.; Cheki, M. The Protective Effect of Herniarin on Genotoxicity and Apoptosis Induced by Cisplatin in Bone Marrow Cells of Rats. Drug Chem. Toxicol. 2022, 45(4), 1470–1475. DOI: 10.1080/01480545.2020.1842883.
  • Sim, M.-O.; Lee, H.-I.; Ham, J. R.; Seo, K.-I.; Kim, M.-J.; Lee, M.-K. Anti-Inflammatory and Antioxidant Effects of Umbelliferone in Chronic Alcohol-Fed Rats. Nutr. Res. Pract. 2015, 9(4), 364. DOI: 10.4162/nrp.2015.9.4.364.
  • Boulebd, H. Are Thymol, Rosefuran, Terpinolene and Umbelliferone Good Scavengers of Peroxyl Radicals? Phytochemistry. 2021, 184, 112670. DOI: 10.1016/j.phytochem.2021.112670.
  • Alotaibi, M. F.; Al-Joufi, F.; Abou Seif, H. S.; Alzoghaibi, M. A.; Djouhri, L.; Ahmeda, A. F.; Mahmoud, A. M. Umbelliferone Inhibits Spermatogenic Defects and Testicular Injury in Lead-Intoxicated Rats by Suppressing Oxidative Stress and Inflammation, and Improving Nrf2/HO-1 Signaling. Drug Des. Devel. Ther. 2020, 14, 4003–4019. DOI: 10.2147/DDDT.S265636.
  • Wang, D.; Wang, X.; Tong, W.; Cui, Y.; Li, X.; Sun, H. Umbelliferone Alleviates Lipopolysaccharide-Induced Inflammatory Responses in Acute Lung Injury by Down-Regulating TLR4/MyD88/NF-Κb Signaling. Inflammation. 2019, 42(2), 440–448. DOI: 10.1007/s10753-018-00953-4.
  • Wang, H.-Q.; Wang, S.-S.; Chiufai, K.; Wang, Q.; Cheng, X.-L. Umbelliferone Ameliorates Renal Function in Diabetic Nephropathy Rats Through Regulating Inflammation and TLR/NF-Κb Pathway. Chin. J. Nat. Med. 2019, 17(5), 346–354. DOI: 10.1016/S1875-5364(19)30040-8.
  • Kundu, M.; Chatterjee, S.; Ghosh, N.; Manna, P.; Das, J.; Sil, P. C. Tumor Targeted Delivery of Umbelliferone via a Smart Mesoporous Silica Nanoparticles Controlled-Release Drug Delivery System for Increased Anticancer Efficiency. Mater. Sci. Eng. C. 2020, 116, 111239. DOI: 10.1016/j.msec.2020.111239.
  • Hassanein, E. H. M.; Ali, F. E. M.; Kozman, M. R.; Abd El-Ghafar, O. A. M. Umbelliferone Attenuates Gentamicin-Induced Renal Toxicity by Suppression of TLR-4/nf-Κb-p65/NLRP-3 and JAK1/STAT-3 Signaling Pathways. Environ. Sci. Pollut. Res. 2021, 28(9), 11558–11571. DOI: 10.1007/s11356-020-11416-5.
  • Sotiropoulou, N. S.; Megremi, S. F.; Tarantilis, P. Evaluation of Antioxidant Activity, Toxicity, and Phenolic Profile of Aqueous Extracts of Chamomile (Matricaria Chamomilla L.) and Sage (Salvia Officinalis L.) Prepared at Different Temperatures. Appl. Sci. 2020, 10(7), 2270. DOI: 10.3390/app10072270.
  • Gao, W.; Wang, C.; Yu, L.; Sheng, T.; Wu, Z.; Wang, X.; Zhang, D.; Lin, Y.; Gong, Y. Chlorogenic Acid Attenuates Dextran Sodium Sulfate-Induced Ulcerative Colitis in Mice Through MAPK/ERK/JNK Pathway. Biomed Res. Int. 2019, 2019, 1–14. DOI: 10.1155/2019/6916189.
  • Kováčik, J.; Klejdus, B. Dynamics of Phenolic Acids and Lignin Accumulation in Metal-Treated Matricaria Chamomilla Roots. Plant Cell Rep. 2008, 27(3), 605–615. DOI: 10.1007/s00299-007-0490-9.
  • Ye, X.; Liu, Y.; Hu, J.; Gao, Y.; Ma, Y.; Wen, D. Chlorogenic Acid-Induced Gut Microbiota Improves Metabolic Endotoxemia. Front. Endocrinol. (Lausanne). 2021, 12, 237–244. DOI: 10.3389/fendo.2021.762691.
  • Bhandarkar, N. S.; Brown, L.; Panchal, S. K. Chlorogenic acid attenuates high-carbohydrate, high-fat diet–induced cardiovascular, liver, and metabolic changes in rats. Nutr. Res. 2019, 62, 78–88. DOI: 10.1016/j.nutres.2018.11.002.
  • Ma, Y.; Gao, M.; Liu, D. Chlorogenic Acid Improves High Fat Diet-Induced Hepatic Steatosis and Insulin Resistance in Mice. Pharm. Res. 2015, 32(4), 1200–1209. DOI: 10.1007/s11095-014-1526-9.
