Next Article in Journal / Special Issue
Persistent Nasal Obstruction: An Expression of the Pro-Inflammatory State?
Previous Article in Journal
The “Scrubbing Brush Technique” for Access to Tight Lateral Recess of the Sphenoid Sinus: A Single Case Report
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on the Medicinal and Pharmacological Properties of Traditional Ethnomedicinal Plant Sonapatha, Oroxylum indicum

by
Ganesh Chandra Jagetia
1,2
1
Department of Zoology, Mizoram University, Aizawl 796004, India
2
10 Maharana Pratap Colony, Sector 13, Hiran Magri, Udaipur 313002, India
Sinusitis 2021, 5(1), 71-89; https://doi.org/10.3390/sinusitis5010009
Submission received: 18 November 2020 / Revised: 3 May 2021 / Accepted: 20 May 2021 / Published: 25 May 2021
(This article belongs to the Special Issue Feature Papers in Sinusitis 2021)

Abstract

:
Oroxylum indicum, Sonapatha is traditionally used to treat asthma, biliousness, bronchitis, diarrhea, dysentery, fevers, vomiting, inflammation, leukoderma, skin diseases, rheumatoid arthritis, wound injury, and deworm intestine. This review has been written by collecting the relevant information from published material on various ethnomedicinal and pharmacological aspects of Sonapatha by making an internet, PubMed, SciFinder, Science direct, and Google Scholar search. Various experimental studies have shown that Sonapatha scavenges different free radicals and possesses alkaloids, flavonoids, cardio glycosides, tannins, sterols, phenols, saponins, and other phytochemicals. Numerous active principles including oroxylin A, chrysin, scutellarin, baicalein, and many more have been isolated from the different parts of Sonapatha. Sonapatha acts against microbial infection, cancer, hepatic, gastrointestinal, cardiac, and diabetic disorders. It is useful in the treatment of obesity and wound healing in in vitro and in vivo preclinical models. Sonapatha elevates glutathione, glutathione-s-transferase, glutathione peroxidase, catalase, and superoxide dismutase levels and reduces aspartate transaminase alanine aminotransaminase, alkaline phosphatase, lactate dehydrogenase, and lipid peroxidation levels in various tissues. Sonapatha activates the expression of p53, pRb, Fas, FasL, IL-12, and caspases and inhibited nuclear factor kappa (NF-κB), cyclooxygenase (COX-2), tumor necrosis factor (TNFα), interleukin (IL6), P38 activated mitogen-activated protein kinases (MAPK), fatty acid synthetase (FAS), sterol regulatory element-binding proteins 1c (SREBP-1c), proliferator-activated receptor γ2 (PPARγ2), glucose transporter (GLUT4), leptin, and HPV18 oncoproteins E6 and E7 at the molecular level, which may be responsible for its medicinal properties. The phytoconstituents of Sonapatha including oroxylin A, chrysin, and baicalein inhibit the replication of SARS-CoV-2 (COVID-19) in in vitro and in vivo experimental models, indicating its potential to contain COVID-19 infection in humans. The experimental studies in various preclinical models validate the use of Sonapatha in ethnomedicine and Ayurveda.

1. Introduction

Oroxylum indicum belongs to the family Bignoniaceae and is also known as Arthrophyllum reticulatum Blume ex Miq., Bignonia lugubris Salisb., Bignonia indica L., Bignonia tripinnata Noronha, Bignonia pentandra Lour., Bignonia quadripinnata Blanco, Bignonia tuberculate Roxb. ex DC., Hippoxylon indica (L.) Raf., Calosanthes indica (L.) Blume, Oroxylum flavum Rehder, and Spathodea indica (L.) Pers. Oroxylum indicum is commonly known as the tree of Damocles in English and Sonapatha or Shyonak in Hindi. Sonapatha is a semi-deciduous tree, which is sparingly branched and grows up to a height of 18–20 m. The trunk of Sonapatha is around 40 cm in diameter, and the stem bark is light brown or greyish-brown colored, which is soft and spongy with numerous corky lenticels. The inner bark layer of Sonapatha is golden-yellow colored and, hence, the name Sonapatha [1,2,3]. The leaves of Sonapatha are crowded, 0.5–2 m long, 3–4 times pinnate with long petiole, and swollen rachis at the points of insertion (Figure 1). The stipules are absent, whereas leaflets are ovate to oblong. The leaflets are 4–15 cm × 3–9 cm in size with a cuneate base and scattered glands on the lower surface. Inflorescence of Sonapatha is an erect raceme, terminal, and 25–150 cm long, with partitioned peduncle rachis (Figure 1). Flowers are bisexual, containing 2–4 cm long pedicel, bracteolate, coriaceous calyx, and campanulate. The buds are 2–4 cm long, 1.5–2 cm in diameter, brown or dirty violet colored, containing water, and become almost woody in fruit (Figure 1). The corolla is funnel shaped, about 10 cm long with 5 lobes, subequal with wrinkled margin, reddish outside, and yellowish to pinkish colored inside. The flowers contain five stamens, inserted in the throat, hairy at the base. The ovary is superior, two-celled, and contains many ovules (Figure 1).

2. Vernacular Names

The different names of Sonapatha are listed in Table 1. It is commonly known as midnight horror, Indian trumpet flower, broken bones plant, Tree of Damocles, Indian calosanthes, Indian caper, and Indian trumpet in English; Shyonak, Sonapatha, Patrorna, Shallaka, Putivriksha, Shuran, Sauna, Son, Aralu, Vatuk, Urru, Bhut-Vriksha, Dirghavrinta, Kutannat, or Manduk (the flower) in Hindi; Shyonaka, Nat, Katvanga, Sukanasa, Prithu Simba, Tuntuka, Dirghavrinta, Mandukparana, Kutannata, Patroma, Katammar, Bhalluka, Mayurjangha, and Aralu in Sanskrit; Tentu in Gujarati; Ane-mungu, Alangi, Patagani, Bunepaale, Tattuna, Sonepatta, Tigadu, and Tattuna in Kannada; Dundilamu, Manduka-paramu, Chettu, Nemali, Pampena, Pampini, or Suka-nasamu in Telugu; Khonha, Sona, Kani-Dingi, Paharijora, Hanghoal, Thona Gach, Aklong-Singh, Krong-Sa-Bang (Marma Tribe), Naori Chilana (Chakma Tribe), Kaak-Rakung (Halam Tribe), Tou-Kharung (Tripura Tribe), or Kanai Dingi (Garo Tribe) in Bengali; Davamadak in Konkani; Achi, Arandei, Cori-Konnai, Venga maram, Palai-yutaicci, Pana, Puta-Puspam (the flower), Pei-maram, Peiarlanke, Palagaipayani, Peruvaagai or Vangam in Tamil; Tetu or Tayitu in Marathi; Vashrppathiri, Palaqapayyani, or Vellapathiri, in Malayalam; Bhatghila, Dingdinga or Toguna in Assamese; Archangkawm, Phanphania, or Tatelo in Oriya; Archangkawm in Mizo. Handy pinyin or Mud huddle in Chinese; Tatelo in Nepalese; Thotila or Totila in Singhalese; Bonglai Kulai or beka in Malay [1,2,3,4,5,6]. It is known as Pongporang (Sundanese), Kayu lanang, or Mungli (Javanese) in Indonesia; pingka-pingkahan (Tagalog), abong-abong (Bisaya), or Kamkampilan (Iloko) in Philippines; Pi ka in Cambodia; Lin may or Ung ka in Laos; Phe kaa (central), Litmai (northern), or Lin faa (north-eastern) in Thailand, and Núc nác, Hoàng bá nam, or Mộc hồ diệp in Vietnam [6,7,8].

3. Geographical Distribution

Sonapatha is distributed throughout the Indian subcontinent except drier regions and in the foothills of Himalaya up to a height of 1000–1200 m including Bhutan and Nepal. It is also widely distributed in Eastern and the Western Ghats as well as the North-East region of India [9]. In China, it grows in Fujian, Guangdong, Guangxi, Guizhou, Sichuan, and Yunnan provinces. Sonapatha is also found in Taiwan, Cambodia, Malaysia, Myanmar, Philippines, Thailand, Indonesia (Java, Sumatra), Sri Lanka (Ceylon), Vietnam, and Laos [4,8,10].

4. Traditional Medicinal and Ethnomedicinal Uses

Natural products have played a crucial role in the healthcare of humans since their evolution and continue to play an important role even in the most modern state-of-the-art therapeutic era. The sword-like fruits or twigs of Sonapatha have been used by the farmers to kill crabs in the paddy fields. Its flowers and fruits are served as a vegetable in Thailand. It is also used as wood, tannins, and dye. Sonapatha is grown as an ornamental plant because of its strange looks [11]. The stem bark of Sonapatha is employed to kill maggots on cattle wounds [11,12,13,14]. Sonapatha has been used to treat several human ailments since ancient times. Various parts of Sonapatha are used for human healthcare traditionally and in Ayurveda, where it is one of the most important ingredients of Brahma Rasayana, Dashmularishta, Dhanawantara Ghrita, Amritarishta, Narayana Taila, Dantyadyarista, and Chyavanprasha [11,15,16]. Ayurveda classifies Sonapatha as shothara (anti-inflammatory); sheetha prashamana (subsides cough and cold), and anuvasanogapa (used as oil enema). Ayurveda describes Sonapatha as astringent, bitter, aphrodisiac, tonic, cooling, light to digest, dry, pungent, hot in potency, and an appetite booster. Sonapatha is given to treat “vata”, asthma, bronchitis, fevers, diarrhea, dysentery, vomiting, biliousness, inflammation, leukoderma, skin diseases, rheumatoid arthritis, and to heal wounds. It is also given to deworm the intestine and to soothe anal troubles. Roots of Sonapatha act as an aphrodisiac, antidiabetic, astringent, antiarthritic, carminative, diaphoretic, constipating, anthelmintic, febrifuges, diuretic, digestive, and expectorant [1,3,11,15]. The root bark paste of Sonapatha prepared in sesame oil (Sesamum indicum) is a digestive tonic, whereas seeds are given orally to treat throat complications and hypertension [17]. The root bark of Sonapatha is given to treat nasopharyngeal cancer, stomatitis, and tuberculosis [18,19]. Sonapatha root bark paste is orally administered to treat dysentery, diarrhea jaundice, and rheumatism by the Naga tribe in India [20]. The medicated oil prepared from Sonapatha relieves pain in the otis when instilled in the ear [5]. Sonapatha seeds are digestive and purgative, and their paste is topically applied to treat boils and wounds. The powdered seeds are given to induce conception in women. The paste made from grounded seeds with fire soot is applied to the neck to treat tonsillitis. The poultice made from the bark of Sonapatha is used in the treatment of inflammations, skin diseases, sprains, and rheumatism, whereas powdered bark and paste are applied on burns and wounds, respectively [13,14,21]. Sonapatha stem bark is also used to treat stomach disorders [22]. A glass of bark decoction with two spoons of sugar is often given to cure jaundice by the halam tribe of northeast India [23]. The leaves are used in the snakebite and to treat ulcers, headache, and enlarged spleen [14,19,24]. Sonapatha fruits are sweet in taste, acrid, anthelmintic, stomachic, and expectorant and are used to improve appetite, treat throat infection, bronchitis, heart disorders, leukoderma, and piles [15,19,24,25,26]. The seeds and bark of Sonapatha are given in conditions of pneumonia, respiratory troubles, and fever by different tribes in India [27,28].
The root decoction of Sonapatha is given in the conditions of dysentery and diarrhea in Nepal [29]. In Malaysia, Sonapatha is given to relieve toothache, treat cholera, dysentery, fever, gastralia, wound injury, splenomegaly, and loss of appetite. The decoction prepared from any part of Sonapatha is used externally during childbirth. The fresh bark of Sonapatha is macerated and applied topically in allergic dermatitis. In the Philippines, the root bark is used as a diaphoretic [30]. In Vietnam, seed decoction is used as a folk medicine to cure bronchitis, cough, and gastritis. In Chinese medicine, the seeds of Sonapatha are given to treat ulcers, boils, liver, and stomach problems. Sonapatha bark is used in the treatment of rheumatism and dysentery in Myanmar, the Philippines, and Vietnam [31].

