Flavonoids from Clerodendrum genus and their biological activities

O.L. Erukainure, O. V. Oke, A.J. Ajiboye, O.Y. Okafor. Nutritional qualities and phytochemical constituents of Clerodendrum volubile, a tropical non-conventional vegetable. International Food Research Journal 18 (2011) 1393-1399. http://www.ifrj.upm.edu.my/18%20(04)%202011/(27)IFRJ-2011-009.pdf

N. Shrivastava, T. Patel. Clerodendrum and Heathcare: An Overview. Medicinal and Aromatic Plant Science and Biotechnology ©2007 Global Science Books 1 (2007) 142-150. http://www.globalsciencebooks.info/Online/GSBOnline/images/0706/MAPSB_1(1)/MAPSB_1(1)142-150o.pdf

P. Kar, A.K. Goyal, A.P. Das, A. Sen. Antioxidant and pharmaceutical potential of Clerodendrum L.: An overview. International Journal of Green Pharmacy 8 (2014). 210-216. https://www.greenpharmacy.info/index.php/ijgp/article/view/415

A. Yadav, V.P. Kuamr, T. Chand, R.H. Bora. Ethno-medicinal knowledge of Clerodendrum L. among different tribes of Nambor reserve forest, Assam, India. Journal of Pharmacognosy and Phytochemistry 7 (2018). 1567-1570. https://www.phytojournal.com/archives/2018/vol7issue5/PartAA/7-5-235-900.pdf

R.M. Rueda. The Genus Clerodendrum (Verbenaceae) in Mesoamerica. Annals of the Missouri Botanical Garden 80 (1993) 870-890. https://doi.org/10.2307/2399934

V. Hamilton. Mabberley’s Plant-Book: A Portable Dictionary of Plants, their Classification and Uses (4th edition). Reference Reviews 32 (2018) 28-29. https://doi.org/10.1108/RR-12-2017-0259

C. Leeratiwong, P. Chantaranothai, A.J. Paton. A Synopsis of the Genus Clerodendrum L. (Lamiaceae ) in Thailand. Tropical Natural History 11 (2011) 177-211. https://www.thaiscience.info/journals/Article/TNAH/10801631.pdf

L. Guang-Wei, M. Katsuyuki, Y. Tokihito, Y. Kenjiro. Effects of extract from Clerodendron trichotomum on blood pressure and renal function in rats and dogs. Journal of Ethnopharmacology 42 (1994) 77-82. https://doi.org/10.1016/0378-8741(94)90100-7

J.J. Patel, S.R. Acharya, N.S. Acharya. Clerodendrum serratum (L.) Moon. - A review on traditional uses, phytochemistry and pharmacological activities. Journal of Ethnopharmacology 154 (2014) 268-285. https://doi.org/10.1016/j.jep.2014.03.071

P. Solapure, Dr. Pradeep, R. Mundugaru, P.L. Hegde. Comparative anti-inflammatory activity of Clerodendrum serratum (Linn) Moon and Solanum xanthocarpum Schrad and Wendl in wistar ablino rats. The Journal of Phytopharmacology 5 (2016) 38-44. http://dx.doi.org/10.31254/phyto.2016.5201

H. Rabiul, M. Subhasish, S. Sinha, M.G. Roy, D. Sinha, S. Gupta. Hepatoprotective activity of clerodendron inerme against paracetamol induced hepatic injury in rats for pharmaceutical product. International Journal of Drug Development and Research 3 (2011) 118-126. https://www.scholarscentral.com/pdfs/109115/hepatoprotective-activity-of-clerodendron-inerme-againstparacetamol-induced-hepatic-injury-in-rats-for-pharmaceutical-product.pdf

T. Payum. Phytoconstituents and proximate composition of clerodendrum colebrookianum walp.: A widely used anti high blood pressure medicinal food plant in eastern himalayas. Pharmacognosy Journal 12 (2020) 1534-1540. http://dx.doi.org/10.5530/pj.2020.12.210

M.K. Kar, T.R. Swain, S.K. Mishra. Antidiabetic activity of clerodendrum philippinum schauer leaves in streptozotocin induced diabetic rats. International Journal of Pharmacy and Pharmaceutical Sciences 7 (2015) 386-389. https://journals.innovareacademics.in/index.php/ijpps/article/view/7428

Ł. Kuźma, J. Gomulski. Biologically Active Diterpenoids in the Clerodendrum Genus—A Review. International Journal of Molecular Sciences 23 (2022) 11001. https://doi.org/10.3390/ijms231911001

J.H. Wang, F. Luan, X.D. He, Y. Wang, M.X. Li. Traditional uses and pharmacological properties of Clerodendrum phytochemicals. Journal of Traditional and Complementary Medicine 8 (2018) 24-38. https://doi.org/10.1016/j.jtcme.2017.04.001

E.L. Santos, B.H.L.N.S. Maia, A.P. Ferriani, S.D. Teixeira. Flavonoids: Classification, Biosynthesis and Chemical Ecology. in: Flavonoids - From Biosynthesis to Human Health, IntechOpen Limited, London, UK, 2017, pp. 1-16. http://dx.doi.org/10.5772/67861

B.H. Havsteen. The biochemistry and medical significance of the flavonoids. Pharmacology and Therapeutics 96 (2002) 67-202. https://doi.org/10.1016/s0163-7258(02)00298-x

R.A. Dixon, G.M. Pasinetti. Flavonoids and isoflavonoids: From plant biology to agriculture and neuroscience. Plant Physiology 154 (2010) 453-457. https://doi.org/10.1104/pp.110.161430

M.C. Dias, D.C.G.A. Pinto, A.M.S. Silva. Plant flavonoids: Chemical characteristics and biological activity. Molecules 26 (2021) 5377. https://doi.org/10.3390/molecules26175377

W. Feng, Z. Hao, M. Li. Isolation and Structure Identification of Flavonoids. in: Flavonoids - From Biosynthesis to Human Health, IntechOpen Limited, London, UK, pp. 17-43. http://dx.doi.org/10.5772/67810

S. Chen, X. Wang, Y. Cheng, H. Gao, X. Chen. A Review of Classification, Biosynthesis, Biological Activities and Potential Applications of Flavonoids. Molecules 28 (2023) 4982. https://doi.org/10.3390/molecules28134982

A.N. Panche, A.D. Diwan, S.R. Chandra. Flavonoids: An overview. Journal of Nutritional Science 5 (2016) e47. https://doi.org/10.1017/jns.2016.41

W. Liu, X. Cui, Y. Zhong, R. Ma, B. Liu, Y. Xia. Phenolic metabolites as therapeutic in inflammation and neoplasms: Molecular pathways explaining their efficacy. Pharmacological Research 193 (2023) 106812. https://doi.org/10.1016/j.phrs.2023.106812

K.G. Prasanth, A. Anandbabu, R. Venkatanarayanan, B. Dineshkumar, V. Sankar. HPTLC Technique: Determination of flavonoid from Clerodendrum viscosum vent roots. Der Pharma Chemica 4 (2012) 926-929. https://www.derpharmachemica.com/pharma-chemica/hptlc-technique-determination-of-flavonoid-from-clerodendrum-viscosum-vent-roots.pdf

J. Zhou, Q. Yang, X. Zhu, T. Lin, D. Hao, J. Xu. Antioxidant activities of clerodendrum cyrtophyllum turcz leaf extracts and their major components. PLoS ONE 15 (2020) e0234435. https://doi.org/10.1371/journal.pone.0234435

J.O. Chaves, M.C. de Souza, L.C. da Silva, D. Lachos-Perez, P.C. Torres-Mayanga, A.P. da F. Machado, T. Forster-Carneiro, M. Vázquez-Espinosa, A.V. González-de-Peredo, G.F. Barbero, M.A. Rostagno. Extraction of Flavonoids From Natural Sources Using Modern Techniques. Frontiers in Chemistry 8 (2020) 1-25. https://doi.org/10.3389/fchem.2020.507887