  • Huang, S.; Wang, L.-L.; Xue, N.-N.; Li, C.; Guo, H.-H.; Ren, T.-K.; Zhan, Y.; Li, W.-B.; Zhang, J.; Chen, X.-G., et al. Chlorogenic Acid Effectively Treats Cancers Through Induction of Cancer Cell Differentiation. Theranostics. 2019, 9(23), 6745–6763. DOI: 10.7150/thno.34674.
  • Santana-Gálvez, J.; Villela Castrejón, J.; Serna-Saldívar, S. O.; Jacobo-Velázquez, D. A. Anticancer potential of dihydrocaffeic acid: a chlorogenic acid metabolite. CyTA - J. Food. 2020, 18(1), 245–248. DOI: 10.1080/19476337.2020.1743762.
  • Rebai, O.; Belkhir, M.; Sanchez-Gomez, M. V.; Matute, C.; Fattouch, S.; Amri, M. Differential Molecular Targets for Neuroprotective Effect of Chlorogenic Acid and Its Related Compounds Against Glutamate Induced Excitotoxicity and Oxidative Stress in Rat Cortical Neurons. Neurochem. Res. 2017, 42(12), 3559–3572. DOI: 10.1007/s11064-017-2403-9.
  • Heitman, E.; Ingram, D. K. Cognitive and neuroprotective effects of chlorogenic acid. Nutr. Neurosci. 2017, 20(1), 32–39. DOI: 10.1179/1476830514Y.0000000146.
  • Shan, S.; Tian, L.; Fang, R. Chlorogenic Acid Exerts Beneficial Effects in 6-Hydroxydopamine-Induced Neurotoxicity by Inhibition of Endoplasmic Reticulum Stress. Med. Sci. Monit. 2019, 25, 453–459. DOI: 10.12659/MSM.911166.
  • Kumar, G.; Mukherjee, S.; Paliwal, P.; Singh, S. S.; Birla, H.; Singh, S. P.; Krishnamurthy, S.; Patnaik, R. Neuroprotective Effect of Chlorogenic Acid in Global Cerebral Ischemia-Reperfusion Rat Model. Naunyn Schmiedebergs Arch. Pharmacol. 2019, 392(10), 1293–1309. DOI: 10.1007/s00210-019-01670-x.
  • Liu, D.; Wang, H.; Zhang, Y.; Zhang, Z. Protective Effects of Chlorogenic Acid on Cerebral Ischemia/Reperfusion Injury Rats by Regulating Oxidative Stress-Related Nrf2 Pathway. Nutr. Neurosci. 2020, 20, 32–39. DOI: 10.1179/1476830514Y.0000000146
  • Alam, M.; Ahmed, S.; Elasbali, A. M.; Adnan, M.; Alam, S.; Hassan, M. I.; Pasupuleti, V. R. Therapeutic Implications of Caffeic Acid in Cancer and Neurological Diseases. Front. Oncol. 2022, 12, 989–1010. DOI: 10.3389/fonc.2022.860508.
  • Kępa, M.; Miklasińska-Majdanik, M.; Wojtyczka, R. D.; Idzik, D.; Korzeniowski, K.; Smoleń-Dzirba, J.; Wąsik, T. J. Antimicrobial Potential of Caffeic Acid against Staphylococcus aureus Clinical Strains. Biomed Res. Int. 2018, 2018, 1–9. DOI: 10.1155/2018/7413504.
  • Rezaei-Seresht, H.; Cheshomi, H.; Falanji, F.; Movahedi-Motlagh, F.; Hashemian, M.; Mireskandari, E. Cytotoxic activity of caffeic acid and gallic acid against MCF-7 human breast cancer cells: An in silico and in vitro study. Avicenna J. Phytomedicine. 2019, 9, 574–586. DOI: 10.22038/AJP.2019.13475.
  • Niu, Y.; Wang, K.; Zheng, S.; Wang, Y.; Ren, Q.; Li, H.; Ding, L.; Li, W.; Zhang, L. Antibacterial Effect of Caffeic Acid Phenethyl Ester on Cariogenic Bacteria and Streptococcus mutans Biofilms. Antimicrob. Agents Chemother. 2020, 64(9), 789. DOI: 10.1128/AAC.00251-20.
  • Malik, J. A.; Ahmed, S.; Mir, A.; Shinde, M.; Bender, O.; Alshammari, F.; Ansari, M.; Anwar, S. The SARS-CoV-2 Mutations versus Vaccine Effectiveness: New Opportunities to New Challenges. J. Infect Public Health. 2022, 15(2), 228–240. DOI: 10.1016/j.jiph.2021.12.014.
  • Adem, Ş.; Eyupoglu, V.; Sarfraz, I.; Rasul, A.; Zahoor, A. F.; Ali, M.; Abdalla, M.; Ibrahim, I. M.; Elfiky, A. A. Caffeic Acid Derivatives (CAFDs) as Inhibitors of SARS-CoV-2: CAFDs-Based Functional Foods as a Potential Alternative Approach to Combat COVID-19. Phytomedicine. 2021, 85, 153310. DOI: 10.1016/j.phymed.2020.153310.
  • Cook, M. T. Mechanism of Metastasis Suppression by Luteolin in Breast Cancer. Breast Cancer Targets Ther. 2018, 10, 89–100. DOI: 10.2147/BCTT.S144202.
  • Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M. A.; Khan, I. A.; Imran, A.; Orhan, I. E.; Rizwan, M.; Atif, M.; et al. Luteolin, a Flavonoid, as an Anticancer Agent: A Review. Biomed. Pharmacother. 2019, 112, 108612. DOI: 10.1016/j.biopha.2019.108612.
  • Ashokkumar, P.; Sudhandiran, G. Luteolin Inhibits Cell Proliferation During Azoxymethane-Induced Experimental Colon Carcinogenesis via Wnt/β-Catenin Pathway. Invest. New Drugs. 2011, 29(2), 273–284. DOI: 10.1007/s10637-009-9359-9.
  • Xu, H.; Yu, W.; Sun, S.; Li, C.; Zhang, Y.; Ren, J. Luteolin Attenuates Doxorubicin-Induced Cardiotoxicity Through Promoting Mitochondrial Autophagy. Front. Physiol. 2020, 11, 10990–10998. DOI: 10.3389/fphys.2020.00113.
  • Guimarães, R.; Calhelha, R. C.; Froufe, H. J. C.; Abreu, R. M. V.; Carvalho, A. M.; Queiroz, M. J.; Ferreira, R. P. Wild Roman Chamomile Extracts and Phenolic Compounds: Enzymatic Assays and Molecular Modelling Studies with VEGFR-2 Tyrosine Kinase. Food Funct. 2016, 7(1), 79–83. DOI: 10.1039/C5FO00586H.
  • Gendrisch, F.; Esser, P. R.; Schempp, C. M.; Wölfle, U. Luteolin as a Modulator of Skin Aging and Inflammation. BioFactors. 2021, 47(2), 170–180. DOI: 10.1002/biof.1699.
  • Kempuraj, D.; Thangavel, R.; Kempuraj, D. D.; Ahmed, M. E.; Selvakumar, G. P.; Raikwar, S. P.; Zaheer, S. A.; Iyer, S. S.; Govindarajan, R.; Chandrasekaran, P. N., et al. Neuroprotective effects of flavone luteolin in neuroinflammation and neurotrauma. BioFactors. 2021, 47(2), 190–197. DOI: 10.1002/biof.1687.
  • Salehi, B.; Machin, L.; Monzote, L.; Sharifi-Rad, J.; Ezzat, S. M.; Salem, M. A.; Merghany, R. M.; El Mahdy, N. M.; Kılıç, C. S.; Sytar, O., et al. Therapeutic Potential of Quercetin: New Insights and Perspectives for Human Health. ACS Omega. 2020, 5(20), 11849–11872. DOI: https://doi.org/10.1021/acsomega.0c01818.
  • Patel, R. V.; Mistry, B. M.; Shinde, S. K.; Syed, R.; Singh, V.; Shin, H.-S. Therapeutic potential of quercetin as a cardiovascular agent. Eur. J. Med. Chem. 2018, 155, 889–904. DOI: 10.1016/j.ejmech.2018.06.053.
  • Lesjak, M.; Beara, I.; Simin, N.; Pintać, D.; Majkić, T.; Bekvalac, K.; Orčić, D.; Mimica-Dukić, N. Antioxidant and Anti-Inflammatory Activities of Quercetin and Its Derivatives. J. Funct. Foods. 2018, 40, 68–75. DOI: 10.1016/j.jff.2017.10.047.
  • Azeem, M.; Hanif, M.; Mahmood, K.; Ameer, N.; Chughtai, F. R. S.; Abid, U. An Insight into Anticancer, Antioxidant, Antimicrobial, Antidiabetic and Anti-Inflammatory Effects of Quercetin: A Review. Polym. Bull. 2023, 80(1), 241–262. DOI: 10.1007/s00289-022-04091-8.
  • Wiczkowski, W.; Szawara-Nowak, D.; Topolska, J.; Olejarz, K.; Zieliński, H.; Piskuła, M. K. Metabolites of Dietary Quercetin: Profile, Isolation, Identification, and Antioxidant Capacity. J. Funct. Foods. 2014, 11, 121–129. DOI: 10.1016/j.jff.2014.09.013.
  • Rauf, A.; Imran, M.; Khan, I. A.; Ur‐Rehman, M.; Gilani, S. A.; Mehmood, Z.; Mubarak, M. S. Anticancer potential of quercetin: A comprehensive review. Phyther. Res. 2018, 32(11), 2109–2130. DOI: 10.1002/ptr.6155.
  • Zaplatic, E.; Bule, M.; Shah, S. Z. A.; Sahab Uddin, M.; Niaz, K. Corrigendum to “Molecular Mechanisms Underlying Protective Role of Quercetin in Attenuating Alzheimer’s disease” [Life Sci. 221 (2019) 109–119]. Life. sci. 2019, 231(231), 116616. DOI: 10.1016/j.lfs.2019.116616.
  • Nouri, Z.; Fakhri, S.; Nouri, K.; Wallace, C. E.; Farzaei, M. H.; Bishayee, A. Targeting Multiple Signaling Pathways in Cancer: The Rutin Therapeutic Approach. Cancers (Basel). 2020, 12(8), 2276. DOI: 10.3390/cancers12082276.