5. Phytochemistry

Table 2 lists various phytochemicals that have been detected in Sonapatha. The chloroform and ethanol stem bark extracts of Sonapatha contained phenolics and flavonoids, whereas saponins in the aqueous extract. The alkaloids were detected in benzene, chloroform, and ethanol extracts [32]. The ethyl acetate fractions of Sonapatha stem and root bark extracted in methanol and water (4:1) showed the presence of glycosides, terpenoids phenols, and alkaloids [33]. The seeds of Sonapatha extracted in acetone, benzene, chloroform, methanol, and water showed the presence of alkaloids, flavonoids, tannins, glycosides, sterols, phenols, saponins, fats, and oils, whereas alkaloids, glycosides, flavonoids, lignins, and quinines were absent in the chloroform seed extract. Acetone, benzene, chloroform, and methanol extracts of Sonapatha stem bark showed the presence of tannins, saponins, sterols, phenols, fats, and oils, whereas glycosides and quinones were absent in all extracts of stem bark. The methanol extract of the bark was weakly positive for alkaloids. The root bark of Sonapatha extracted in acetone, benzene, chloroform, methanol, and water showed the presence of flavonoids, alkaloids, glycosides, tannins, sterols, phenols, lignins, saponins, fats, and oils; however, quinones were absent [34]. The alkaloids, flavonoids, tannins, and glycosides were detected in the 95% ethanol extract of Sonapatha [35]. Chloroform and ethanol extracts of Sonapatha stem bark showed the presence of alkaloids. The flavonoids and tannins were detected in the chloroform, ethanol, and aqueous extracts, whereas cardiac glycosides were detected only in chloroform and ethanol extracts. The saponins were present only in ethanol and aqueous extracts of Sonapatha and the steroids were absent in the latter. The phlobatannins were altogether absent in Sonapatha stem bark [13]. The methanol extract of tender leaves of Sonapatha has been reported to possess phlobatannins, flavonoids, phenols, tannins, and glycosides [36].

5.1. Isolation of Active Chemicals

Several chemicals have been isolated from different parts of Sonapatha (Table 3).

5.1.1. Leaf

The leaves of Sonapatha contained baicalein; baicalein-7-O-glucoside; baicalein-7-O-diglucoside; baicalein-7-O-glucuronide; chrysin; chrysin-diglucoside; chrysin-7-O-glucuronide; oroxylin-A; scutellarin; baicalein-6-0-glucuronide [37]. The leaf oil of Sonapatha showed the presence of Ar-tumerone, methyl hexadecanoate, aurenan-2-one, and isopropyl butanoate [38].

5.1.2. Stem

Oroxylin A, chrysin, scutellarin, baicalein, baicalein-6-O-glucuronide, dihydrobaicalein, ellagic acid, dihydrooroxylin-A, methyl-3,4,5-trihydroxy-6-(5-hydroxy-6-methoxy-4-oxo-2-phenyl-chroman-7-yloxy)-tetrahydro-2H-pyran-2-carboxylate, 5-hydroxyl-7-methoxy-2-(2-methoxy-6-(3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yoloxy)phenyl)-4H-chromen-4-one, or-oxyloside, hispidulin, apigenin, ficusal, balanophonin, 2-(1-hydroxymethylethyl)-4H, 9H-naphtho [2,3-β]furan-4,9-dione, salicylic acid, ρ -hydroxybenzoic acid, protocatechuic acid, isovanillin, β-hydroxypropiovanillon, oroxylin, pinostrobin, stigmast-7-en-3-ol, and 3,4′,5,7 tetra-hydroxy flavonol (kaempferol), dehydro-iso-α-lapachone, and lapachol were isolated from the stem of Sonapatha [6,14,39,40,41,42]. Additionally, 5,6,7-trimethoxy flavone-8-O-β-d-glucopyranoside, oroxylin A-7-Odglucuronide butylester, 6-methoxy-baicalein, oroxylin-A-7-O-glucoside, 5,7-dihydroxy–flavone, baicalein, and 6-methoxy-7-glucuronide have also been separated from the stem of Sonapatha [6,14,39,40,41,42,43].

5.1.3. Roots

The roots of Sonapatha have been reported to contain ellagic acid, baicalein, chrysin, biochanin-A, oroxylin A, hispidulin, 2,5-dihydroxy-6,7-dimethoxy flavone and 3,7, 3′,5′-tetramethoxy-4-hydroxy flavone, lapachol, and β-sitosterol [6,44,45,46].

5.1.4. Seeds

Several phytochemicals have been isolated from seeds of Sonapatha including oroxylin-A, scutellarin, baicalein, oroxindin, baicalein-6-glucoside (tetuin), oroxin A, oroxin B, chrysin, baicalin, quercetin, apigenin, kaempferol, quercetin-3-O-ara-binopyranoside, lupeol, lup-20 (29)-ene-2α,3β-diol, pinosylvin, dihydropinosylvin, cholest-5-ene-3, 7-diol, rengyol, isorengyol, zarzissine, (E)-pinosylvin-3-Odglucopyranoside, adenosine, sitosterol, daucosterol, chrysin 6-C-βdglucopyranosyl-8-Odglucuronopyranoside, baicalein 7-Odglucuronopyranosyl-(1→3)[βdglucopyranosyl(1→6)]βdglucopyranoside, scutellarin 7-Odglucopyranosyl-(1-6)-βdglucopyranoside, chrysin-7-O-gentiobioside, baicalein- 7-O-diglucoside, baicalein-7-O-glucoside, scutellarin-7-O-glucopyranoside, 2-methyl-6-phenyl 4H-pyran-4-one, chrysin-7-O-glucuronide, chrysin-6-C-βddimethyl sulfone, glucopyranosyl-8-C-α-L-arabinopyranoside, pinocembrin, pinoban- ksin, lupeol, 2α-hydroxyllupeol, echinulin, β-sitosterol, baicalein-7-O-gentiobioside, baicalein-7-O-glucoside, baicalein-7-O-glucuronide, oroxin C, and oroxin D [47,48,49,50,51,52,53,54].

5.1.5. Flowers and Fruits

Baicalein and chrysin have been isolated from the flowers of Sonapatha [55], whereas the fruits have been shown to possess oroxylin A, orlumin A, chrysin, and ursolic acid [56,57].

6. Pharmacological Activities

Sonapatha has shown various pharmacological activities in different study systems in vitro and in vivo as listed below.

6.1. Free Radical Scavenging and Antioxidant Activities

Table 4 lists the free radical scavenging ability of various extracts of Sonapatha. The estimation of antioxidant activity by reduction in Mo+4 to Mo+5 showed that petroleum ether, benzene, chloroform, ethanol, and aqueous stem bark extracts of Sonapatha dose dependently increased the antioxidant activity, and the highest antioxidant potential was detected for the chloroform followed by ethanol extract, whereas the petroleum ether extract was the least active [32]. The methanol extract of different parts of Sonapatha also scavenged hydroxyl, nitric oxide, superoxide, and DPPH free radicals in vitro [58]. The leaves of Sonapatha extracted in petroleum ether, chloroform, ethanol, and water inhibited the formation of DPPH, hydroxyl, nitric oxide, and superoxide radicals concentration dependently, where the ethanol extract was most potent. The reducing power assay further confirmed that the ethanol leaf extract of Sonapatha was more powerful than the other extracts [59]. Similarly, leaves of Sonapatha extracted in ethyl acetate, methanol, and water scavenged DPPH radical depending on their concentration, and the ethyl acetate extract was most potent followed by methanol and water extracts [60]. The hexane, chloroform, ethyl acetate, and water extracts of stem bark of Sonapatha showed a concentration-dependent inhibition of DPPH radicals, and the ethyl acetate extract was more effective than the other three extracts [61]. The petroleum ether, dichloromethane, and methanol extracts of Sonapatha have been found to scavenge DPPH, and superoxide radicals and possessed Fe3+ reducing potential. The methanol extract was more potent than the other two extracts [62]. The radical scavenging activity of chloroform, ethanol, and aqueous extracts of the stem bark of Sonapatha showed that it inhibited DPPH, superoxide anion, hydroxyl, and nitric oxide free radicals in a concentration-dependent manner, and 200 μg/mL of chloroform and ethanol extracts scavenged the DPPH radicals to the maximum extent. This concentration for hydroxyl radical inhibition was 6000 and 8000 μg/mL for chloroform and ethanol extracts, respectively. The highest scavenging of superoxide anion radical was observed at a concentration of 2500 μg/mL for both chloroform and ethanol extracts, whereas it took 20,000 μg/mL to inhibit the nitric oxide radical to the maximum extent. The ferric reducing power of different Sonapatha stem bark extracts increased depending on their concentration and a dose of 2500 μg/mL of all three extracts resulted in the maximum reduction in ferric iron [63]. The ethanol and aqueous extracts of Sonapatha seeds have been reported to inhibit DPPH radicals, whereas the fruit extracts were less effective than the seed extracts [64].