D.K. Magozwi, M. Dinala, N. Mokwana, X. Siwe-Noundou, R.W.M. Krause, M. Sonopo, L.J. McGaw, W.A. Augustyn, V.J. Tembu. Flavonoids from the genus euphorbia: Isolation, structure, pharmacological activities and structure-activity relationships. Pharmaceuticals 14 (2021) 428. https://doi.org/10.3390/ph14050428

W. Li, X. Zhang, S. Wang, X. Gao, X. Zhang. Research Progress on Extraction and Detection Technologies of Flavonoid Compounds in Foods. Foods 13 (2024) 628. https://doi.org/10.3390/foods13040628

T.R. Prashith Kekuda, S.J. Sudharshan. Ethnobotanical uses, phytochemistry and biological activities of Clerodendrum paniculatum L. (Lamiaceae): A comprehensive review. Journal of Drug Delivery and Therapeutics 8 (2018) 28-34. https://jddtonline.info/index.php/jddt/article/view/1930

P.N. Leena, N.A. Aleykutty. Isolation and Spectral identification of Quercetin fron the alcoholic root extract of Clerodendrum paniculatum. International Journal of Pharma Sciences and Research 7 (2016) 47-50. http://www.ijpsr.info/docs/IJPSR16-07-01-005.pdf

R. Kopilakkal, K. Chanda, M.M. Balamurali. Hepatoprotective and Antioxidant Capacity of Clerodendrum paniculatum Flower Extracts against Carbon Tetrachloride-Induced Hepatotoxicity in Rats. ACS Omega 6 (2021) 26489-26498. https://doi.org/10.1021/acsomega.1c03722

D. Pertiwi, P. Sitorus, I. Hafiz, D. Satria. Analysis of Component and Antibacterial Activity of Ethanol Extract and Etyl Acetate Fraction of Pagoda (Clerodendrum paniculatum L.) Leaves against Pseudomonas aeruginosa and MRSA. Research Journal of Pharmacy and Technology 15 (2022) 3047-3050. https://doi.org/10.52711/0974-360X.2022.00509

I. Hafiz, Rosidah, J. Silalahi. Antioxidant and anti-inflammatory activity of pagoda leaves (Clerodendrum paniculatum l.) ethanolic extract in white male rats (Rattus novergicus). International Journal of PharmTech Research 9 (2016) 165-170. https://sphinxsai.com/2016/ph_vol9_no5/1/(165-170)V9N5PT.pdf

N.P. Hegde, B.S. Hungund. Phytochemical profiling of Clerodendrum paniculatum leaf extracts: GC-MS, LC-MS analysis and comparative evaluation of antimicrobial, antioxidant & cytotoxic effects. Natural Product Research (2022) 2957-2964. https://doi.org/10.1080/14786419.2022.2140339

Venkatesh S, Aswani K, Asheena Asharaf V V, Anjitha P, Suresh A, Babu G. Anti-diabetic activity of Clerodendrum paniculatum leaves by In-vitro, In-vivo and Ex-vivo methods. GSC Biological and Pharmaceutical Sciences 16 (2021) 211-218. https://doi.org/10.30574/gscbps.2021.16.1.0210

S. Varghese, P. Kannappan, D. Kanakasabapathi, S.R. Madathil, M. Perumalsamy. Antidiabetic and antilipidemic effect of Clerodendrum paniculatum flower ethanolic extract. An in vivo investigation in Albino Wistar rats. Biocatalysis and Agricultural Biotechnology 36 (2021) 102095. https://doi.org/10.1016/j.bcab.2021.102095

K. Priyanka, I. Kuppast, B. Ramesh, S. Gururaj, H. Annegowda. Screening of aerial parts of the plant Clerodendrum paniculatum Linn for anti-anxiety activity. GSC Biological and Pharmaceutical Sciences 8 (2019) 46-50. http://dx.doi.org/10.30574/gscbps.2019.8.1.0159

S. Sundaraganapathy, P.N. L.eena. Development and Assessment of Anti Cancer Activity of Phytosome Using Isolated Compound from Clerodendron Paniculatum Linn Root Extract. International Journal of Pharma Research and Health Sciences 4 (2016) 1399-1402. https://pharmahealthsciences.net/pdfs/volume4-issue52016/11.vol4-issue5-2016-MS-15331.pdf

M. Barman, A. Barman, S. Ray. Clerodendrum inerme (L.) Gaertn.: a critical review on current progress in traditional uses, phytochemistry, pharmacological aspects and toxicity. Phytochemistry Reviews (2024). http://dx.doi.org/10.1007/s11101-024-09934-y

W.J. Huang, H.J. Lee, H.L. Chen, P.C. Fan, Y.L. Ku, L.C. Chiou. Hispidulin, a constituent of Clerodendrum inerme that remitted motor tics, alleviated methamphetamine-induced hyperlocomotion without motor impairment in mice. Journal of Ethnopharmacology 166 (2015) 18-22. https://doi.org/10.1016/j.jep.2015.03.001

B. Achari, C. Chaudhuri, C.R. Saha, P.K. Dutta, S.C. Pakrashi. A clerodane diterpene and other constituents of Clerodendron inerme. Phytochemistry 29 (1990) 3671-3673. https://doi.org/10.1016/0031-9422(90)85302-V

S.R.M. Ibrahim, K.Z. Alshali, M.A. Fouad, E.S. Elkhayat, R.A. Al Haidari, G.A. Mohamed. Chemical constituents and biological investigations of the aerial parts of Egyptian Clerodendrum inerme. Bulletin of Faculty of Pharmacy, Cairo University 52 (2014) 165-170. https://doi.org/10.1016/j.bfopcu.2014.05.002

Y.S. Yankanchi, S.A. Koli. Anti-inflammatory and Analgesic activity of mature leaves methanol extract of clerodendrum inerme L. (Gaertn). Journal of Pharmaceutical Sciences and Research 2 (2010) 782-785. https://www.europub.co.uk/articles/anti-inflammatory-and-analgesic-activity-of-mature-leaves-methanol-extract-of-clerodendrum-inerme-l-gaertn-A-155215

M. Nindatu, F. Noya, Y. Taihuttu. Efektivitas antimalaria rebusan tanaman lamburung meit (Clerondrum inerme Linn) pada penderita malaria di daerah pelayanan puskesmas Kairatu Barat, Kabupaten Seram Barat, Maluku. Molucca Medica (2018) 11-19. http://dx.doi.org/10.30598/molmed.2018.v11.i2.11

S.A. Khan, N. Rasool, M. Riaz, R. Nadeem, U. Rashid, K. Rizwan, M. Zubair, I.H. Bukhari, T. Gulzar. Evaluation of antioxidant and cytotoxicity studies of clerodendrum inerme. Asian Journal of Chemistry 25 (2013) 7457-7462. http://dx.doi.org/10.14233/ajchem.2013.14831

P.C. Fan, W.J. Huang, L.C. Chiou. Intractable chronic motor tics dramatically respond to clerodendrum inerme (L) gaertn. Journal of Child Neurology 24 (2009) 887-890. https://doi.org/10.1177/0883073808331088

H.L. Chen, H.J. Lee, W.J. Huang, J.F. Chou, P.C. Fan, J.C. Du, Y.L. Ku, L.C. Chiou. Clerodendrum inerme leaf extract alleviates animal behaviors, hyperlocomotion, and prepulse inhibition disruptions, mimicking tourette syndrome and schizophrenia. Evidence-Based Complementary and Alternative Medicine (2012) 284301. https://doi.org/10.1155/2012/284301

Y. Chowdhary. Chemical Composition of Clerodendrum Phlomidis: A Review. Asian Journal of Research in Pharmaceutical Sciences 12:2 (2022) 133-136. https://doi.org/10.52711/2231-5659.2022.00022