  • Enogieru, A. B.; Haylett, W.; Hiss, D. C.; Bardien, S.; Ekpo, O. E. Rutin as a Potent Antioxidant: Implications for Neurodegenerative Disorders. OXID. MED. CELL LONGEV. 2018, 2018, 1–17. DOI: 10.1155/2018/6241017.
  • Irfan, F.; Jameel, F.; Khan, I.; Aslam, R.; Faizi, S.; Salim, A. Role of Quercetin and Rutin in Enhancing the Therapeutic Potential of Mesenchymal Stem Cells for Cold Induced Burn Wound. Regen. Ther. 2022, 21, 225–238. DOI: 10.1016/j.reth.2022.07.011.
  • Imani, A.; Maleki, N.; Bohlouli, S.; Kouhsoltani, M.; Sharifi, S.; Maleki Dizaj, S. Molecular mechanisms of anticancer effect of rutin. Phyther. Res. 2021, 35(5), 2500–2513. DOI: 10.1002/ptr.6977.
  • Sun, X.; Li, L.; Dong, Q.-X.; Zhu, J.; Huang, Y.; Hou, S.; Yu, X.; Liu, R. Rutin Prevents Tau Pathology and Neuroinflammation in a Mouse Model of Alzheimer’s Disease. J. Neuroinflammation. 2021, 18(1), 131. DOI: 10.1186/s12974-021-02182-3.
  • Satyal, P.; Shrestha, S.; Setzer, W. N. Composition and Bioactivities of an (E)-β-Farnesene Chemotype of Chamomile (Matricaria Chamomilla) Essential Oil from Nepal. Nat. Prod. Commun. 2015, 10(8), 1934578X1501000. DOI: 10.1177/1934578X1501000835.
  • Roby, M. H. H.; Sarhan, M. A.; Selim, K. A.-H.; Khalel, K. I. Antioxidant and antimicrobial activities of essential oil and extracts of fennel (Foeniculum vulgare L.) and chamomile (Matricaria chamomilla L.). Ind. Crops Prod. 2013, 44, 437–445. DOI: 10.1016/j.indcrop.2012.10.012.
  • Seyedjavadi, S. S.; Khani, S.; Zare‐Zardini, H.; Halabian, R.; Goudarzi, M.; Khatami, S.; Imani Fooladi, A. A.; Amani, J.; Razzaghi‐Abyaneh, M. Isolation, Functional Characterization, and Biological Properties of MCh‐AMP1, a Novel Antifungal Peptide from Matricaria Chamomilla L. Chem. Biol. Drug Des. 2019, 93(5), 949–959. DOI: 10.1111/cbdd.13500.
  • Tolouee, M.; Alinezhad, S.; Saberi, R.; Eslamifar, A.; Zad, S. J.; Jaimand, K.; Taeb, J.; Rezaee, M.-B.; Kawachi, M.; Shams-Ghahfarokhi, M.; et al. Effect of Matricaria Chamomilla L. Flower Essential Oil on the Growth and Ultrastructure of Aspergillus Niger van Tieghem. Int. J. Food Microbiol. 2010, 139(3), 127–133.
  • Enas, M. A. Phytochemical Composition, Antifungal, Antiaflatoxigenic, Antioxidant, and Anticancer Activities of Glycyrrhiza Glabra L. and Matricaria Chamomilla L. Essential Oils. J. Med. Plants Res. 2013, 7(29), 2197–2207. DOI: 10.5897/JMPR12.5134.
  • EL-Hefny, M.; Abo Elgat, W.; Al-Huqail, A.; Ali, H. Essential and Recovery Oils from Matricaria Chamomilla Flowers as Environmentally Friendly Fungicides Against Four Fungi Isolated from Cultural Heritage Objects. Processes. 2019, 7(11), 809. DOI: 10.3390/pr7110809.
  • Dadashpour, M.; Firouzi-Amandi, A.; Pourhassan-Moghaddam, M.; Maleki, M. J.; Soozangar, N.; Jeddi, F.; Nouri, M.; Zarghami, N.; Pilehvar-Soltanahmadi, Y. Biomimetic Synthesis of Silver Nanoparticles Using Matricaria Chamomilla Extract and Their Potential Anticancer Activity Against Human Lung Cancer Cells. Mater. Sci. Eng. C. 2018, 92, 902–912. DOI: 10.1016/j.msec.2018.07.053.
  • Kamali, A. M.; Nikseresht, M.; Delaviz, H.; Barmak, M. J. S.; Ardakani, M.; Mahmoudi, M. T. In vitro Cytotoxic Activity of Matricaria Chamomilla Root Extract in Human Breast Cancer Cell Line MCF-7. Life Sci. J. 2014, 11, 403–406.
  • Nikseresht, M.; Kamali, A.; Rahimi, H.; Delaviz, H.; Toori, M.; Kashani, I.; Mahmoudi, R. The Hydroalcoholic Extract of Matricaria Chamomilla Suppresses Migration and Invasion of Human Breast Cancer MDA-MB-468 and MCF-7 Cell Lines. Pharmacognosy. Res. 2017, 9(1), 87. DOI: 10.4103/0974-8490.199778.
  • Shaaban, M.; El-Hagrassi, A. M.; Osman, A. F.; Soltan, M. M. Bioactive Compounds from Matricaria Chamomilla: Structure Identification, in vitro Antiproliferative, Antimigratory, Antiangiogenic, and Antiadenoviral Activities. Zeitschrift für Naturforsch. C. 2022, 77(3–4), 85–94. DOI: 10.1515/znc-2021-0083.