6.2. Anti-Inflammatory and Analgesic Activities

The 50 μg/mL of dichloromethane root extracts of Sonapatha resulted in 100% inhibition of leukocyte lipoxygenase activity indicating its anti-inflammatory potential [6]. The aqueous leaf extract of Sonapatha has shown anti-inflammatory activity in carrageenan-induced rat paw edema test where 300 mg/kg body weight (b. wt.) extract was found to be superior over 150 mg/kg b. wt. [65]. A similar effect has been reported for the Sonapatha root and stem bark decoction in water that reduced the carrageenan-induced rat paw edema [66]. The rats treated with aqueous root extract of Sonapatha at a dose of 100, 200 and 400 mg/kg b. wt. for seven or four days significantly inhibited the induction of dinitrobenzene sulfonic acid-induced colitis [67]. The oral administration of 500 mg/kg b. wt. reduced acetic acid writhing, which was comparable to standard analgesic agent aminopyrine given at a dose of 50 mg/kg [68]. The aqueous extract of Sonapatha has been reported to reduce mouse ear edema in an earlier study [69]. The ethanol extract of Sonapatha (250 and 300 mg/kg b. wt.) was evaluated for its anti-inflammatory and analgesic activities and 300 mg/kg ethanol extract reduced mice ear and paw edema and also exerted analgesic effect to the maximum extent [70]. The hydroalcoholic extract of Sonapatha stem bark extract exerted anti-inflammatory activity on carrageenan-induced rat paw edema [71]. The Sonapatha extracted in methanol and given at a dose of 100, 200, and 400 mg/kg b. wt. has been reported to reduce carrageenan-induced rat paw edema depending on its dose. The histological evaluation of rat paw showed reduced cell infiltration and suppressed inflammation in a dose-dependent manner [72].

6.3. Anti-Allergic and Antiasthmatic Effect

Allergic rhinitis and asthma are linked and affect 40% of the world population. They reduce the quality of life and performance and their prevalence has been increasing in the population throughout the world [73]. The studies on the antiallergic and antiasthmatic effect of Oroxylum indicum are scanty. The antiallergic and anti-asthmatic effects of oroxylin A extracted from Sonapatha have been studied in female Balb/c mice and cultured rat RBL-2H3 mast cells. The rat RBL-2H3 mast cells sensitized with monoclonal anti-dinitrophenyl specific mouse IgE and then exposed to 0.1, 0.3, 1, 3, 10, and 30 µM oroxylin A reduced the antigen-induced degranulation in a concentration-dependent manner as indicated by the release of β-hexosaminidase. The maximum degranulation and β-hexosaminidase release were estimated at a concentration of 30 µM oroxylin A in RBL-2H3 mast cells. The female Balb/c mice were injected with 1 and 5 mg/ kg b. wt. oroxylin A intraperitoneally 30 before OVA administration to induce asthma inhibited the accumulation of eosinophils in bronchoalveolar lavage fluid of mice by 51% and 84%, respectively. The histologic observations of mice lungs revealed the accumulation of eosinophils around bronchioles whereas oroxylin A administration alleviated the eosinophils around bronchioles depending on its dose. The oroxylin A administration also reduced the OVA-induced mucin production in the mice lungs. The study on various cytokine expression revealed that oroxylin A suppressed the expression of IL-4, IL-5, and IL-13, INF-γ and IL-2 in the lungs of OVA administered mice, which may be responsible for its antiallergic and antiasthmatic activities in Balb/c mice [74].

6.4. Antimicrobial Activity

The antimicrobial activity of the dichloromethane extract of Sonapatha was studied against the Gram-positive and Gram-negative bacteria, and it was found to inhibit the growth of both the Gram-positive Bacillus subtilis and Staphylococcus aureus and the Gram-negative Escherichia coli and Pseudomonas aeruginosa bacteria and the fungi Candida albicans [6]. The dichloromethane extract of Sonapatha also inhibited the growth of dermatophytes and wood root fungi [75]. The root and stem of Sonapatha extracted in alcohol suppressed the growth of Klebsiella, Escherichia coli, Proteus, Pseudomonas, and Staphylococcus aureus, and the stem extract was found to be superior to the root extract [33]. The methanol extract of stem bark of Sonapatha has been reported to be active against Bacillus cereus, Bacillus megaterium, Bacillus subtilis, Staphylococcus aureus, Sarcina lutea (Gram-positive), Escherichia coli, Pseudomonas aeruginosa, Salmonella paratyphi, Salmonella typhi, Shigella boydii, Shigella dysenteriae, Vibrio mimicus, Vibrio parahemolyticus (Gram-negative), and the fungi Saccharomyces cerevisiae, Candida albicans and Aspergillus niger [42].
The petroleum, ether, dichloromethane, and methanol extracts of stem bark of Sonapatha were active against the Klebsiella pneumoniae, Pseudomonas aeruginosa, Escherichia coli, Bacillus subtilis, Bacillus cereus, and Staphylococcus aureus bacteria and Aspergillus flavus, Aspergillus fumigatus, and Macrofomina phaeolina fungi [62]. The hydroalcoholic extract of stem bark of Sonapatha reduced the growth of Bacillus cereus, Bacillus subtilis, Staphylococcus aureus, and Staphylococcus albus bacteria [76]. The aqueous extracts of Sonapatha seeds and fruits have been reported to inhibit the growth of Staphylococcus intermedius and Streptococcus suis bacteria in a concentration-dependent manner and the aqueous extract was less effective. The seed extracts were more effective than the fruit extracts [36]. The stem bark of Sonapatha extracted in methanol, ethyl acetate and ethanol inhibited the growth of Bacillus subtilis, Escherichia coli (Gram-positive), and Pseudomonas aeruginosa (Gram negative), and the ethanol extract was found to be most potent [77]. The hexane, chloroform, ethyl acetate, and water extracts of stem bark of Sonapatha have shown antimicrobial activity against Escherichia coli, Bacillus subtilis, Pseudomonas aeruginosa, Staphylococcus aureus, and Candida albicans. The chloroform and ethyl acetate extracts were most effective than the hexane and water extracts [61]. The tender leaves of Sonapatha extracted in methanol inhibited the growth of Pseudomonas aeruginosa and Bacillus subtilis studied by zone inhibition and well methods, respectively [36]. The orlumin A isolated from the fruits of Sonapatha has shown the highest antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA) with a MIC value of 3.1 μg/mL [57].

6.5. Anthelmintic Effect

The methanol extract of Sonapatha stem bark has been tested against the anthelmintic activity in Hymenolepis diminuta. The artificially excysted juveniles and adult H. diminuta worms were treated with 10, 20, and 30 mg/mL of stem bark Sonapatha decoction. The exposure of juvenile worms to 30 mg/mL of Sonapatha bark extract resulted in worm mortality as early as 0.25 ± 0.00 h after treatment. The scanning electron microscopic examination revealed that the extract treatment led to wrinkled scolex, distorted integument, and eroded microtriches in both the juvenile and adult worms [78]. The in vivo study in H. diminuta infected Wistar rats revealed that administration of 1000 mg/kg b. wt. Sonapatha extract caused 79.3% reduction in eggs per gram of feces count and 70.8% reduction in worm counts indicating that it was more effective in killing juvenile worms than the adult worms [78]. A study of methanol extract of Sonapatha stem bark and aqueous fruit extracts was conducted against the killing of adult Pheretima posthuma earthworm. The exposure of P. posthuma to 10, 50, and 100 mg/mL of stem bark and fruit extracts of Sonapatha resulted in a concentration-dependent paralysis and death of earthworms depending on the concentration [79].

6.6. Hepatoprotective Effect

The hepatoprotective effect of petroleum ether, chloroform, ethanol, and aqueous extracts of Sonapatha leaves was studied against the carbon tetrachloride-induced liver toxicity. All four extracts protected the liver against the carbon tetrachloride-induced toxicity as indicated by a decline in the rat serum aspartate transaminase (AST) and alanine aminotransaminase (ALT) alkaline phosphatase (ALP) and total bilirubin levels. However, ethanol extract was found to be most effective than the other three extracts [59]. The administration of methanol stem bark extract of Sonapatha at a dose of 500 mg/kg b. wt. protected rat liver against the carbon tetrachloride-induced toxicity by reducing the serum AST, ALT, ALP, and total bilirubin levels. It also reduced carbon tetrachloride-induced rat liver damage and necrosis [80]. The hepatoprotective action of 100, 200, and 400 mg/kg b. wt. of aqueous root bark extract of Sonapatha was studied against the paracetamol-induced liver toxicity in Wistar rats, where the aqueous extract was found to reduce serum AST, ALT, and ALP levels in a dose-related manner, and their lowest activities were reported for 400 mg/kg extract. This was backed by histological examination where the aqueous Sonapatha root bark extract reduced the paracetamol-induced degenerative changes in the liver of Wistar rats [81]. The mouse treated with petroleum ether, ethyl acetate, methanol, and ethanol extracts of Sonapatha stem bark protected the liver against carbon tetrachloride-induced liver toxicity. The ethyl acetate extract was superior to other extracts as it reduced the pathological damage to the liver to a maximum extent followed by the higher reduction in AST, ALT, ALP, and total bilirubin levels in mouse serum than the other extracts [23]. The methanol-dichloromethane extract of the whole Sonapatha plant has been reported to protect liver explant and chronic liver damage in rats as indicated by a significant depletion in AST, ALT, and ALP levels in both explant and rat serum. Besides, the serum bilirubin level declined significantly. The histology of the liver showed mild hepatic swelling and a reduction in the carbon tetrachloride-induced hepatic injury [82]. The administration of 10, 50, and 100 mg/kg b. wt. aqueous-methanol extract of stem bark of Sonapatha reduced the acetaminophen-induced liver toxicity in a dose-dependent manner, where the serum levels of AST, ALT, and lipid peroxidation (LOO) were significantly low in Sprague-Dawley rats [83]. The aqueous and ethanol extracts of Sonapatha protected Wistar rats against the antitubercular drug AKT-4 induced hepatotoxicity significantly by alleviating the AKT4 induced increase in ALT, AST, lactate dehydrogenase (LDH), and total bilirubin levels. Additionally, the aqueous and ethanol extracts of Sonapatha significantly increased catalase, superoxide dismutase (SOD), and glutathione (GSH) levels, whereas LOO level declined significantly in the AKT-4 treated rat liver. AKT4 induced pathological alterations in the hepatocytes of the rat with distorted hepatic cords, degenerative, and necrobiotic changes, accumulation of fat globules in the hepatocytes with their swelling and hemorrhage, whereas treatment of rats with aqueous and ethanol extracts of Sonapatha, showed no such histopathological changes in the liver [84]. The 70% ethanol extract of stem bark of Sonapatha reduced serum, ALT, ALP, γ-glutamyltransferase (GGT), and total bilirubin levels at different posttreatment days in the carbon tetrachloride treated Wistar rats [85]. The seed extract of Sonapatha has been reported to protect against the progression of nonalcoholic fatty liver disease into nonalcoholic steatohepatitis in vitro and in vivo by inhibiting the transcriptional activity of NF-κB and its phosphorylation and increasing the nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor (IκB) level [86].