P.R.S. Rajamanoharan. An ethno botanical survey of medicinal plants in Sillalai, Jaffna, Northern Province, Sri Lanka. International Journal of Herbal Medicine 1:4 (2014) 22-30. https://www.cabidigitallibrary.org/doi/pdf/10.5555/20143146451

J. Santosh Kumar, M. Krishna Chaitanya, A.J. Semotiuk, V. Krishna. Indigenous knowledge of medicinal plants used by ethnic communities of South India. Ethnobotany Research and Applications 18 (2019) 1-112. http://dx.doi.org/10.32859/era.18.4.1-112

M.K. Mohan Maruga Raja, S.H. Mishra. Comprehensive review of Clerodendrum phlomidis: A traditionally used bitter. Journal of Chinese Integrative Medicine 8 (2010) 510-524. http://dx.doi.org/10.3736/jcim20100602

C. Muthu, A.D. Reegan, S. Kingsley, S. Ignacimuthu. Larvicidal activity of pectolinaringenin from Clerodendrum phlomidis L. against Culex quinquefasciatus Say and Aedes aegypti L. (Diptera: Culicidae). Parasitology Research 111 (2012) 1059-1065. https://doi.org/10.1007/s00436-012-2932-8

Y.P. Bharitkar, A. Hazra, S. Shah, S. Saha, A.K. Matoori, N.B. Mondal. New flavonoid glycosides and other chemical constituents from Clerodendrum phlomidis leaves: Isolation and characterisation. Natural Product Research 29 (2015) 1850-1856. https://doi.org/10.1080/14786419.2015.1009457

A.K. Yadav, J.P. Thakur, J. Agrawal, D. Saikia, A. Pal, M.M. Gupta. Bioactive chemical constituents from the root of Clerodendrum phlomidis. Medicinal Chemistry Research 24 (2015) 1112-1118. https://doi.org/10.1007/s00044-014-1191-x

Y. Vaghasiya, S. V. Chanda. Screening of methanol and acetone extracts of fourteen Indian medicinal plants for antimicrobial activity. Turkish Journal of Biology 31 (2007) 243-248. https://journals.tubitak.gov.tr/cgi/viewcontent.cgi?article=2053&context=biology

S.P. Dhanabal, M.K.M. Marugaraja, B. Suresh. Antidiabetic activity of Clerodendron phlomoidis leaf extract in alloxan-induced diabetic rats. Indian Journal of Pharmaceutical Sciences 70 (2008) 841-844. https://pmc.ncbi.nlm.nih.gov/articles/PMC3040893/pdf/IJPhS-70-841.pdf

V.R. Chidrawar, K.N. Patel, H.R. Chitme, S.S. Shiromwar. Pre-clinical evolutionary study of Clerodendrum phlomidis as an anti-obesity agent against high fat diet induced C57BL/6J mice. Asian Pacific Journal of Tropical Biomedicine 2 (2012) S1509-1519. https://doi.org/10.1016/S2221-1691(12)60446-8

J. Bhangale, R. V Patel, R. Jat, J.O. Bhangale. Antiarthritic activity of crude extract of Clerodendrum phlomidis (L.) leaves in FCA induced arthritis in rats. World Journal of Pharmaceutical Research 9 (2020) 715-729. https://www.researchgate.net/publication/348693789_ANTIARTHRITIC_ACTIVITY_OF_CRUDE_EXTRACT_OF_CLERODENDRUM_PHLOMIDIS_L_LEAVES_IN_FCA_INDUCED_ARTHRITIS_IN_RATS

R.H. Gokani, S.K. Lahiri, D.D. Santani, M.B. Shah. Evaluation of immunomodulatory activity of Clerodendrum phlomidis and Premna integrifolia root. International Journal of Pharmacology 3 (2007) 352-356. https://doi.org/10.3923/ijp.2007.352.356

H. Joshi, K. Megeri. Antiamnesic evaluation of C. phlomidis Linn. bark extract in mice. Revista Brasileira de Ciencias Farmaceuticas/Brazilian Journal of Pharmaceutical Sciences 44 (2008) 717-725. https://doi.org/10.1590/S1516-93322008000400019

S. Rani, N. Ahamed, S. Rajaram, R. Saluja, S. Thenmozhi, T. Murugesan. Anti-diarrhoeal evaluation of Clerodendrum phlomidis Linn. leaf extract in rats. Journal of Ethnopharmacology 68 (1999) 315-319. https://doi.org/10.1016/S0378-8741(99)00103-8

J.M. Brimson, N. Onlamoon, T. Tencomnao, P. Thitilertdecha. Clerodendrum petasites S. Moore: The therapeutic potential of phytochemicals, hispidulin, vanillic acid, verbascoside, and apigenin. Biomedicine and Pharmacotherapy 118 (2019) 1-6. https://doi.org/10.1016/j.biopha.2019.109319

C. Singharachai, C. Palanuvej, H. Kiyohara, H. Yamada, N. Ruangrungsi. Pharmacognostic specification of five root species in Thai traditional medicine remedy: Ben-Cha-Lo-Ka-Wi-Chian. Pharmacognosy Journal 3 (2011) 1-11. https://doi.org/10.5530/pj.2011.21.1

A. Panthong, D. Kanjanapothi, W.C. Taylor. Ethnobotanical review of medicinal plants from thai traditional books, Part I: Plants with anti-inflammatory, anti-asthmatic and antihypertensive properties. Journal of Ethnopharmacology 18 (1986) 213-228. https://doi.org/10.1016/0378-8741(86)90001-2

A. Hazekamp, R. Verpoorte, A. Panthong. Isolation of a bronchodilator flavonoid from the Thai medicinal plant Clerodendrum petasites. Journal of Ethnopharmacology 78 (2001) 45-49. https://doi.org/10.1016/S0378-8741(01)00320-8

Hasriadi, A. Jongchanapong, W. Thongphichai, P.W. Dasuni Wasana, S. Sukrong, R. Suttisri, S. Amnuoypol, P. Towiwat. Antinociceptive efficacy of Clerodendrum petasites S. Moore, a Thai medicinal plant, and its CNS safety profiles. Journal of Traditional and Complementary Medicine 13 (2023) 81-92. https://doi.org/10.1016/j.jtcme.2022.11.001

P. Arjsri, K. Srisawad, S. Mapoung, W. Semmarath, P. Thippraphan, S. Umsumarng, S. Yodkeeree, P. Dejkriengkraikul. Hesperetin from Root Extract of Clerodendrum petasites S. Moore Inhibits SARS-CoV-2 Spike Protein S1 Subunit-Induced NLRP3 Inflammasome in A549 Lung Cells via Modulation of the Akt/MAPK/AP-1 Pathway. International Journal of Molecular Sciences 23 (2022) 10346. https://doi.org/10.3390/ijms231810346

T. Kwuansawat, W. Putalun, W. Tassaneeyakul, P. Mahakunakorn. Phytochemical constituents and anti-inflammatory activities of Clerodendrum petasites. International Journal of Health Sciences (2022) 3497-3510. https://doi.org/10.53730/ijhs.v6nS1.5542

P. Thitilertdecha, R.H. Guy, M.G. Rowan. Characterisation of polyphenolic compounds in Clerodendrum petasites S. Moore and their potential for topical delivery through the skin. Journal of Ethnopharmacology 154 (2014) 400-407. https://doi.org/10.1016/j.jep.2014.04.021

A. Panthong, D. Kanjanapothi, T. Taesotikul, T. Wongcome, V. Reutrakul. Anti-inflammatory and antipyretic properties of Clerodendrum petasites S. Moore. Journal of Ethnopharmacology 85 (2003) 151-156. https://doi.org/10.1016/S0378-8741(02)00368-9