  • Mitoshi, M.; Kuriyama, I.; Nakayama, H.; Miyazato, H.; Sugimoto, K.; Kobayashi, Y.; Jippo, T.; Kanazawa, K.; Yoshida, H.; Mizushina, Y. Effects of Essential Oils from Herbal Plants and Citrus Fruits on DNA Polymerase Inhibitory, Cancer Cell Growth Inhibitory, Antiallergic, and Antioxidant Activities. J. Agric. Food. Chem. 2012, 60(45), 11343–11350. DOI: 10.1021/jf303377f.
  • El Joumaa, M. M.; Taleb, R. I.; Rizk, S.; Borjac, J. M. Protective effect of Matricaria chamomilla extract against 1,2-dimethylhydrazine-induced colorectal cancer in mice. J. Complement Integr. Med. 2020, 17(3), 20190143. DOI: 10.1515/jcim-2019-0143.
  • Shebbo, S.; El Joumaa, M.; Kawach, R.; Borjac, J. Hepatoprotective Effect of Matricaria Chamomilla Aqueous Extract Against 1,2-Dimethylhydrazine-Induced Carcinogenic Hepatic Damage in Mice. Heliyon. 2020, 6(6), e04082. DOI: 10.1016/j.heliyon.2020.e04082.
  • Hassanpour, H.; Ghanbarzadeh, M. Induction of cell division and antioxidative enzyme activity of Matricaria chamomilla L. cell line under clino-rotation. Plant Cell Tissue Organ Cult. 2021, 146(2), 215–224. DOI: 10.1007/s11240-021-02060-z.
  • Ranjbar, A.; Mohsenzadeh, F.; Chehregani, A.; Khajavi, F.; Zijoud, S.-M.; Ghasemi, H. Ameliorative Effect of Matricaria Chamomilla.L on Paraquat: Induced Oxidative Damage in Lung Rats. Pharmacognosy. Res. 2014, 6(3), 199. DOI: 10.4103/0974-8490.132595.
  • Kolodziejczyk-Czepas, J.; Bijak, M.; Saluk, J.; Ponczek, M. B.; Zbikowska, H. M.; Nowak, P.; Tsirigotis-Maniecka, M.; Pawlaczyk, I. Radical Scavenging and Antioxidant Effects of Matricaria Chamomilla Polyphenolic–Polysaccharide Conjugates. Int. J. Biol. Macromol. 2015, 72, 1152–1158. DOI: 10.1016/j.ijbiomac.2014.09.032.
  • Aksoy, L.; Sözbilir, N. B. Effects of Matricaria Chamomilla L. on Lipid Peroxidation, Antioxidant Enzyme Systems, and Key Liver Enzymes in CCl 4 -Treated Rats. Toxicol. Environ. Chem. 2012, 94(9), 1780–1788. DOI: 10.1080/02772248.2012.729837.
  • Iverson, C. D.; Zahid, S.; Li, Y.; Shoqafi, A. H.; Ata, A.; Samarasekera, R. Glutathione S-Transferase Inhibitory, Free Radical Scavenging, and Anti-Leishmanial Activities of Chemical Constituents of Artocarpus Nobilis and Matricaria Chamomilla. Phytochem. Lett. 2010, 3(4), 207–211. DOI: 10.1016/j.phytol.2010.07.008.
  • Ortiz, M. I.; Cariño‐Cortés, R.; Ponce‐Monter, H. A.; González‐García, M. P.; Castañeda‐Hernández, G.; Salinas‐Caballero, M. Synergistic Interaction of Matricaria Chamomilla Extract with Diclofenac and Indomethacin on Carrageenan‐Induced Paw Inflammation in Rats. Drug Dev. Res. 2017, 78(7), 360–367. DOI: 10.1002/ddr.21401.
  • El Mihyaoui, A.; Esteves da Silva, J. C. G.; Charfi, S.; Candela Castillo, M. E.; Lamarti, A.; Arnao, M. B. Chamomile (Matricaria Chamomilla L.): A Review of Ethnomedicinal Use. Phytochemistry And Pharmacological Uses. Life. 2022, 12(4), 479. DOI: 10.3390/life12040479.
  • Wu, Y.; Xu, Y.; Yao, L. Anti-inflammatory and Anti-allergic Effects of German Chamomile (Matricaria chamomilla L.). J. Essent. Oil Bear. Plants. 2012, 15(1), 75–83. DOI: 10.1080/0972060X.2012.10644022.
  • Sayyar, Z.; Yazdinezhad, A.; Hassan, M.; Jafari Anarkooli, I. Protective Effect of Matricaria Chamomilla Ethanolic Extract on Hippocampal Neuron Damage in Rats Exposed to Formaldehyde. OXID. MED. CELL LONGEV. 2018, 2018, 1–10. DOI: 10.1155/2018/6414317.
  • Asgharzade, S.; Rabiei, Z.; Rafieian-Kopaei, M. Effects of Matricaria Chamomilla Extract on Motor Coordination Impairment Induced by Scopolamine in Rats. Asian Pac. J. Trop. Biomed. 2015, 5(10), 829–833. DOI: 10.1016/j.apjtb.2015.06.006.