6.7. Gastroprotective Effect

The gastroprotective activity of 100 and 300 mg/kg b. wt. of 50% hydroalcoholic, petroleum ether, chloroform, ethyl acetate, and n-butanol extracts of root bark of Sonapatha were studied against ethanol-induced gastric damage. All extracts at a dose of 300 mg/kg showed a significant reduction in the ulcer index. The petroleum ether, n-butanol, and chloroform extracts were most effective when compared to ethyl acetate and hydroalcoholic root extracts. Likewise, these extracts of Sonapatha also reduced LOO and increased the activities of SOD, and catalase, and GSH levels, especially at a dose of 300 mg/kg in Wistar rats [87]. Treatment of 100 and 250 mg/kg of hexane and acetone stem bark extracts of Sonapatha had a mild to moderate protective effect against different models of gastric ulcerations. The isolated chrysin and compound 1 from the stem bark of Sonapatha were superior gastroprotective agents in pylorus ligation and cold restrain-induced ulceritis models in rats [40]. The butanol, petroleum ether, and ethanol extracts of stem bark of Sonapatha reduced the ethanol-induced gastric ulcers with an inhibitory index of 0.07 ± 0.007 (99%), 0.27 ± 0.011 (96%), and 0.87 ± 0.044 (86%), respectively [72].

6.8. Cardioprotective Effect

The cardioprotective effect of 70% methanol extract of root bark of Sonapatha was studied in doxorubicin-treated Sprague Dawley rats. The oral administration of animals with 200 and 400 mg/kg b. wt. Sonapatha extract for 14 days protected rats against doxorubicin-induced cardiotoxicity. The methanol extract of Sonapatha led to normalization of Electrocardiogram (ECG), ST-segment depression, and QRS complex in the hearts of doxorubicin treated rats. The serum marker enzymes like creatinine phosphokinase (CPK), AST, and LDH were significantly reduced by methanol extract of Sonapatha. The assessment of LOO, GSH, glutathione peroxidase, (GPx), and SOD in the heart tissue revealed a significant decline in LOO and rise in the GSH, GPx, and SOD levels in the Sonapatha treated group when compared to doxorubicin treatment alone. Histopathologic evaluation revealed focal degeneration, fragmentation, disorganization of myofibrils, and necrotic changes in the heart tissue after doxorubicin treatment whereas Sonapatha extract administration reduced these changes [88].

6.9. Antidiabetic Effect

Administration of 300 and 500 mg/kg b. wt. of aqueous and ethanol extracts of Sonapatha for 21 days significantly reduced serum glucose levels in alloxan and dexamethasone-induced diabetes in Wistar rats [89]. Methanol extract of the heartwood of Sonapatha reduced the activity of α-glucosidase (GAA; an enzyme involved in carbohydrate digestion and glycoprotein biosynthesis, which is highly activated in diabetes) indicating its antidiabetic potential [41]. Similarly, hydroalcoholic extract (50% ethanol) of Sonapatha inhibited GAA activity in vitro. Sonapatha extract has been found to improve insulin sensitivity in cultured 3T3-L1 mature adipocytes. Oral administration of 250 mg/kg b. wt. of 50% ethanol extract of Sonapatha has reduced fasting blood glucose, low-density lipoprotein (LDL), and glycosylated hemoglobin (HbA1c) levels and elevated high-density lipoprotein (HDL) in streptozotocin-induced diabetic rat serum after 28 days post-treatment. Homeostasis model assessment-insulin resistance (HOMA-IR) index and quantitative insulin sensitivity check index (QUICKI) were also significantly reduced in Sonapatha treated diabetic rats after 28 days post-treatment [90]. The seeds of Sonapatha extracted in 90% ethanol inhibited the rat intestinal GAA activity. The administration of 50 and 250 mg/kg b. wt. of alcoholic seed extract of Sonapatha non-significantly reduced the fasting glucose level in alloxan-induced diabetic mice however, its combination with acarbose significantly reduced the fasting glucose level [91]. Similarly, 90% ethanol seed extract of Sonapatha at a dose of 50 and 200 mg/kg b. wt. in conjunction with acarbose inhibited glucose level in streptozotocin-induced diabetic mice significantly. The Sonapatha seed extract increased the antioxidative capacity by elevating GSH, SOD, and HDL followed by reduced LOO and LDL levels in prediabetic mice [92]. The administration of 300 mg/kg b. wt. of aqueous and methanol extracts of Sonapatha leaves reduced the glucose, total cholesterol, triglyceride, protein, urea, and creatinine levels in alloxan-induced diabetic rats [93]. Administration of 200 and 400 mg/kg b. wt. of 50% ethanol seed extract of Sonapatha for 21 days reduced serum glucose, triglyceride, and cholesterol levels in the glibenclamide-induced diabetic rats [94].

6.10. Antiobesic Effect

Obesity is one of the major health problems in humans and 13% of the adult world population is clinically obese. Obesity is associated with cardiovascular diseases, type II diabetes and certain cancers and Sonapatha has shown promise to reduce obesity preclinically. The 3T3-L1 adipocytes treated with hexanes, dichloromethane, ethyl acetate and methanol extracts of Sonapatha bark inhibited the lipid accumulation and lipase activity and the ethyl acetate extract was most effective [95]. Similarly, 3T3-L1 adipocytes exposed to 50, 100, 150 or 200 μg/mL of 95% Sonapatha ethanol fruit extract for 2 and 10 days suppressed the accumulation of lipids and lipase activity in 3T3-L1 adipocytes concentration-dependently. Sonapatha fruit extract also reduced lipids, lipid esters, nucleic acids, glycogen and carbohydrates in 3T3-L1 adipocytes [96]. The treatment of 3T3-L1 preadipocytes with 50, 100, 150 or 200 μg/mL fruit extract of Sonapatha inhibited lipid accumulation and the greatest effect was observed for 200 μg/mL. The study of mRNA expression showed that 200 μg/mL fruit extract suppressed the expression of fatty acid synthetase (FAS), sterol regulatory element-binding proteins 1c (SREBP-1c), proliferator-activated receptor-γ2 (PPARγ2), glucose transporter (GLUT4), and leptin in adipocytes indicating its potential as an antiobesity agent [97].

6.11. Anticancer Effect

The aqueous and methanol extracts of Sonapatha stem bark induced apoptosis in MDA-MB-435S (human breast carcinoma), Hep3B (human hepatic carcinoma), and PC-3 (human prostate cancer) cells [98]. The exposure of HeLa cells to 50, 100, 200, 250 and 500 petroleum ether, dichlromethane and methanol extracts of Sonapatha stem bark caused a concentration-dependent rise in the cytotoxic effect with an IC50 of 112.3 ± 4.4, 171.7 ± 7.4 and 315.7 ± 6.5 μg/mL for petroleum ether, dichlromethane and methanol extracts, respectively. The petroleum ether extract was most cytotoxic and induced apoptosis indicated by membrane blebbing, nuclear fragmentation and apoptotic bodies and DNA fragmentation [62]. The petroleum and chloroform stem bark extract of Sonapatha has been reported to exert dose-dependent cytotoxicity in MDA-MB-231 and MCF-7 breast cancer cells by XTT assay and also induced apoptosis indicated by DNA fragmentation measured by ELISA [99]. The Sonapatha leaves extracted in methanol have shown the cytotoxic effect on HeLa cells with an IC50 of 3.87 μg/mL, whereas it did not exert any cytotoxicity on normal Vero and MDCK cells. The methanol leaf extract of Sonapatha induced apoptosis, which was characterized by nuclear fragmentation, condensation, and apoptotic bodies. The apoptosis induction by methanol extract was further characterized by FITC-Annexin V and propidium iodide assay by flow cytometry analysis. The flow cytometric evaluation has revealed the collection of cells in the G1/S phase of the cell cycle by increasing p53 expression [100]. The cytotoxic effects of ethanol extract of wild Sonapatha and twigs cultured in vitro have triggered dose-dependent cytotoxicity in SiHa and HepG2 cells with IC50 of 440 and 480 µg mL−1, respectively for wild samples. However, the cytotoxicity was lower in the case of in vitro samples that exhibited the IC50 of 530 and 550 µg mL−1 for SiHa and HepG2 cells, respectively [101]. The root bark of Sonapatha extracted in chloroform, ethyl acetate and n-butanol showed lethality in the brine shrimp assay and it was also cytotoxic to MCF-7 cells [102]. The leaf, bark, pod, and seeds of Sonapatha extracted in ethanol showed a concentration-dependent increase in cytotoxicity by sulforhodamine B and clonogenic assays with the IC50 of 161.2 ± 8.63, 286.73 ± 33.01, 149.03 ± 8.81, and 107.06 ± 5.66 μg/mL for leaf, bark, pod, and seed extracts, respectively. The seed extract was most cytotoxic as indicated by colony-forming assay [103]. The leaves and fruit of Sonapatha extracted in ethanol were cytotoxic to MCF-7 cells in sulforhodamine B and clonogenic assays having an IC50 of 57.02 ± 2.85 µg/mL and 131.3 ± 19.2 µg/mL, respectively. The leaf extract also inhibited cell migration, activated caspase 3, and triggered apoptosis and suppressed the expression of MMP 9, ICAMP1, and Rac1 genes in MCF-7 cells. RhoA was significantly elevated by both leaf and fruit extracts in MCF-7 cells [104]. The methanol extract of Sonapatha leaves triggered cytotoxicity in a concentration-dependent manner with an IC50 of 6.25 ± 1.06 µg/mL. The cytotoxic effect of Sonapatha seems to be mediated by the downmodulation of HPV18 oncoproteins E6 and E7, followed by the activation of caspase-8 and caspase-3 and higher mRNA expressions of p53, pRb, Fas, and FasL in HeLa cells. The Sonapatha methanol leaf extract increased IL-12 and reduced the IL-6 production in HeLa cells [105].