P. Thitilertdecha, M.G. Rowan, R.H. Guy. Topical formulation and dermal delivery of active phenolic compounds in the Thai medicinal plant - Clerodendrum petasites S. Moore. International Journal of Pharmaceutics 478 (2015) 39-45. https://doi.org/10.1016/j.ijpharm.2014.11.004

A.A. Ajao, O.M. Oseni, O.T. Oladipo, Y.A. Adams, Y.O. Mukaila, A.A. Ajao. Clerodendrum volubile P. Beauv (Lamiaceae), an underutilized indigenous vegetable of utmost nutritive and pharmacological importance. Beni-Suef University Journal of Basic and Applied Sciences 7 (2018) 606-611. https://doi.org/10.1016/j.bjbas.2018.07.003

K. Okaiyeto, A.O. Falade, O.O. Oguntibeju. Traditional uses, nutritional and pharmacological potentials of clerodendrum volubile. Plants 10 (2021) 1893. https://doi.org/10.3390/plants10091893

O.L. Erukainure, M.A. Mesaik, O. Atolani, A. Muhammad, C.I. Chukwuma, M.S. Islam. Pectolinarigenin from the leaves of Clerodendrum volubile shows potent immunomodulatory activity by inhibiting T − cell proliferation and modulating respiratory oxidative burst in phagocytes. Biomedicine and Pharmacotherapy 93 (2017) 529-535. https://doi.org/10.1016/j.biopha.2017.06.060

R.N. Ugbaja, T.F. Akinhanmi, A.S. James, E.I. Ugwor, A.A. Babalola, E.O. Ezenandu, V.C. Ugbaja, E.A. Emmanuel. Flavonoid-rich fractions from Clerodendrum volubile and Vernonia amygdalina extenuates arsenic-invoked hepato-renal toxicity via augmentation of the antioxidant system in rats. Clinical Nutrition Open Science 35 (2021) 12-25. https://doi.org/10.1016/j.nutos.2020.12.003

O.L. Erukainure, M.Z. Zaruwa, M.I. Choudhary, S.A. Naqvi, N. Ashraf, R.M. Hafizur, A. Muhammad, O.A.T. Ebuehi, G.N. Elemo. Dietary fatty acids from leaves of Clerodendrum volubile induce cell cycle arrest, downregulate matrix metalloproteinase-9 expression, and modulate redox status in human breast cancer. Nutrition and Cancer 68 (2016) 634-645. https://doi.org/10.1080/01635581.2016.1156714

O.L. Erukainure, N. Narainpersad, M. Singh, S. Olakunle, M.S. Islam. Clerodendrum volubile inhibits key enzymes linked to type 2 diabetes but induces cytotoxicity in human embryonic kidney (HEK293) cells via exacerbated oxidative stress and proinflammation. Biomedicine and Pharmacotherapy 106 (2018) 1144-1152. https://doi.org/10.1016/j.biopha.2018.07.013

A. Saheed, G. Olufunke, Olorundare Gideon, S. Deeba N, M. Hasan, R. Albrecht, K. Mamoru. Cytotoxic Potentials of Clerodendrum Volubile against Prostate Cancer Cells and Its Possible Proteomic Targets. Journal of Clinical Nutrition and Food Science 2 (2019) 46-53. https://www.researchgate.net/publication/339075343_Cytotoxic_Potentials_of_Clerodendrum_Volubile_against_Prostate_Cancer_Cells_and_Its_Possible_Proteomic_Targets

T.H. Ogunwa, T.T. Adeyelu, R.Y. Fasimoye, M.B. Oyewale, T.A. Ademoye, O.C. Ilesanmi, O.B. Awe, S.A. Ajiboye, B.O. Oloye, D.R. Sholanke. Phytochemical evaluation and in vitro antioxidant status of Clerodendrum volubile (an indigenous medicinal plant). Pakistan Journal of Pharmaceutical Research 2 (2016) 77-88. https://journals.indexcopernicus.com/api/file/viewByFileId/120019

S.A. Adefegha, G. Oboh. Antioxidant and inhibitory properties of Clerodendrum volubile leaf extracts on key enzymes relevant to non-insulin dependent diabetes mellitus and hypertension. Journal of Taibah University for Science 10 (2016) 521-533. https://doi.org/10.1016/j.jtusci.2015.10.008

R.K. Verma, T. Paraidathathu. Herbal medicines used in the traditional Indian medicinal system as a therapeutic treatment option for overweight and obesity management: A review. International Journal of Pharmacy and Pharmaceutical Sciences 6 (2014) 40-47. https://www.researchgate.net/publication/260677281_Herbal_medicines_used_in_the_traditional_Indian_medicinal_system_as_a_therapeutic_treatment_option_for_overweight_and_obesity_management_A_review

R.N. Jadeja, M.C. Thounaojam, T.B. Singh, R. V. Devkar, A. V. Ramachandran. Traditional uses, phytochemistry and pharmacology of Clerodendron glandulosum Coleb - a review. Asian Pacific Journal of Tropical Medicine 5 (2012) 1-6. https://doi.org/10.1016/S1995-7645(11)60236-8

P.K. Deb, P. Khound, S. Bhattacharjee, P. Choudhury, H. Sarma, R. Devi, B. Sarkar. Variation in chemical constituents, in-vitro bioactivity and toxicity profile among different parts of Clerodendrum glandulosum Lindl. (C. colebrookianum Walp.). South African Journal of Botany 140 (2021) 50-61. https://doi.org/10.1016/j.sajb.2021.03.023

P. Khound, P.K. Deb, S. Bhattacharjee, K.D. Medina, P.P. Sarma, B. Sarkar, R. Devi. Phenolic enriched fraction of Clerodendrum glandulosum Lindl. leaf extract ameliorates hyperglycemia and oxidative stress in streptozotocin-nicotinamide induced diabetic rats. Journal of Ayurveda and Integrative Medicine 15 (2024) 100906. https://doi.org/10.1016/j.jaim.2024.100906

R.N. Jadeja, M.C. Thounaojam, Ansarullah, V.B. Patel, R. V. Devkar, A. V. Ramachandran. Protective effect of Clerodendron glandulosum extract against experimentally induced metabolic syndrome in rats. Pharmaceutical Biology 48 (2010) 1312-1319. https://doi.org/10.3109/13880201003739304

R.N. Jadeja, M.C. Thounaojam, U. V. Ramani, R. V. Devkar, A. V. Ramachandran. Anti-obesity potential of Clerodendron glandulosum.Coleb leaf aqueous extract. Journal of Ethnopharmacology 135 (2011) 338-343. https://doi.org/10.1016/j.jep.2011.03.020

R. Jadeja, M. Thounaojam, Ansarullah, A. V. Ramachandran, R. Devkar. Phytochemical constituents and free radical scavenging activity of Clerodendron glandulosum.Coleb methanolic extract. Journal of Complementary and Integrative Medicine 6 (2009). https://doi.org/10.2202/1553-3840.1226.