  • Alibabaei, Z.; Rabiei, Z.; Rahnama, S.; Mokhtari, S.; Rafieian-Kopaei, M. Matricaria Chamomilla Extract Demonstrates Antioxidant Properties Against Elevated Rat Brain Oxidative Status Induced by Amnestic Dose of Scopolamine. Biomed. Aging Pathol. 2014, 4(4), 355–360. DOI: 10.1016/j.biomag.2014.07.003.
  • Amsterdam, J. D.; Li, Q. S.; Xie, S. X.; Mao, J. J. Putative Antidepressant Effect of Chamomile (Matricaria Chamomilla L.) Oral Extract in Subjects with Comorbid Generalized Anxiety Disorder and Depression. J. Altern. Complement. Med. 2020, 26(9), 815–821. DOI: 10.1089/acm.2019.0252.
  • Abbasinia, H.; Alizadeh, Z.; Vakilian, K.; Jafari, Z.; Poor, P. M.; Ranjbaran, M. Effect of Chamomile Extract on Sleep Disorder in Menopausal Women. Iran. J. Obstet. Gynecol. Infertil. 2016, 19, 1–7. DOI: 10.22038/IJOGI.2016.7631.
  • Abad, A. N. A.; Nouri, M. H. K.; Gharjanie, A.; Tavakoli, F. Effect of Matricaria chamomilla Hydroalcoholic Extract on Cisplatin-induced Neuropathy in Mice. Chin. J. Nat. Med. 2011, 9, 126–131. DOI: 10.3724/SP.J.1009.2011.00126.
  • Nouri, K. Antinociceptive effect of Matricaria chamomilla on vincristine-induced peripheral neuropathy in mice. Afr J. Pharm. Pharmacol. 2012, 6(1), 24–29. DOI: 10.5897/AJPP11.340.
  • Ioniță, R. Anxiolytic and Antidepressant Effects of Matricaria Chamomilla Hydroalcoholic Extract in a Rat Model of Scopolamine. Farmacia. 2019, 67(1), 68–72. DOI: 10.31925/farmacia.2019.1.9.
  • Reis, L. S. L. D. S.; Pardo, P. E.; Oba, E.; Kronka, S. D. N.; Frazatti-Gallina, N. M. Matricaria chamomilla CH 12 decreases handling stress in Nelore calves. J. Vet. Sci. 2006, 7(2), 189. DOI: 10.4142/jvs.2006.7.2.189.
  • Moore, E.; Lockwood, D. Treatment of Visceral Leishmaniasis. J. Glob. Infect. Dis. 2010, 2(2), 151. DOI: 10.4103/0974-777X.62883.
  • Karam, T. K.; Ortega, S.; Ueda Nakamura, T.; Auzély-Velty, R.; Nakamura, C. V. Development of Chitosan Nanocapsules Containing Essential Oil of Matricaria Chamomilla L. for the Treatment of Cutaneous Leishmaniasis. Int. J. Biol. Macromol. 2020, 162, 199–208. DOI: 10.1016/j.ijbiomac.2020.06.149.
  • Luize, P. S.; Tiuman, T. S.; Morello, L. G.; Maza, P. K.; Ueda-Nakamura, T.; Dias Filho, B. P.; Cortez, D. A. G.; Mello, J. C. P. D.; Nakamura, C. V. Effects of Medicinal Plant Extracts on Growth of Leishmania (L.) Amazonensis and Trypanosoma Cruzi. Rev. Bras. Cienc. Farm. 2005, 41(1), 85–94. DOI: 10.1590/S1516-93322005000100010.
  • Macchioni, F.; Perrucci, S.; Cecchi, F.; Cioni, P. L.; Morelli, I.; Pampiglione, S. Acaricidal activity of aqueous extracts of camomile flowers, Matricaria chamomilla, against the mite Psoroptes cuniculi. Med. Vet. Entomol. 2004, 18(2), 205–207. DOI: 10.1111/j.0269-283X.2004.00488.x.
  • Bahmani, M.; Saki, K.; Gholami-Ahangaran, M.; Parsaei, P.; Mohsenzadegan, A.; Zia-Jahromi, N. Evaluating the Anti-Leech Activity of Methanolic Extract of Matricaria Chamomilla L. Comparing with Ivermectin, Mebendasole, Praziquantel, Rafoxanide, Febantel and Albendasole. Middle-East J. Sci. Res. 2012, 12, 260–263. DOI: 10.5829/idosi.mejsr.2012.12.2.6446.
  • Mollabashi, E. N.; Ziaie, T.; Khalesi, Z. B.; Leili, E. K.; Bekhradi, R. Effect of Chamomile capsule on premenstrual syndrome symptoms relief. Iran. J. Obstet. Gynecol. Infertil. 2018, 21, 72–80. DOI: 10.22038/IJOGI.2018.11799.
  • Johari, H.; Khavarian, M.; Mokhtari, M.; Kamali, M.; Kargar Jahromi, H. The Effects of Hydro Alcoholic Extract of Matricaria Chamomilla Flower on Testosterone and Gonadotropins Hormone in Adult Male Rat. Pars Jahrom Univ. Med. Sci. 2014, 12(4), 37–41. DOI: 10.29252/jmj.12.4.6.