6.12. Wound Healing Effect

The root bark of Sonapatha extracted in methanol has been reported to heal partial burn wounds in mice receiving the topical application of 1 and 2.5% extract containing ointment. This was evident by higher wound contraction and reduced wound healing time. The histological evaluation revealed a time-dependent progression in the formation of dermis and reepithelialization. The methanol extract of Sonapatha also increased collagen synthesis and 2.5% extract was superior to 1% [106]. Topical application of 5, 10, 20, and 30% ethanol stem bark extract of Sonapatha increased wound contraction and reduced the mean wound healing time in a dose-dependent manner of the full thickness excision wounds of mice. The 10% extract was most effective as it produced the highest wound contraction and reduced healing time maximally. The biochemical analysis of collagen and DNA syntheses showed a dose-dependent rise, accompanied by a decline in the LOO. The Western blot analysis showed that infliction of excision wound upregulated NF-κB and COX-2, whereas topical application of Sonapatha ethanol extract reduced their expression [14].

6.13. Effect against COVID-19 Infection

Coronavirus disease 2019 (COVID-19) is an infectious disease caused by coronavirus-2 (SARS-CoV-2). Covid-19 became a pandemic at the beginning of 2020 and since then several deaths have occurred worldwide. Covid-19 infection is characterized by a severe life-threatening acute respiratory syndrome caused by hyperinflammatory response, vascular damage, microangiopathy, angiogenesis and widespread thrombosis. Currently, no specific treatment is available against COVID19 infection [107]. The COVID-19 entry into the primary host-cell is mediated by specific binding of coronavirus spike proteins (s) to the angiotensin-converting enzyme II (ACE2) receptors of human organs. Therefore, blocking of ACE2 can reduce COVID-19 entry into the cell. The molecular docking and the surface plasmon resonance studies have shown that oroxylin A present in Sonapatha can suppress the entry of the SARS-CoV-2-spiked pseudotyped virus into ACE2 cells, as it binds well to the ACE2 (blocking its availability for COVID-19) receptors as shown by surface plasmon resonance and cell membrane chromatography [108]. The 200 mg/kg baicalein another constituent of Sonapatha has been found to inhibit the replication of the COVID-19 virus and loss of body weight and reduce lung damage in hACE2 transgenic mice infected with SARS-CoV-2 and it also reduced SARS-CoV-2-induced injury in cultured Vero cells [109]. Baicalein inhibited the replication of SARS-CoV-2 in vitro with an IC50 of 10 μM in Vero E6 cells by reducing oxygen consumption rate, oxidative phosphorylation and mitochondrial membrane potential [110]. Molecular docking studies against the SARS-CoV-2 virus replication enzyme main protease (Mpro) revealed that four (baicalein-7-O-diglucoside, chrysin-7-O-glucuronide, oroxindin and scutellarein) of the eighteen constituents of Sonapatha had the potential to inhibit the activity of this enzyme and restrain COVID-19 infection [111].

7. Mechanisms of Action

The actual mechanism of action of Sonapatha in mitigating various disorders is not well understood. Sonapatha synthesizes several primary and secondary metabolites, and they individually or collectively may interact with multiple targets in the cells to bring the desired action against numerous ailments with less adverse effect than its constituents (Figure 2). Reactive oxygen species (ROS) generation is a fundamental mechanism, which is produced during oxidative phosphorylation as well as defense against foreign invaders into the body. However, excess ROS production is linked to inflammation and most human diseases. Therefore, one of the important mechanisms of action of Sonapatha seems to be its ability to scavenge different free radicals and increase various antioxidants such as GSH, glutathione-S-transferase (GST), GPx, catalase, and SOD and alleviate LOO, ALT, AST, ALP, LDH, GAA and GGT in normal tissues. Different extracts of Sonapatha have been reported to arrest cells in the G1/S phase of the cell cycle, suppress the expression of NF-κB, COX-2, TNFα, IL2, IL3, IL5, IL13, IL12, IL4, IL6, INF-γ, P38MAPK, MMP 9, ICAMP1, Rac1, FAS, SREBP-1c, PPARγ2, GLUT4, leptin, ACE2, and HPV18 oncoproteins E6 and E7. In contrast, Sonapatha upregulates the expression of p53, Bax, pRb, Fas, FasL, IL-12, RhoA, and caspases leading to various beneficial effects of Sonapatha in various study systems discussed above [14,23,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,81,82,83,84,85,86,87,88,90,91,92,96,97,100,104,105,109,110].
ACE2: angiotensin-converting enzyme II; ALP: alkaline phosphatase; ALT: alanine aminotransaminase; AST: aspartate transaminase; COX: cyclooxygenase; CPK: creatinine phosphokinase; DF: DNA fragmentation; FasL: Fas ligand; FAS: fatty acid synthetase; FRS: free radical scavenging; GAA: α-glucosidase; GLUT4: glucose transporter; GPx: glutathione peroxidase; GSH: glutathione; GST: glutathione-s-transferase; HPV: human papilloma virus; IL: interleukin; IκB: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor; INF-γ: interferon-γ; LDH: lactate dehydrogenase; LOO: lipid peroxidation; MAPK: microtubule associated protein kinase; MMP: matrix metalloproteinase; NF-κB: nuclear factor kappa B; Rac: Ras-related C3 botulinum toxin substrate; Rb: retinoblastoma protein; Rho: Ras homolog family member; SOD: superoxide dismutase; TNF: tumor necrosis factor; sterol regulatory element-binding proteins 1c (SREBP-1c); PPARγ2: proliferator-activated receptor-γ 2.

8. Conclusions

Sonapatha is used in traditional and Ayurvedic systems of medicine to treat numerous human disorders. The scientific evaluation of Sonapatha has established its analgesic, anti-inflammatory, anthelmintic, antimicrobial, antidiabetic, anti-obesity, hepatoprotective, cardioprotective, gastroprotective, anticancer, and wound healing activities in various preclinical models in vitro and in vivo. The therapeutic potential of Sonapatha seems to be due to its ability to scavenge free radicals, elevate GSH, GST, GPx, catalase, and SOD levels, and reduce LOO, as most human ailments are triggered by increased oxidative stress. In addition, Sonapatha reduces lipid accumulation and lipase activity. At the molecular level, Sonapatha inhibits the activation of NF-κB, COX-2, TNFα, IL2, IL3, IL5, IL13, IL12, IL4, IL6, INF-γ, P38 MAPK, MMP 9, ICAMP1, Rac1, HPV18 oncoproteins E6 and E7, FAS, SREBP-1c, PPARγc2, GLUT4, and leptin. Sonapatha upregulates the activation of p53, pRb, Fas, FasL, IL-12, RhoA, and caspases 8/3, which may be related to its cytotoxic, anti-inflammatory, antidiabetic, antiobesity, hepatoprotective, gastroprotective, cardioprotective, and wound healing activities. The experimental and molecular docking studies have revealed that active constituents of Sonapatha including oroxylin A, baicalein, and chrysin have the potential to act against the COVID-19 infection by blocking the ACE2 receptors and attenuating the activity of enzyme main protease, which aid in the replication of SARS-CoV-2 in humans.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Acknowledgments

I wish to acknowledge the constant support, encouragement, and patience of my wife Mangla Jagetia during the writing of this manuscript.

Conflicts of Interest

The author does not have any conflict-of-interest statement to declare.

Abbreviations

ACE2: angiotensin-converting enzyme II; ALP: alkaline phosphatase; ALT: alanine aminotransaminase; AST: aspartate transaminase; COX: cyclooxygenase; CPK: creatinine phosphokinase; FasL: Fas ligand; FAS: fatty acid synthetase; FRS: free radical scavenging; GAA: α-glucosidase; GLUT4: glucose transporter; GPx: glutathione peroxidase; GSH: glutathione; GST: glutathione-s-transferase; HDL: high-density lipoprotein; HPV: human papilloma virus; IκB: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor; LDH: lactate dehydrogenase; LOO: lipid peroxidation; LDL: low-density lipoprotein; HDL: high-density lipoprotein; MAPK: microtubule associated protein kinase; MMP: matrix metalloproteinase; Mo: molybdenum; INF-γ: interferon-γ; IL: interleukin; NF-κB: nuclear factor kappa B; PPARc2: proliferator-activated receptor γ 2; Rac: Ras-related C3 botulinum toxin substrate; Rb: retinoblastoma protein; Rho: Ras homolog family member; SOD: superoxide dismutase; SREBP-1c: sterol regulatory element-binding proteins 1c; TNF: tumor necrosis factor.