R.N. Jadeja, M.C. Thounaojam, Ansarullah, R. V. Devkar, A. V. Ramachandran. Clerodendron glandulosum Coleb., Verbenaceae, ameliorates high fat diet-induced alteration in lipid and cholesterol metabolism in rats. Revista Brasileira de Farmacognosia 20 (2010) 117-123. http://dx.doi.org/10.1590/S0102-695X2010000100023

R.N. Jadeja, M.C. Thounaojam, Ansarullah, R. V. Devkar, A. V. Ramachandran. A preliminary study on hypolipidemic effect of aqueous leaf extract of Clerodendron glandulosum.Coleb. International Journal of Green Pharmacy 3 (2009) 285-289. https://www.greenpharmacy.info/index.php/ijgp/article/view/102

J.M. Davis, E.A. Murphy, M.D. Carmichael. Effects of the dietary flavonoid quercetin upon performance and health. Current Sports Medicine Reports 8 (2009) 206-213. http://dx.doi.org/10.1249/JSR.0b013e3181ae8959

P.S. Bustos, R. Deza-Ponzio, P.L. Páez, I. Albesa, J.L. Cabrera, M.B. Virgolini, M.G. Ortega. Protective effect of quercetin in gentamicin-induced oxidative stress in vitro and in vivo in blood cells. Effect on gentamicin antimicrobial activity. Environmental Toxicology and Pharmacology 48 (2016) 253-264. https://doi.org/10.1016/j.etap.2016.11.004

R. Amorati, A. Baschieri, A. Cowden, L. Valgimigli. The antioxidant activity of quercetin in water solution. Biomimetics 2 (2017) 9. https://doi.org/10.3390/biomimetics2030009

J.H. Kim, M.J. Kang, H.N. Choi, S.M. Jeong, Y.M. Lee, J.I. Kim. Quercetin attenuates fasting and postprandial hyperglycemia in animal models of diabetes mellitus. Nutrition Research and Practice 5 (2011) 107-111. https://doi.org/10.4162/nrp.2011.5.2.107

I.U.H. Bhat, R. Bhat. Quercetin: A bioactive compound imparting cardiovascular and neuroprotective benefits: Scope for exploring fresh produce, their wastes, and by-products. Biology 10 (2021) 586. https://doi.org/10.3390/biology10070586

S. Chen, H. Jiang, X. Wu, J. Fang. Therapeutic Effects of Quercetin on Inflammation, Obesity, and Type 2 Diabetes. Mediators of Inflammation 2016 (2016) 340637. https://doi.org/10.1155/2016/9340637

M.C. Serban, A. Sahebkar, A. Zanchetti, D.P. Mikhailidis, G. Howard, D. Antal, F. Andrica, A. Ahmed, W.S. Aronow, P. Muntner, G.Y.H. Lip, I. Graham, N. Wong, J. Rysz, M. Banach. Effects of Quercetin on Blood Pressure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Journal of the American Heart Association 5 (2016). https://doi.org/10.1161/jaha.115.002713.

D. Szwajgier, K. Borowiec, J. Zapp. Activity-guided purification reveals quercetin as the most efficient cholinesterase inhibitor in wild strawberry (Fragaria vesca L.) and apricot (Prunus armeniaca L.) fruit extract. Emirates Journal of Food and Agriculture 31 (2019) 386-394. https://doi.org/10.9755/ejfa.2019.v31.i5.1957

A. Rauf, M. Imran, I.A. Khan, M. ur-Rehman, S.A. Gilani, Z. Mehmood, M.S. Mubarak. Anticancer potential of quercetin: A comprehensive review. Phytotherapy Research 32 (2018) 2109-2130. https://doi.org/10.1002/ptr.6155

H.A.S. El-Nashar, M.I. Gamal El-Din, L. Hritcu, O.A. Eldahshan. Insights on the inhibitory power of flavonoids on tyrosinase activity: A survey from 2016 to 2021. Molecules 26 (2021) 7546. https://doi.org/10.3390/molecules26247546

N. Salem Alrawaiq, A. Abdullah. A review of flavonoid quercetin: Metabolism, Bioactivity and antioxidant properties. International Journal of PharmTech Research 6 (2014) 933-941. https://sphinxsai.com/2014/phvolpt3/1/(933-941)Jul-Aug14.pdf

Chen Yuting, Zheng Rongliang, Jia Zhongjian, Ju Yong. Flavonoids as superoxide scavengers and antioxidants. Free Radical Biology and Medicine 9 (1990) 19-21. https://doi.org/10.1016/0891-5849(90)90045-K

C. Manach, C. Morand, O. Texier, M.L. Favier, G. Agullo, C. Demigne, F. Regerat, C. Remesy. Quercetin metabolites in plasma of rats fed diets containing rutin or quercetin. Journal of Nutrition 125 (1995) 1911-1922. https://doi.org/10.1093/jn/125.7.1911

A.N. Begum, J. Terao. Protective effect of quercetin against cigarette tar extract-induced impairment of erythrocyte deformability. Journal of Nutritional Biochemistry 13 (2002) 265-272. https://doi.org/10.1016/S0955-2863(01)00219-4

A.D. Mariee, G.M. Abd-Allah, H.A. El-Beshbishy. Protective effect of dietary flavonoid quercetin against lipemic-oxidative hepatic injury in hypercholesterolemic rats. Pharmaceutical Biology 50 (2012) 265-272. https://doi.org/10.1016/S0955-2863(01)00219-4

A. Yarahmadi, F. Khademi, Z. Mostafavi-Pour, F. Zal. In-Vitro Analysis of Glucose and Quercetin Effects on m-TOR and Nrf-2 Expression in HepG2 Cell Line (Diabetes and Cancer Connection). Nutrition and Cancer 70 (2018) 770-775. https://doi.org/10.1080/01635581.2018.1470654

M. Vessal, M. Hemmati, M. Vasei. Antidiabetic effects of quercetin in streptozocin-induced diabetic rats. Comparative Biochemistry and Physiology - C Toxicology and Pharmacology 135 (2003) 357-364. https://doi.org/10.1016/S1532-0456(03)00140-6

O. Coskun, M. Kanter, A. Korkmaz, S. Oter. Quercetin, a flavonoid antioxidant, prevents and protects streptozotocin-induced oxidative stress and β-cell damage in rat pancreas. Pharmacological Research 51 (2005) 117-123. https://doi.org/10.1016/j.phrs.2004.06.002

M. Kobori, S. Masumoto, Y. Akimoto, Y. Takahashi. Dietary quercetin alleviates diabetic symptoms and reduces streptozotocin-induced disturbance of hepatic gene expression in mice. Molecular Nutrition and Food Research 53 (2009) 859-868. https://doi.org/10.1002/mnfr.200800310

S. Wein, N. Behm, R.K. Petersen, K. Kristiansen, S. Wolffram. Quercetin enhances adiponectin secretion by a PPAR-γ independent mechanism. European Journal of Pharmaceutical Sciences 41 (2010) 16-22. https://doi.org/10.1016/j.ejps.2010.05.004

O.M. Ahmed, T. Mohamed, H. Moustafa, H. Hamdy, R.R. Ahmed, E. Aboud. Quercetin and low level laser therapy promote wound healing process in diabetic rats via structural reorganization and modulatory effects on inflammation and oxidative stress. Biomedicine and Pharmacotherapy 101 (2018) 58-73. https://doi.org/10.1016/j.biopha.2018.02.040

O. Maksymchuk, A. Shysh, I. Rosohatska, M. Chashchyn. Quercetin prevents type 1 diabetic liver damage through inhibition of CYP2E1. Pharmacological Reports 69 (2017) 1386-1392. https://doi.org/10.1016/j.pharep.2017.05.020

R.M. Maciel, F.B. Carvalho, A.A. Olabiyi, R. Schmatz, J.M. Gutierres, N. Stefanello, D. Zanini, M.M. Rosa, C.M. Andrade, M.A. Rubin, M.R. Schetinger, V.M. Morsch, C.C. Danesi, S.T.A. Lopes. Neuroprotective effects of quercetin on memory and anxiogenic-like behavior in diabetic rats: Role of ectonucleotidases and acetylcholinesterase activities. Biomedicine and Pharmacotherapy 84 (2016) 559-568. https://doi.org/10.1016/j.biopha.2016.09.069

C.S. Kim, Y. Kwon, S.Y. Choe, S.M. Hong, H. Yoo, T. Goto, T. Kawada, H.S. Choi, Y. Joe, H.T. Chung, R. Yu. Quercetin reduces obesity-induced hepatosteatosis by enhancing mitochondrial oxidative metabolism via heme oxygenase-1. Nutrition and Metabolism 12 (2015) 33. https://doi.org/10.1186/s12986-015-0030-5