  • Karbalay-Doust, S.; Noorafshan, A.; Dehghani, F.; Panjehshahin, M. R.; Monabati, A. Effects of Hydroalcoholic Extract of Matricaria Chamomilla on Serum Testosterone and Estradiol Levels, Spermatozoon Quality, and Tail Length in Rat. Iran. J. Med. Sci. 2010, 35, 122–128.
  • Shadi, T. Z.; Talal, A. Z. A Review of Four Common Medicinal Plants Used to Treat Eczema. J. Med. Plants Res. 2015, 9(24), 702–711. DOI: 10.5897/JMPR2015.5831.
  • Dolati, S. A Review of the Therapeutic Effects of Chamomile (Matricaria Chamomile) in Traditional and Modern Medicine. Res. Sport Sci. Med. Plants. 2021, 1, 1–12. DOI: 10.30495/VARZESH.2021.1918705.1003.
  • Boroujeni, H.; Parvin, N.; Mirzaeian, P.; Marandi, S. Formulation and Clinical Trial Study of AJMT Cream in Treatment of Eczema. Iioab. J. 2017, 8, 46–50.
  • Niknam, S.; Tofighi, Z.; Faramarzi, M. A.; Abdollahifar, M. A.; Sajadi, E.; Dinarvand, R.; Toliyat, T. Polyherbal Combination for Wound Healing: Matricaria Chamomilla L. and Punica Granatum L. DARU J. Pharm. Sci. 2021, 29(1), 133–145. DOI: 10.1007/s40199-021-00392-x.
  • Soltani, M.; Moghimian, M.; Abtahi-Eivari, S. H.; Shoorei, H.; Khaki, A.; Shokoohi, M. Protective Effects of Matricaria Chamomilla Extract on Torsion/detorsion-Induced Tissue Damage and Oxidative Stress in Adult Rat Testis. Int. J. Fertil. Steril. 2018, 12(3), 242–248. DOI: 10.22074/ijfs.2018.5324.
  • Jarrahi, M. An Experimental Study of the Effects of Matricaria Chamomilla Extract on Cutaneous Burn Wound Healing in Albino Rats. Nat. Prod. Res. 2008, 22(5), 422–427. DOI: 10.1080/14786410701591713.
  • Cemek, M.; Kağa, S.; Şimşek, N.; Büyükokuroğlu, M. E.; Konuk, M. Antihyperglycemic and Antioxidative Potential of Matricaria Chamomilla L. in Streptozotocin-Induced Diabetic Rats. J. Nat. Med. 2008, 62(3), 284–293. DOI: 10.1007/s11418-008-0228-1.
  • Mahadeva Rao, U. S.; Subramanian, S. Biochemical Evaluation of Antihyperglycemic and Antioxidative Effects of Morinda Citrifolia Fruit Extract Studied in Streptozotocin-Induced Diabetic Rats. Med. Chem. Res. 2009, 18(6), 433–446. DOI: 10.1007/s00044-008-9140-1.
  • Najla, O. A.; Olfat, A. K.; Kholoud, S. R.; Enas, N. D.; Hanan, I. Hypoglycemic and Biochemical Effects of Matricaria Chamomilla Leave Extract in Streptozotocin-Induced Diabetic Rats. J. Heal. Sci. 2012, 2, 43–48. DOI: 10.5923/j.health.20120205.01.
  • Soliman, G. A. The Potential Cardioprotective Effect of Matricaria Chamomilla Extract Against Diabetes-Induced Oxidative Stress in Rats. Farmacia. 2020, 68(2), 269–279. DOI: 10.31925/farmacia.2020.2.12.
  • Heidarianpour, A.; Mohammadi, F.; Keshvari, M.; Mirazi, N. Ameliorative Effects of Endurance Training and Matricaria Chamomilla Flowers Hydroethanolic Extract on Cognitive Deficit in Type 2 Diabetes Rats. Biomed. Pharmacother. 2021, 135, 111230. DOI: 10.1016/j.biopha.2021.111230.
  • Hajizadeh-Sharafabad, F.; Varshosaz, P.; Jafari-Vayghan, H.; Alizadeh, M.; Maleki, V. Chamomile (Matricaria recutita L.) and diabetes mellitus, current knowledge and the way forward: A systematic review. Complement Ther. Med. 2020, 48, 102284. DOI: 10.1016/j.ctim.2019.102284.
  • Tavakol, H.; Farzad, K.; Fariba, M.; Abdolkarim, C.; Hassan, G.; Seyed-Mostafa, H.; Akram, R. Hepatoprotective Effect of Matricaria Chamomilla.L in Paraquat Induced Rat Liver Injury. Drug Res. (Stuttg). 2014, 65(2), 61–64. DOI: 10.1055/s-0033-1363999.
  • Madrigal-Santillán, E. Review of Natural Products with Hepatoprotective Effects. World J. Gastroenterol. 2014, 20(40), 14787. DOI: 10.3748/wjg.v20.i40.14787.
  • Chani, J. M.; Jasim, N. Z. Hepatoprotective Effect of Matricaria Chamomilla Hot Aqueous Extract Against Methomyl 90%- Induced Hepatotoxicity in Mice. Al-Kufa Univ. J. Biol. 2016, 8, 185–195.