References

  1. Kirtikar, K.; Basu, B. Indian Medicinal Plants; Oriental Enterprises: Deharadun, India, 2001; Volume 4. [Google Scholar]
  2. Dalal, N.V.; Rai, V.R. In vitro propagation of Oroxylum indicum Vent. a medicinally important forest tree. J. For. Res. 2004, 9, 61–65. [Google Scholar] [CrossRef]
  3. Paranjpe, P. Indian Medicinal Plants; Chaukhamba Sanskrit Pratishthan: Delhi, India, 2005. [Google Scholar]
  4. Theobald, W. Bignoniaceae. In A Revised Handbook to the Flora of Ceylon; Dassanayake, M.D., Fosberg, F.R., Eds.; Amerind Publishing Co. Pvt. Ltd.: New Delhi, India, 1981. [Google Scholar]
  5. Chauhan, N. Medicinal and Aromatic Plants of Himachal Pradesh; Indus Publishing: New Delhi, India, 1999. [Google Scholar]
  6. Rasadah, M.A.; Houghton, P.J.; Raman, A.; Hoult, J.R.S. Antimicrobial and antiinflammatory activities of extracts and constituents of Oroxylum indicum (L.) Vent. Phytomedicine 1998, 5, 375–381. [Google Scholar] [CrossRef]
  7. Rasadah, M.A. Oroxylum indicum (L.) Kurz. In Plant Resources of South-East Asia No. 12 (2): Medicinal and Poisonous Plants 2; van Valkenberg, J.L.C.H., Bunyapraphatsara, N., Eds.; Backhuys Publisher: Leiden, The Netherlands, 2001; ISBN 9057820994. [Google Scholar]
  8. Dev, L.R.; Anurag, M.; Rajiv, G. Oroxylum indicum: A review. Pharmacogn. J. 2010, 2, 304–310. [Google Scholar] [CrossRef]
  9. Jayaram, K.; Prasad, M. Genetic diversity in Oroxylum indicum (L.) Vent. (Bignoniaceae), a vulnerable medicinal plant by random amplified polymorphic DNA marker. Afr. J. Biotechnol. 2008, 7, 254–262. [Google Scholar]
  10. Singh, A.K. Morphological expressions in seedlings of Oroxylum indicum (L.) Vent—A well known medicinal tree of tropics and subtropics. Mod. Phytomorphol. 2015, 8, 41–48. [Google Scholar]
  11. Warrier, P.; Nambiar, V.; Ramankutty, C. Indian Medicinal Plants—A Compendium of 500 Species; Orient Longman Ltd.: Chennai, India, 1995. [Google Scholar]
  12. Nakahara, K.; Trakoontivakorn, G.; Alzoreky, N.S.; Ono, H.; Onishi-Kameyama, M.; Yoshida, M. Antimutagenicity of some edible Thai plants, and a bioactive carbazole alkaloid, mahanine, isolated from Micromelum minutum. J. Agric. Food Chem. 2002, 50, 4796–4802. [Google Scholar] [CrossRef]
  13. Lalrinzuali, K.; Vabeiryureilai, M.; Jagetia, G.C. Phytochemical and TLC profiling of Oroxylum indicum and Milletia pachycarpa. J. Plant Biochem. Physiol. 2015, 3. [Google Scholar] [CrossRef] [Green Version]
  14. Lalrinzuali, K.; Vabeiryureilai, M.; Jagetia, G.C. Topical application of stem bark ethanol extract of Sonapatha, Oroxylum indicum (L.) Kurz accelerates healing of deep dermal excision wound in Swiss albino mice. J. Ethnopharmacol. 2018, 227, 290–299. [Google Scholar] [CrossRef]
  15. Dinda, B.; Silsarma, I.; Dinda, M.; Rudrapaul, P. Oroxylum indicum (L.) Kurz, an important Asian traditional medicine: From traditional uses to scientific data for its commercial exploitation. J. Ethnopharmacol. 2015, 161, 255–278. [Google Scholar] [CrossRef]
  16. Balkrishna, A. Ayurved Jadi-Booti Rahasya; Divyaprakasan, Divya Yog Mandir Trust: Haridwar, India, 2005; ISBN 81-89235-44-8. [Google Scholar]
  17. Singh, H.B.; Prasad, P.; Rai, L.K. Folk Medicinal Plants in the Sikkim Himalayas of India. Asian Folk Studs 2002, 61, 295–310. [Google Scholar] [CrossRef]
  18. Bhattacharje, S. Use of flavours and fragrances. In Handbook of Aromatic Plants, 2nd ed.; Bhattacharje, S.K., Ed.; Pointer Publishers: Jaipur, India, 2005; ISBN 8171324071. [Google Scholar]
  19. Khare, C. Indian Herbal Remedies: Rational Western Therapy, Ayurvedic, and Other Traditional Usage, Botany, 4th ed.; Springer: Berlin/Heidelberg, Germany; New York, NY, USA, 2004; ISBN 3-540-01026-2. [Google Scholar]
  20. Jamir, N.S.; Takatemjen; Limasemba. Traditional knowledge of Lotha-Naga tribes in Wokha district, Nagaland. Indian J. Tradit. Knowl. 2010, 9, 45–48. [Google Scholar]
  21. Gaur, R.D.; Sharma, J. Indigenous knowledge on the utilization of medicinal plant diversity in the Siwalik region of Garhwal Himalaya, Uttarakhand. J. For. Environ. Sci. 2011, 27, 23–31. [Google Scholar]
  22. Rout, S.D.; Panda, T.; Mishra, N. Ethno-medicinal plants used to cure different diseases by tribals of Mayurbhanj district of north Orissa. Stud. Ethno-Med. 2009, 3, 27–32. [Google Scholar] [CrossRef]
  23. Das, S.; Duttachoudhury, M.; Mandal, S.C.; Talukdar, A. Traditional knowledge of ethnomedicinal hepatoprotective plants used by certain ethnic communities of Tripura state. Indian J. Fundam. Appl. Life Sci. 2012, 2, 84–97. [Google Scholar]
  24. Nadkarni, K. Indian Materia Medica, 3rd ed.; Popular Prakashan: Bombay, India, 1982. [Google Scholar]
  25. Chopra, R.N.; Nayar, S.L.; Chopra, I.C. Glossary of Indian Medicinal Plants; National Institute of Science Communication and Information Resources: New Delhi, India, 2002. [Google Scholar]
  26. Drury, C.H. Ayurvedic Useful Plants of India; Asiatic Publishing House: Delhi, India, 2010; ISBN 819080913X. [Google Scholar]
  27. Patil, G.; Mali, P.; Bhadane, V. Folk remedies used against respiratory disorders in Jalgaon district, Maharashtra. Nat. Prod. Radiance 2008, 7, 354–358. [Google Scholar]
  28. Panghal, M.; Arya, V.; Yadav, S.; Kumar, S.; Yadav, J.P. Indigenous knowledge of medicinal plants used by Saperas community of Khetawas, Jhajjar District, Haryana, India. J. Ethnobiol. Ethnomed. 2010, 6. [Google Scholar] [CrossRef] [Green Version]
  29. National Register of Medicinal Plants; IUCN Nepal: Kathmandu, Nepal, 2000; ISBN 92-9144-048-5.
  30. Fazlin, A.; Ahmad, Z.; Lim, H.H. Compendium of Medicinal Plants Used in Malaysia; Herbal Medicine Research Centre, Institute for Medical Research, Aktif Bestari Sdn Bhd: Kuala Lumpur, Malaysia, 2002; Volume 2. [Google Scholar]
  31. DeFilipps, R.A.; Krupnick, G.A. The medicinal plants of Myanmar. PhytoKeys 2018, 102, 1–341. [Google Scholar] [CrossRef] [PubMed]
  32. Kalaivani, T.; Mathew, L. Phytochemistry and free radical scavenging activities of Oroxylum indicum. Environ. We Int. J. Sci. Technol. 2009, 4, 45–52. [Google Scholar]
  33. Radhika, L.G.; Meena, C.V.; Peter, S.; Rajesh, K.S.; Rosamma, M.P. Phytochemical and antimicrobial study of Oroxylum indicum. Anc. Sci. Life 2011, 30, 114–120. [Google Scholar]
  34. Samatha, T.; Srinivas, P.; Shyamsundarachary, R.; Rajinikanth, M.; Rama Swamy, N. Phytochemical analysis of seeds, stem bark and root of an endangered medicinal forest tree Oroxylum indicum (L.) Kurz. Int. J. Pharma Bio Sci. 2012, 3, B1063–B1075. [Google Scholar]
  35. Das, B.K.; Al-Amin, M.M.; Russel, S.M.; Kabir, S.; Bhattacherjee, R.; Hannan, J.M.A. Phytochemical screening and evaluation of analgesic activity of Oroxylum indicum. Indian J. Pharm. Sci. 2014, 76, 571–575. [Google Scholar] [PubMed]
  36. SatyaEswari, J.; Dhagat, S.; Naik, S.; Dibya, S. Oroxylum indicum leaf extracts for screening of antimicrobial properties and phytochemicals. Pharm. Bioprocess. 2018, 6, 7–14. [Google Scholar]
  37. Subramanian, S.; Nair, A. Flavanoids of the leaves of Oroxylum indicum and Pajanelia longifolia. Phytochemistry 1972, 11, 439–440. [Google Scholar] [CrossRef]
  38. Zaghloul, S.; Azzam, S.; Eid, H.; Hassan, H.; Sleem, A. Chemical and biological investigation of essential oil of Oroxylum indicum L. leaves cultivated in Egypt. Int. J. Pharmacogn. Phytochem. Res. 2015, 7, 570–575. [Google Scholar]
  39. Luitel, H.; Rajbhandari, M.; Kalauni, S.; Awale, S.; Masuda, K.; Gewali, M. Chemical constituents from Oroxylum indicum (L.) Kurz of Nepalese Origin. Sci. World 2010, 8, 66–68. [Google Scholar] [CrossRef] [Green Version]
  40. Babu, T.; Manjulatha, K.; Kumar, G.; Hymavathi, A.; Tiwari, A.; Purohit, M.; Rao, J.; Babu, K. Gastroprotective flavonoid constituents from Oroxylum indicum Vent. Bioorg. Med. Chem. Lett. 2010, 20, 117–120. [Google Scholar] [CrossRef]
  41. Nguyen, M.; Nguyen, N.; Nguyen, X.; Huynh, T.; Min, B. Screening of α-glucosidase inhibitory activity of Vietnamese medicinal plants: Isolation of active principles from Oroxylum indicum. Nat. Prod. Sci. 2012, 18, 47–51. [Google Scholar]