Z. Jing, Z. Wang, X. Li, X. Li, T. Cao, Y. Bi, J. Zhou, X. Chen, D. Yu, L. Zhu, S. Li. Protective Effect of Quercetin on Posttraumatic Cardiac Injury. Scientific Reports 6 (2016) 30812 . https://doi.org/10.1038/srep30812

Y.M. Zhang, Z.Y. Zhang, R.X. Wang. Protective Mechanisms of Quercetin Against Myocardial Ischemia Reperfusion Injury. Frontiers in Physiology 11 (2020) 30812. https://doi.org/10.1038/srep30812

F. Elbarbry, K. Abdelkawy, N. Moshirian, A.M. Abdel-Megied. The antihypertensive effect of quercetin in young spontaneously hypertensive rats; role of arachidonic acid metabolism. International Journal of Molecular Sciences 21 (2020) 6554. https://doi.org/10.3390/ijms21186554

S. Muhammad, N. Fatima. In silico analysis and molecular docking studies of potential angiotensin-converting enzyme inhibitor using quercetin glycosides. Pharmacognosy Magazine 11 (2015) S123-S126. https://pmc.ncbi.nlm.nih.gov/articles/PMC4461951/

I.E. Orhan. Cholinesterase Inhibitory Potential of Quercetin towards Alzheimer’s Disease - A Promising Natural Molecule or Fashion of the Day? - A Narrowed Review. Current Neuropharmacology 19 (2020) 2205-2213. http://dx.doi.org/10.2174/1570159X18666201119153807

A.M. Sabogal-Guáqueta, J.I. Muñoz-Manco, J.R. Ramírez-Pineda, M. Lamprea-Rodriguez, E. Osorio, G.P. Cardona-Gómez. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 93 (2015) 134-145. https://doi.org/10.1016/j.neuropharm.2015.01.027

A.B. Granado-Serrano, M.A. Martín, L. Bravo, L. Goya, S. Ramos. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). Journal of Nutrition 136 (2006) 2715-2721. https://doi.org/10.1093/jn/136.11.2715

N. Gulati, B. Laudet, V.M. Zohrabian, R. Murali, M. Jhanwar-Uniyal. The antiproliferative effect of Quercetin in cancer cells is mediated via inhibition of the PI3K-Akt/PKB pathway. Anticancer Research 26 (2006) 1177-1181. https://ar.iiarjournals.org/content/anticanres/26/2A/1177.full.pdf

Y.H. Kim, Y.J. Lee. TRAIL apoptosis is enhanced by quercetin through Akt dephosphorylation. Journal of Cellular Biochemistry 100 (2007) 998-1009. https://doi.org/10.1002/jcb.21098

Q. Wu, P.W. Needs, Y. Lu, P.A. Kroon, D. Ren, X. Yang. Different antitumor effects of quercetin, quercetin-3′-sulfate and quercetin-3-glucuronide in human breast cancer MCF-7 cells. Food and Function 9 (2018) 1736-1746. https://doi.org/10.1039/C7FO01964E

A. Constantinou, R. Mehta, C. Runyan, K. Rao, A. Vaughan, R. Moon. Flavonoids as DNA topoisomerase antagonists and poisons: Structure-activity relationships. Journal of Natural Products 58 (1995) 217-225. https://doi.org/10.1021/np50116a009

M. Fan, G. Zhang, X. Hu, X. Xu, D. Gong. Quercetin as a tyrosinase inhibitor: Inhibitory activity, conformational change and mechanism. Food Research International 100 (2017) 226-233. https://doi.org/10.1016/j.foodres.2017.07.010

S. Zolghadri, A. Bahrami, M.T. Hassan Khan, J. Munoz-Munoz, F. Garcia-Molina, F. Garcia-Canovas, A.A. Saboury. A comprehensive review on tyrosinase inhibitors. Journal of Enzyme Inhibition and Medicinal Chemistry 34 (2019) 279-309. https://doi.org/10.1080/14756366.2018.1545767

K. Jakimiuk, S. Sari, R. Milewski, C.T. Supuran, D. Şöhretoğlu, M. Tomczyk. Flavonoids as tyrosinase inhibitors in in silico and in vitro models: basic framework of SAR using a statistical modelling approach. Journal of Enzyme Inhibition and Medicinal Chemistry 37 (2022) 421-430. https://doi.org/10.1080/14756366.2021.2014832

M. Fan, H. Ding, G. Zhang, X. Hu, D. Gong. Relationships of dietary flavonoid structure with its tyrosinase inhibitory activity and affinity. LWT 107 (2019) 25-34. https://doi.org/10.1016/j.lwt.2019.02.076

S. a. Park, J. Jegal, K.W. Chung, H.J. Jung, S.G. Noh, H.Y. Chung, J. Ahn, J. Kim, M.H. Yang. Isolation of tyrosinase and melanogenesis inhibitory flavonoids from Juniperus chinensis fruits. Bioscience, Biotechnology and Biochemistry 82 (2018) 2041-2048. https://doi.org/10.1080/09168451.2018.1511367

S.R. Alizadeh, M.A. Ebrahimzadeh. Quercetin derivatives: Drug design, development, and biological activities, a review. European Journal of Medicinal Chemistry 229 (2022) 49-56. https://doi.org/10.1016/j.ejmech.2021.114068

S. Singh, V. Kushwah, A.K. Agrawal, S. Jain. Insulin- and quercetin-loaded liquid crystalline nanoparticles: Implications on oral bioavailability, antidiabetic and antioxidant efficacy. Nanomedicine 13 (2018) 521-537. https://doi.org/10.2217/nnm-2017-0278

M.S. Valencia, M. Franco da Silva Júnior, F.H. Xavier Júnior, B. de Oliveira Veras, E. Fernanda de Oliveira Borba, T. Gonçalves da Silva, V.L. Xavier, M. Pessoa de Souza, M. das G. Carneiro-da-Cunha. Bioactivity and cytotoxicity of quercetin-loaded, lecithin-chitosan nanoparticles. Biocatalysis and Agricultural Biotechnology 31 (2021) 101879. https://doi.org/10.1016/j.bcab.2020.101879

R.G.R. Pinheiro, A. Granja, J.A. Loureiro, M.C. Pereira, M. Pinheiro, A.R. Neves, S. Reis. Quercetin lipid nanoparticles functionalized with transferrin for Alzheimer’s disease. European Journal of Pharmaceutical Sciences 148 (2020) 105314. https://doi.org/10.1016/j.ejps.2020.105314

W. Chen, X. Ju, R.E. Aluko, Y. Zou, Z. Wang, M. Liu, R. He. Rice bran protein-based nanoemulsion carrier for improving stability and bioavailability of quercetin. Food Hydrocolloids 108 (2020) 106042. https://doi.org/10.1016/j.foodhyd.2020.106042

N. Wu, Y. Zhang, J. Ren, A. Zeng, J. Liu. Preparation of quercetin-nicotinamide co-crystals and their evaluation under in vivo and in vitro conditions. RSC Advances 10 (2020) 21852-21859. https://doi.org/10.1039/D0RA03324C

M.H. Choi, S.H. Yang, D.S. Kim, N.D. Kim, H.J. Shin, K. Liu. Novel quercetin derivative of 3,7-dioleylquercetin shows less toxicity and highly potent tyrosinase inhibition activity. International Journal of Molecular Sciences 22 (2021) 4264. https://doi.org/10.3390/ijms22084264

D. Kashyap, A. Sharma, H.S. Tuli, K. Sak, V.K. Garg, H.S. Buttar, W.N. Setzer, G. Sethi. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. Journal of Functional Foods 48 (2018) 457-471. https://doi.org/10.1016/j.jff.2018.07.037