  • Salama, R. M. Matricaria chamomilla attenuates cisplatin nephrotoxicity. Saudi J. Kidney Dis. Transplant. 2012, 23(4), 765. DOI: 10.4103/1319-2442.98158.
  • Salama, R. H. M.; Abd-El-Hameed, N. A.; Abd-El-Ghaffar, S. K.; Mohammed, Z. T.; Ghandour, N. M. A. Nephroprotective Effect of Nigella Sativa and Matricaria Chamomilla in Cisplatin Induced Renal Injury—Supportive Treatments in Cisplatin Nephrotoxicity. Int. J. Clin. Med. 2011, 02(3), 185–195. DOI: 10.4236/ijcm.2011.23031.
  • Mehmood, M. H.; Munir, S.; Khalid, U. A.; Asrar, M.; Gilani, A. H. Antidiarrhoeal, Antisecretory and Antispasmodic Activities of Matricaria Chamomilla are Mediated Predominantly Through K±channels Activation. BMC Complement. Altern. Med. 2015, 15(1), 75. DOI: 10.1186/s12906-015-0595-6.
  • Romero, M. D. C.; Valero, A.; Martín-Sánchez, J.; Navarro-Moll, M. C. Activity of Matricaria Chamomilla Essential Oil Against Anisakiasis. Phytomedicine. 2012, 19(6), 520–523. DOI: 10.1016/j.phymed.2012.02.005.
  • Bahrami, A. M.; Doosti, A.; Moosavi, A. B. Effect of Matricaria Chamomilla L. Plant Extraction on Experimental Infected Lamb with Ostertagia Ostertagi Parasits. Int. J. Pharmacol. 2010, 6(5), 712–718. DOI: 10.3923/ijp.2010.712.718.
  • Hajaji, S.; Sifaoui, I.; López-Arencibia, A.; Reyes-Batlle, M.; Jiménez, I. A.; Bazzocchi, I. L.; Valladares, B.; Pinero, J. E.; Lorenzo-Morales, J.; Akkari, H. Correlation of Radical-Scavenging Capacity and Amoebicidal Activity of Matricaria Recutita L. (Asteraceae). Exp. Parasitol. 2017, 183, 212–217. DOI: 10.1016/j.exppara.2017.09.011.
  • Váradyová, Z.; Pisarčíková, J.; Babják, M.; Hodges, A.; Mravčáková, D.; Kišidayová, S.; Königová, A.; Vadlejch, J.; Várady, M. Ovicidal and Larvicidal Activity of Extracts from Medicinal-Plants Against Haemonchus Contortus. Exp. Parasitol. 2018, 195, 71–77. DOI: 10.1016/j.exppara.2018.10.009.
  • Cemek, M.; Yilmaz, E.; Büyükokuroğlu, M. E. Protective Effect of Matricaria Chamomilla on Ethanol-Induced Acute Gastric Mucosal Injury in Rats. Pharm. Biol. 2010, 48(7), 757–763. DOI: 10.3109/13880200903296147.
  • Morshedi, M.; Gol, A.; Mohammadzadeh, A. The Effect of Matricaria Chamomilla on the Treatment of Ibuprofen-Induced Gastric Ulcers in Male Rats. Hormozgan Med. J. 2014, 20, 270–275.
  • Karbalay-Doust, S.; Noorafshan, A. Antiulcerogenic Effects of Matricaria Chamomilla Extract in Experimental Gastric Ulcer in Mice. Iran. J. Med. Sci. 2009, 34, 198–203.
  • Naema, F.; J, N.; Ali, A.; Ahmed, A. Effect of the Aqueous Extract of Matricaria Chamomilla on Stress-Ethanol Induced Acute Gastric Ulceration in Rabbits. Med. J. Basrah Univ. 2006, 24(1), 66–69. DOI: 10.33762/mjbu.2006.46423.
  • El Souda, S. S. E. D.; Ahmed, K. M.; Grace, M. H.; Elkherassy, E. E. A.; Farrag, A. R. H.; Abdelwahab, S. M. Flavonoids and Gastroprotective Effect of Matricaria Chamomilla Against Indomethacin-Induced Ulcer in Rats. J. Herbs. Spices Med. Plants. 2015, 21(2), 111–117. DOI: 10.1080/10496475.2014.919372.
  • Gomaa, A.; Hashem, T.; Mohamed, M.; Ashry, E. Matricaria Chamomilla Extract Inhibits Both Development of Morphine Dependence and Expression of Abstinence Syndrome in Rats. J. Pharmacol. Sci. 2003, 92(1), 50–55. DOI: 10.1254/jphs.92.50.
  • de Souza Reis, L. S. L.; Frazatti-Gallina, N. M.; de Lima Paoli, R.; Giuffrida, R.; Albas, A.; Oba, E.; Pardo, P. E. Efficiency of Matricaria chamomilla CH12 and number of doses of rabies vaccine on the humoral immune response in cattle. J. Vet. Sci. 2008, 9(4), 433. DOI: 10.4142/jvs.2008.9.4.433.
  • Dada, R.; Toghyani, M.; Tabeidian, S. A. The Effect of Chamomile Flower (Matricaria Chamomilla L.) Extract and Powder as Growth Promoter on Growth Performance and Digestive Organs of Broiler Chickens. Res. Opin. Anim. Vet. Sci. 2015, 5, 290–294.