  42. Islam, K.; Eti, Z.; Chowdhury, J.A. Phytochemical and antimicrobial analysis on the extract of Oroxylum indicum Linn. stem-bark. Iran. J. Pharmacol. Ther. 2010, 9, 25–28. [Google Scholar]
  43. Fan, Q.F.; Hu, Z.Y.; Na, Z.; Tang, H.S.; Zuo, G.Y.; Song, Q.S. One new flavonoid from Oroxylum indicum. Nat. Prod. Res. 2015, 29, 1828–1832. [Google Scholar] [CrossRef]
  44. Zaveri, M.; Khandhar, A.; Jain, S. Quantification of baicalein, chrysin, biochanin-A and ellagic acid in root bark of Oroxylum indicum by RP-HPLC with UV detection. Eurasian J. Anal. Chem. 2008, 3, 245–257. [Google Scholar]
  45. Uddin, K.; Sayeed, A.; Islam, A.; Rahman, A.; Ali, A.; Khan, G.R.M.A.; Sadik, M. Purification, characterization and cytotoxic activity of two flavonoids from Oroxylum indicum Vent. (Bignoniaceae). Asian J. Plant Sci. 2003, 2, 515–518. [Google Scholar] [CrossRef]
  46. Yadav, A.K.; Manika, N.; Bagchi, D.; Gupta, M. Simultaneous determination of flavonoids in Oroxylum indicum by RP-HPLC. Med. Chem. Res. 2013, 22, 2222–2227. [Google Scholar] [CrossRef]
  47. Mehta, C.; Mehta, T. Tetuin, a glucoside from the seeds of Oroxylum indicum Vent. Curr. Sci. 1953, 22, 114. [Google Scholar]
  48. Nair, A.; Joshi, B. Oroxindin—A new flavone glucuronide from Oroxylum indicum Vent. Proc. Indian Acad. Sci. 1979, 88, 323–327. [Google Scholar]
  49. Chen, L.J.; Games, D.E.; Jones, J. Isolation and identification of four flavonoid constituents from the seeds of Oroxylum indicum by high-speed counter-current chromatography. J. Chromatogr. A 2003, 988, 95–105. [Google Scholar] [CrossRef]
  50. Yan, R.Y.; Cao, Y.Y.; Chen, C.Y.; Dai, H.Q.; Yu, S.X.; Wei, J.L.; Li, H.; Yang, B. Antioxidant flavonoids from the seed of Oroxylum indicum. Fitoterapia 2011, 82, 841–848. [Google Scholar] [CrossRef]
  51. Wei, X.N.; Lin, B.; Xie, G.Y.; Li, J.W.; Qin, M.J. Chemical constituents of seeds of Oroxylum indicum. Zhongguo Zhongyao Zazhi 2013, 38, 204–207. [Google Scholar] [CrossRef]
  52. Krüger, A.; Ganzera, M. Oroxylum indicum seeds-analysis of flavonoids by micellar electrokinetic chromatography. Chromatography 2013, 1, 1–8. [Google Scholar] [CrossRef] [Green Version]
  53. Li, G.; Wang, G.; Tong, Y.; Zhu, J.; Yun, T.; Ye, X.; Li, F.; Yuan, S.; Liu, Q. Concise synthesis and antidiabetic activity of natural flavonoid glycosides, oroxins C and D, isolated from the seeds of Oroxylum indium. J. Chem. Res. 2021, 45, 68–75. [Google Scholar] [CrossRef]
  54. Rojsanga, P.; Bunsupa, S.; Sithisarn, P. Flavones contents in extracts from Oroxylum indicum seeds and plant tissue cultures. Molecules 2020, 25, 1545. [Google Scholar] [CrossRef] [Green Version]
  55. Rojsanga, P.; Bunsupa, S.; Brantner, A.H.; Sithisarn, P. Comparative phytochemical profiling and in vitro antioxidant activity of extracts from raw materials, tissue-cultured plants, and callus of Oroxylum indicum (L.) Vent. Evid. Based Complement. Altern. Med. 2017, 2017, 6853212. [Google Scholar] [CrossRef] [PubMed]
  56. Jiwajinda, S.; Santisopasri, V.; Murakami, A.; Kim, O.; Kim, H.; Ohigashi, H. Suppressive effects of edible Thai plants on superoxide and nitric oxide generation. Asian Pac. J Cancer Prev. 2002, 3, 215–223. [Google Scholar] [PubMed]
  57. Chumkaew, P.; Srisawat, T. A new flavone from Oroxylum indicum and its antibacterial activity. Chem. Nat. Compd. 2021, 57, 274–276. [Google Scholar] [CrossRef]
  58. Mishra, S.L.; Sinhamahapatra, P.K.; Nayak, A.; Das, R.; Sannigrahi, S. In vitro antioxidant potential of different parts of Oroxylum indicum: A comparative study. Indian J. Pharm. Sci. 2010, 72, 267–269. [Google Scholar] [CrossRef] [Green Version]
  59. Tenpe, C.; Upaganlawar, A.; Burle, S.; Yeole, P. In vitro antioxidant and preliminary hepatoprotective activity of Oroxylum indicum Vent leaf extracts. Pharmacologyonline 2009, 1, 35–43. [Google Scholar]
  60. Gupta, R.C.; Sharma, V.; Sharma, N.; Kumar, N.; Singh, B. In vitro antioxidant activity from leaves of Oroxylum indicum (L.) Vent.—A North Indian highly threatened and vulnerable medicinal plant. J. Pharm. Res. 2008, 1, 65–72. [Google Scholar]
  61. Kumar, V.; Chaurasia, A.K.; Naglot, A.; Gopalakrishnan, R.; Gogoi, B.J.; Singh, L.; Srivastava, R.B.; Deka, D.C. Antioxidant and antimicrobial activities of stem bark extracts of Oroxylum indicum Vent. (Bignoniaceae)—A medicinal plant of northeastern India. South Asian J. Exp. Biol. 2011, 1, 152–157. [Google Scholar]
  62. Moirangthem, D.S.; Talukdar, N.C.; Bora, U.; Kasoju, N.; Das, R.K. Differential effects of Oroxylum indicum bark extracts: Antioxidant, antimicrobial, cytotoxic and apoptotic study. Cytotechnology 2013, 65, 83–95. [Google Scholar] [CrossRef] [Green Version]
  63. Lalrinzuali, K.; Vabeiryureilai, M.; Jagetia, G.C.; Lalawmpuii, P.C. Free radical scavenging and antioxidant potential of different extracts of Oroxylum indicum in vitro. Adv. Biomed. Pharma 2015, 2, 120–130. [Google Scholar] [CrossRef]
  64. Sithisarn, P.; Nantateerapong, P.; Rojsanga, P.; Sithisarn, P. Screening for antibacterial and antioxidant activities and phytochemical analysis of Oroxylum indicum fruit extracts. Molecules 2016, 21, 446. [Google Scholar] [CrossRef]
  65. Upaganlawar, A.; CR, T.; Yeole, P. Antiinflammatory activity of aqueous extract of Oroxylum indicum Vent. leaves extract-preliminary study. Pharmacol. Online 2009, 1, 22–26. [Google Scholar]
  66. Doshi, K.; Ilanchezhian, R.; Acharya, R.; Patel, B.R.; Ravishankar, B. Anti-inflammatory activity of root bark and stem bark of Shyonaka. J. Ayurveda Integr. Med. 2012, 3, 194–197. [Google Scholar] [CrossRef] [Green Version]
  67. Joshi, S.V.; Vyas, B.A.; Shah, P.D.; Shah, D.R.; Shah, S.A.; Gandhi, T.R. Protective effect of aqueous extract of Oroxylum indicum Linn. (root bark) against DNBS-induced colitis in rats. Indian J. Pharmacol. 2011, 43, 656–661. [Google Scholar] [CrossRef]
  68. Asaduzzaman, M.; Nasrin, N.; Muhit, A.; Raihan, S.; Apu, A.; Akbar, A. Antidiarrheal, analgesic and cytotoxic activities of crude extract of Oroxylum indicum (L.) stem bark. J. Pharm. Res. 2011, 4, 4296–4298. [Google Scholar]
  69. Hu, T.; Liu, S.; Zhao, L.; Qin, K. Preparation of Oroxylum indicum extracts and their anti-bacterial and anti-inflammatory activities. China Anim. Husb. Vet. Med. 2010, 37, 225–228. [Google Scholar]
  70. Lalrinzuali, K.; Vabeiryureilai, M.; Jagetia, G.C. Investigation of the anti-inflammatory and analgesic activities of ethanol extract of stem bark of sonapatha Oroxylum indicum in vivo. Int. J. Inflam. 2016, 2016, 8247014. [Google Scholar] [CrossRef] [Green Version]
  71. Laloo, D.; Gogoi, B.; Lyngdoh, W.; Zaman, K.; Sharma, H. Antioxidant, analgesic and anti-inflammatory activities of bark of Oroxylum indicum Vent: An endemic medicinal plant of Northeast India. Asian J. Chem. 2016, 28, 2272–2276. [Google Scholar] [CrossRef]
  72. Begum, M.M.; Islam, A.; Begum, R.; Uddin, M.S.; Rahman, M.S.; Alam, S.; Akter, W.; Das, M.; Rahman, M.S.; Imon, A.H.M.R. Ethnopharmacological inspections of organic extract of Oroxylum indicum in rat models: A promising natural gift. Evid. Based. Complement. Alternat. Med. 2019, 2019, 1562038. [Google Scholar] [CrossRef] [Green Version]
  73. Bergeron, C.; Hamid, Q. Relationship between asthma and rhinitis: Epidemiologic, pathophysiologic, and therapeutic aspects. Allergy Asthma Clin. Immunol. 2005, 1, 81–87. [Google Scholar] [CrossRef]
  74. Lee, A.Y.; Kang, S.; Park, S.J.; Huang, J.; Im, D.S. Anti-allergic effect of oroxylin a from Oroxylum indicum using in vivo and in vitro experiments. Biomol. Ther. 2016, 24, 283–290. [Google Scholar] [CrossRef] [Green Version]
  75. Rasadah, M.A.; Houghton, P.J.; Hoo, T.S. Antifungal activity of some Bignoniaceae found in Malaysia. Phyther. Res. 1998, 12, 331–334. [Google Scholar] [CrossRef]
  76. Samatha, T.; Sampath, A.; Sujatha, K.; Rama Swamy, N. Antibacterial activity of stem bark extracts of Oroxylum indicum an endangered ethnomedicinal forest tree. IOSR J. Pharm. 2011, 7, 24–28. [Google Scholar]
  77. Das, S.; Choudhury, M.D. Antimicrobial activity of stem bark extracts from the plant Oroxylum indicum Vent. Assam Univ. J. Sci. Technol. 2010, 5, 95–99. [Google Scholar]