J. Madunić, I.V. Madunić, G. Gajski, J. Popić, V. Garaj-Vrhovac. Apigenin: A dietary flavonoid with diverse anticancer properties. Cancer Letters 413 (2018) 11-22. https://doi.org/10.1016/j.canlet.2017.10.041

C. Huang, Y.X. Wei, M.C. Shen, Y.H. Tu, C.C. Wang, H.C. Huang. Chrysin, abundant in Morinda citrifolia fruit water-EtOAc extracts, combined with apigenin synergistically induced apoptosis and inhibited migration in human breast and liver cancer cells. Journal of Agricultural and Food Chemistry 64 (2016) 4235-4245. https://doi.org/10.1021/acs.jafc.6b00766

S. Nabavi, S. Habtemariam, M. Daglia, S. Nabavi. Apigenin and Breast Cancers: From Chemistry to Medicine. Anti-Cancer Agents in Medicinal Chemistry 15 (2015) 728-735. http://dx.doi.org/10.2174/1871520615666150304120643

X. Xu, M. Li, W. Chen, H. Yu, Y. Yang, L. Hang. Apigenin Attenuates Oxidative Injury in ARPE-19 Cells thorough Activation of Nrf2 Pathway. Oxidative Medicine and Cellular Longevity 2016 (2016) 378461. https://doi.org/10.1155/2016/4378461

S.Q. Cai, Z.M. Tang, C. Xiong, F.F. Wu, J.R. Zhao, Q. Zhang, L. Wang, X.N. Zhang, X.H. Zhao. The anti-inflammatory effects of apigenin and genistein on the rat intestinal epithelial (IEC-6) cells with TNF-α stimulation in response to heat treatment. Current Research in Food Science 5 (2022) 918-926. https://doi.org/10.1016/j.crfs.2022.05.011

A.H. Rahmani, M.A. Alsahli, A. Almatroudi, M.A. Almogbel, A.A. Khan, S. Anwar, S.A. Almatroodi. The Potential Role of Apigenin in Cancer Prevention and Treatment. Molecules 27 (2022) 6051. https://doi.org/10.3390/molecules27186051

H. Sharma, R. Kanwal, N. Bhaskaran, S. Gupta. Plant flavone apigenin binds to nucleic acid bases and reduces oxidative DNA damage in prostate epithelial cells. PLoS ONE 9 (2014) e91588. https://doi.org/10.1371/journal.pone.0091588

S. Shukla, S. Gupta. Abstract 3804: Transcriptional repression of androgen receptor in human prostate cancer cells by plant flavone apigenin. Cancer Research 70 (2010) 3804. https://doi.org/10.1158/1538-7445.AM10-3804

B. Mafuvadze, Y. Liang, C. Besch-Williford, X. Zhang, S.M. Hyder. Apigenin Induces Apoptosis and Blocks Growth of Medroxyprogesterone Acetate-Dependent BT-474 Xenograft Tumors. Hormones and Cancer 3 (2012) 160-171. https://doi.org/10.1007/s12672-012-0114-x

P.S. Wu, J.H. Yen, M.C. Kou, M.J. Wu. Luteolin and apigenin attenuate 4-hydroxy- 2-nonenal-mediated cell death through modulation of UPR, Nrf2-ARE and MAPK pathways in PC12 cells. PLoS ONE 10 (2015) e0130599. https://doi.org/10.1371/journal.pone.0130599

S. Shukla, S. Gupta. Apigenin-induced cell cycle arrest is mediated by modulation of MAPK, PI3K-Akt, and loss of cyclin D1 associated retinoblastoma dephosphorylation in human prostate cancer cells. Cell Cycle 6 (2007) 1102-1114. https://doi.org/10.4161/cc.6.9.4146

R.H. Patil, R.L. Babu, M. Naveen Kumar, K.M. Kiran Kumar, S.M. Hegde, G.T. Ramesh, S. Chidananda Sharma. Apigenin inhibits PMA-induced expression of pro-inflammatory cytokines and AP-1 factors in A549 cells. Molecular and Cellular Biochemistry 403 (2015) 95-106. https://doi.org/10.1007/s11010-015-2340-3

O.H. Kang, J.H. Lee, D.Y. Kwon. Apigenin inhibits release of inflammatory mediators by blocking the NF-κB activation pathways in the HMC-1 cells. Immunopharmacology and Immunotoxicology 33 (2011) 473-479. https://doi.org/10.3109/08923973.2010.538851

D. Li, L. Wang, Y. Jing, B. Jiang, L. Zhao, Y. Miao, S. Xin, C. Ge. Exploring Molecular Targets and Mechanisms of Apigenin in the Treatment of Papillary Thyroid Carcinoma Based on Network Pharmacology and Molecular Docking Analysis. Natural Product Communications 17(10) (2022). https://doi.org/10.1177/1934578X221135435

R. Zhang, X. Hu, B. Zhang, Z. Wang, C. Hao, J. Xin, Q. Guo. Whitening Activity of Constituents Isolated from the Trichosanthes Pulp. Evidence-Based Complementary and Alternative Medicine 2020 (2020) 582579. https://doi.org/10.1155/2020/2582579

E.S. Karaoğlan, M. Koca. Tyrosinase, cholinesterase inhibitory activity and molecular docking studies on apigenin and vitexin. İstanbul Journal of Pharmacy 50 (2020) 268-271. http://dx.doi.org/10.26650/IstanbulJPharm.2019.0076

K. Chauhan, F. Goel, S. Singh. Apigenin protects melanocytes and improve tyrosinase activity in a hydroquinone induced vitiligo mouse model targeting P38 MAP kinase signaling: histopathology and immunohistochemistry analysis. Naunyn-Schmiedeberg’s Archives of Pharmacology (2023) 4859-4869. https://doi.org/10.1007/s00210-023-02917-4

Y. Ye, H. Wang, J.H. Chu, G.X. Chou, Z.L. Yu. Activation of p38 MAPK pathway contributes to the melanogenic property of apigenin in B16 cells. Experimental Dermatology 20 (2011) 755-757. https://doi.org/10.1111/j.1600-0625.2011.01297.x

L. Apaza Ticona, C. Thiebaut Estrada, Á. Rumbero Sánchez. Inhibition of melanin production and tyrosinase activity by flavonoids isolated from Loranthus acutifolius. Natural Product Research 35 (2021) 4690-4693. https://doi.org/10.1080/14786419.2019.1709185

L. Wang, Q. Ma. Clinical benefits and pharmacology of scutellarin: A comprehensive review. Pharmacology and Therapeutics 190 (2018) 105-127. https://doi.org/10.1016/j.pharmthera.2018.05.006

G.H. Zhang, Q. Wang, J.J. Chen, X.M. Zhang, S.C. Tam, Y.T. Zheng. The anti-HIV-1 effect of scutellarin. Biochemical and Biophysical Research Communications 334 (2005) 812-816. https://doi.org/10.1016/j.bbrc.2005.06.166

D. Wang, L. Wang, J. Gu, H. Yang, N. Liu, Y. Lin, X. Li, C. Shao. Scutellarin inhibits high glucose-induced and hypoxia-mimetic agent-induced angiogenic effects in human retinal endothelial cells through reactive oxygen species/hypoxia-inducible factor-1a/vascular endothelial growth factor pathway. Journal of Cardiovascular Pharmacology 64 (2014) 218-227. https://doi.org/10.1097/fjc.0000000000000109

H. Chen, Q. Du. Potential natural compounds for preventing SARS-CoV-2 (2019-nCoV) infection. Preprints (2020). http://dx.doi.org/10.20944/preprints202001.0358.v3

Q.Y. Jia, H.L. Chen, Z. Qi, X.L.N. Zhang, L.Y. Zheng, T.T. Liu, Y. Yuan, L. Yang, C.Y. Wu. Network pharmacology to explore the mechanism of scutellarin in the treatment of brain ischaemia and experimental verification of JAK2/STAT3 signalling pathway. Scientific Reports 13 (2023) 7557 1-11. https://doi.org/10.1038/s41598-023-33156-5

Y. Yuan, H. Zha, P. Rangarajan, E.A. Ling, C. Wu. Anti-inflammatory effects of Edaravone and Scutellarin in activated microglia in experimentally induced ischemia injury in rats and in BV-2 microglia. BMC Neuroscience 15 (2014) 125. https://doi.org/10.1186/s12868-014-0125-3

P. Luo, Z.H. Tan, Z.F. Zhang, H. Zhang, X.F. Liu, Z.J. Mo. Scutellarin isolated from Erigeron multiradiatus inhibits high glucose-mediated vascular inflammation. Yakugaku Zasshi 128 (2008) 1293-1299. https://doi.org/10.1248/yakushi.128.1293

L. Long, J. Wang, X. Lu, Y. Xu, S. Zheng, C. Luo, Y. Li. Protective effects of scutellarin on type II diabetes mellitus-induced testicular damages related to reactive oxygen species/Bcl-2/Bax and reactive oxygen species/microcirculation/staving pathway in diabetic rat. Journal of Diabetes Research 2015 (2015) 52530. https://doi.org/10.1155/2015/252530

Y. Su, W. Liu, L. Ma, X. Liu, Z. Liu, B. Zhu. Scutellarin inhibits translocation of protein kinase C in diabetic thoracic aorta of the rat. Clinical and Experimental Pharmacology and Physiology 39 (2012) 136-140. https://doi.org/10.1111/j.1440-1681.2011.05645.x

Z. Wang, P. Zhang, Y. Zhao, F. Yu, S. Wang, K. Liu, X. Cheng, J. Shi, Q. He, Y. Xia, L. Cheng. Scutellarin Protects Against Mitochondrial Reactive Oxygen Species-Dependent NLRP3 Inflammasome Activation to Attenuate Intervertebral Disc Degeneration. Frontiers in Bioengineering and Biotechnology 10 (2022) 883118. https://doi.org/10.3389/fbioe.2022.883118

H. Xu, S. Zhang. Scutellarin-induced apoptosis in HepG2 hepatocellular carcinoma cells via a STAT3 pathway. Phytotherapy Research 27 (2013) 1524-1528. https://doi.org/10.1002/ptr.4892

X. Shi, G. Chen, X. Liu, Y. Qiu, S. Yang, Y. Zhang, X. Fang, C. Zhang, X. Liu. Scutellarein inhibits cancer cell metastasis in vitro and attenuates the development of fibrosarcoma in vivo. International Journal of Molecular Medicine 35 (2015) 31-38. https://doi.org/10.3892/ijmm.2014.1997

C.Y. Li, Q. Wang, X. Wang, G. Li, S. Shen, X. Wei. Scutellarin inhibits the invasive potential of malignant melanoma cells through the suppression epithelial-mesenchymal transition and angiogenesis via the PI3K/Akt/mTOR signaling pathway. European Journal of Pharmacology 858 (2019) 172463. https://doi.org/10.1016/j.ejphar.2019.172463

L. Shi, Y. Wu, D. liang LV, L. Feng. Scutellarein selectively targets multiple myeloma cells by increasing mitochondrial superoxide production and activating intrinsic apoptosis pathway. Biomedicine and Pharmacotherapy 109 (2019) 2109-2118. https://doi.org/10.1016/j.biopha.2018.09.024

X.P. Sun, L.L. Wan, Q.J. Yang, Y. Huo, Y.L. Han, C. Guo. Scutellarin protects against doxorubicin-induced acute cardiotoxicity and regulates its accumulation in the heart. Archives of Pharmacal Research 40 (2017) 875-883. https://doi.org/10.1007/s12272-017-0907-0

L.L. Lin, A.J. Liu, J.G. Liu, X.H. Yu, L.P. Qin, D.F. Su. Protective effects of scutellarin and breviscapine on brain and heart ischemia in rats. Journal of Cardiovascular Pharmacology 50 (2007) 327-332. https://doi.org/10.1097/fjc.0b013e3180cbd0e7

L. Dai, L. Gu, K. Maeda. Inhibitory effect and mechanism of scutellarin on melanogenesis. Cosmetics 8 (2021) 15. https://doi.org/10.3390/cosmetics8010015

Q. Chen, C. Shang, M. Han, C. Chen, W. Tang, W. Liu. Inhibitory mechanism of scutellarin on tyrosinase by kinetics, spectroscopy and molecular simulation. Spectrochimica Acta - Part A: Molecular and Biomolecular Spectroscopy 296 (2023) 122644. https://doi.org/10.1016/j.saa.2023.122644

K. Patel, D.K. Patel. Medicinal importance, pharmacological activities, and analytical aspects of hispidulin: A concise report. Journal of Traditional and Complementary Medicine 7 (2017) 360-366. https://doi.org/10.1016/j.jtcme.2016.11.003

M. Han, H. Gao, P. Ju, M. quan Gao, Y. ping Yuan, X. hong Chen, K. li Liu, Y. tao Han, Z. wu Han. Hispidulin inhibits hepatocellular carcinoma growth and metastasis through AMPK and ERK signaling mediated activation of PPARγ. Biomedicine and Pharmacotherapy 103 (2018) 272-283. https://doi.org/10.1016/j.biopha.2018.04.014

D.E. Kim, K. jin Min, M.J. Kim, S.H. Kim, T.K. Kwon. Hispidulin inhibits mast cell-mediated allergic inflammation through down-regulation of histamine release and inflammatory cytokines. Molecules 24 (2019) 2131. 10.3390/molecules24112131.

Y.C. Lin, C.M. Hung, J.C. Tsai, J.C. Lee, Y.L.S. Chen, C.W. Wei, J.Y. Kao, T. Der Way. Hispidulin potently inhibits human glioblastoma multiforme cells through activation of AMP-activated protein kinase (AMPK). Journal of Agricultural and Food Chemistry 58 (2010) 9511-9517. https://doi.org/10.1021/jf1019533

J.M. Yang, C.M. Hung, C.N. Fu, J.C. Lee, C.H. Huang, M.H. Yang, C.L. Lin, J.Y. Kao, T. Der Way. Hispidulin sensitizes human ovarian cancer cells to TRAIL-induced apoptosis by AMPK activation leading to Mcl-1 block in translation. Journal of Agricultural and Food Chemistry 58 (2010) 10020-10026. https://doi.org/10.1021/jf102304g

L. He, Y. Wu, L. Lin, J. Wang, Y. Wu, Y. Chen, Z. Yi, M. Liu, X. Pang. Hispidulin, a small flavonoid molecule, suppresses the angiogenesis and growth of human pancreatic cancer by targeting vascular endothelial growth factor receptor 2-mediated PI3K/Akt/mTOR signaling pathway. Cancer Science 102 (2011) 219-225. https://doi.org/10.1111/j.1349-7006.2010.01778.x

H. Gao, H. Wang, J. Peng. Hispidulin Induces Apoptosis Through Mitochondrial Dysfunction and Inhibition of P13k/Akt Signalling Pathway in HepG2 Cancer Cells. Cell Biochemistry and Biophysics 69 (2014) 27-34. https://doi.org/10.1007/s12013-013-9762-x

A. Mouri, H.J. Lee, T. Mamiya, Y. Aoyama, Y. Matsumoto, H. Kubota, W.J. Huang, L.C. Chiou, T. Nabeshima. Hispidulin attenuates the social withdrawal in isolated disrupted-in-schizophrenia-1 mutant and chronic phencyclidine-treated mice. British Journal of Pharmacology 177 (2020) 3210-3224. https://doi.org/10.1111/bph.15043

D. Kavvadias, P. Sand, K.

留言 (0)

沒有登入
gif