  78. Deori, K.; Yadav, A.K. Anthelmintic effects of Oroxylum indicum stem bark extract on juvenile and adult stages of Hymenolepis diminuta (Cestoda), an in vitro and in vivo study. Parasitol. Res. 2016, 115, 1275–1285. [Google Scholar] [CrossRef]
  79. Islam, M.; Rahman, M.M.; Hasan, M.; Rahaman, M.; Khan, M.R.H.; Al-Faysal, A. Evaluation of anthelmintic activity of crude extracts and different fractions of stem bark and fruits of Oroxylum indicum. World J. Pharm. Res. 2016, 5, 287–294. [Google Scholar] [CrossRef]
  80. Tripathy, B.; Panda, S.; Sahoo, S.; Mishra, S.; Nayak, L. Phytochemical analysis and hepatoprotective effect of stem bark of Oroxylum indicum (L) Vent. on carbon tetrachloride induced hepatotoxicity in rat. Int. J. Pharm. Biol. Arch. 2011, 2, 1714–1717. [Google Scholar]
  81. Sastry, A.; Sastry, V.; Mallikarjun, P.; Srinivas, K. Chemical and pharmacological evaluation of aqueous extract of root bark of “Oroxylum indicum” Vent. Int. J. Pharm. Technol. 2011, 3, 1796–1806. [Google Scholar]
  82. Mishra, S.; Sahoo, S. In vitro and in vivo antihepatotoxic activity of Oroxylum indicum against carbon-tetrachloride induced hepatic damage. Int. J. Pharm. Sci. Res. 2013, 4, 3202–3207. [Google Scholar] [CrossRef]
  83. Bharali, M.K.; Konya, H.; Bahadur, C.L. Protective effect of Oroxylum indicum on acetaminophen induced liver injury in rat. Int. Curr. Pharm. J. 2014, 3, 223–227. [Google Scholar] [CrossRef] [Green Version]
  84. More, A.; Shah, T.; Parab, P.; Apte, K. Oroxylum indicum (Linn.) whole stem extract regulates expression of TNFα, IL6, NFkB, P38 MAPK and oxidative status in antitubercular therapy induced hepatotoxicity in Wistar rats. Matters 2017, 3, e201704000014. [Google Scholar] [CrossRef] [Green Version]
  85. Mohapatra, S.S.; Roy, R.; Mohan, P.; Upadhyaya, T.; Sarma, J. Phytochemical analysis and hepatoprotective effect of hydroethanolic extract of stem bark of Oroxylum indicum. Int. J. Curr. Microbiol. Appl. Sci. 2018, 7, 1000–1006. [Google Scholar] [CrossRef]
  86. Sun, W.; Liu, P.; Yang, B.; Wang, M.; Wang, T.; Sun, W.; Wang, X.; Zheng, W.; Song, X.; Li, J. A network pharmacology approach: Inhibition of the NF-κB signaling pathway contributes to the NASH preventative effect of an Oroxylum indicum seed extract in oleic acid-stimulated HepG2 cells and high-fat diet-fed rats. Phytomedicine 2021. [Google Scholar] [CrossRef]
  87. Zaveri, M.; Jain, S. Gastroprotective effects of root bark of Oroxylum indicum, Vent. J. Nat. Remedies 2007, 7, 269–277. [Google Scholar] [CrossRef]
  88. Menon, S.; Lawrence, L.; Sivaram, V.P.; Padikkala, J. Oroxylum indicum root bark extract prevents doxorubicin-induced cardiac damage by restoring redox balance. J. Ayurveda Integr. Med. 2019, 10, 159–165. [Google Scholar] [CrossRef] [PubMed]
  89. Tamboli, A.; Karpe, S.; Shaikh, S.; Manikrao, A. Hypoglycemic activity of extracts of Oroxylum indicum (L.) vent roots in animal models. Pharmacologyonline 2011, 2, 890–899. [Google Scholar]
  90. Singh, J.; Kakkar, P. Modulation of liver function, antioxidant responses, insulin resistance and glucose transport by Oroxylum indicum stem bark in STZ induced diabetic rats. Food Chem. Toxicol. 2013, 62, 722–731. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, B.W.; Sang, Y.B.; Sun, W.; Yu, H.S.; Ma, B.P.; Xiu, Z.L.; Dong, Y.S. Combination of flavonoids from Oroxylum indicum seed extracts and acarbose improves the inhibition of postprandial blood glucose: In vivo and in vitro study. Biomed. Pharmacother. 2017, 91, 890–898. [Google Scholar] [CrossRef]
  92. Sun, W.; Sang, Y.; Zhang, B.; Yu, X.; Xu, Q.; Xiu, Z.; Dong, Y. Synergistic effects of acarbose and an Oroxylum indicum seed extract in streptozotocin and high-fat-diet induced prediabetic mice. Biomed. Pharmacother. 2017, 87, 160–170. [Google Scholar] [CrossRef] [PubMed]
  93. Kaldate, P.; Tenpe, C.; Rapid, P.Y. Antidiabetic activity of leaves of Oroxylum indicum in alloxan-induced diabetic rats. Inven. Rapid Ethnopharmacol. 2011, 1, 468. [Google Scholar]
  94. Mishra, S.B.; Simon, M.; Mukerjee, A.; Rani, S.; Parashar, T. Phytochemical investigation and antidiabetic activity of Oroxylum indicum Vent. seed. SSRN Electron. J. 2018. [Google Scholar] [CrossRef]
  95. Mangal, P.; Khare, P.; Jagtap, S.; Bishnoi, M.; Kondepudi, K.K.; Bhutani, K.K. Screening of six Ayurvedic medicinal plants for anti-obesity potential: An investigation on bioactive constituents from Oroxylum indicum (L.) Kurz bark. J. Ethnopharmacol. 2017, 197, 138–146. [Google Scholar] [CrossRef] [PubMed]
  96. Hengpratom, T.; Lowe, G.M.; Thumanu, K.; Suknasang, S.; Tiamyom, K.; Eumkeb, G. Oroxylum indicum (L.) Kurz extract inhibits adipogenesis and lipase activity in vitro. BMC Complement. Altern. Med. 2018, 18, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Hengpratom, T.; Ngernsoungnern, A.; Ngernsoungnern, P.; Lowe, G.M.; Eumkeb, G. Antiadipogenesis of Oroxylum indicum (L.) Kurz extract via PPAR γ 2 in 3T3-L1 adipocytes. Evid. Based Complement. Altern. Med. 2020, 2020, 6720205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Rajkumar, V.; Guha, G.; Kumar, R. Induction of apoptosis in MDA-MB-435S, Hep3B and PC-3 cell lines by extracts of Oroxylum indicum. J. Pharm. Res. 2011, 4, 2054–2056. [Google Scholar]
  99. Kumar, D.R.N.; George, V.C.; Suresh, P.K.; Kumar, R.A. Cytotoxicity, apoptosis induction and anti-metastatic potential of Oroxylum indicum in human breast cancer cells. Asian Pac. J. Cancer Prev. 2012, 13, 2729–2734. [Google Scholar] [CrossRef] [Green Version]
  100. Zazali, K.E.; Abdullah, H.; Jamil, N.I.N. Methanol extract of Oroxylum indicum leaves induces G1/S cell cycle arrest in HeLa cells via p53-mediated pathway. Int. J. Med. Plant Res. 2013, 2, 225–237. [Google Scholar]
  101. Kumari, N.; Kumar, R.; Sinha, K.; Kumar, S. Anticancerous potentiality of in vivo grown twigs and in vitro generated plantlets of endangered Oroxylum indicum (L.) Vent. IOSR J. Biotechnol. Biochem. 2018, 4, 21–27. [Google Scholar] [CrossRef]
  102. Dhru, B.; Bhatt, D.; Jethva, K.; Zaveri, M. In vitro cytotoxicity studies of the anti-cancer potential of fractions of root bark of Oroxylum indicum in human breast carcinoma cells. Int. J. Pharm. Sci. Rev. Res. 2016, 38, 18–21. [Google Scholar]
  103. Buranrat, B.; Noiwetch, S.; Suksar, T.; Ta-ut, A.; Boontha, S. Cytotoxic and antimigration effects of different parts of Oroxylum indicum extract on human breast cancer MCF-7 cells. Sci. Technol. Asia 2018, 23, 42–52. [Google Scholar] [CrossRef]
  104. Buranrat, B.; Noiwetch, S.; Suksar, T.; Ta-ut, A. Inhibition of cell proliferation and migration by Oroxylum indicum extracts on breast cancer cells via Rac1 modulation. J. Pharm. Anal. 2020, 10, 187–193. [Google Scholar] [CrossRef]
  105. Wahab, N.; Din, N.; Lim, Y.; Jamil, N.; Mat, N. Proapoptotic activities of Oroxylum indicum leave extract in HeLa cells. Asian Pac. J. Trop. Biomed. 2019, 9, 339–345. [Google Scholar]
  106. Singh, H.; Singh, V.; Kumar, D.; Chaudhary, A. Wound healing and antimicrobial potential of Oroxylum indicum Vent. in albino mice. Nat. Prod. J. 2011, 1, 128–134. [Google Scholar] [CrossRef]
  107. Stasi, C.; Fallani, S.; Voller, F.; Silvestri, C. Treatment for COVID-19: An overview. Eur. J. Pharmacol. 2020, 889, 173644. [Google Scholar] [CrossRef] [PubMed]
  108. Gao, J.; Ding, Y.; Wang, Y.; Liang, P.; Zhang, L.; Liu, R. Oroxylin A is a severe acute respiratory syndrome coronavirus 2-spiked pseudotyped virus blocker obtained from Radix scutellariae using angiotensin-converting enzyme II/cell membrane chromatography. Phyther. Res. 2021, 10, 1002. [Google Scholar] [CrossRef]
  109. Song, J.; Zhang, L.; Xu, Y.; Yang, D.; Yang, S.; Zhang, W.; Wang, J.; Tian, S.; Yang, S.; Yuan, T.; et al. The comprehensive study on the therapeutic effects of baicalein for the treatment of COVID-19 in vivo and in vitro. Biochem. Pharmacol. 2021, 183, 114302. [Google Scholar] [CrossRef] [PubMed]
  110. Huang, S.; Liu, Y.; Zhang, Y.; Zhang, R.; Zhu, C.J.; Fan, L.; Pei, G.; Zhang, B.; Shi, Y. Baicalein inhibits SARS-CoV-2/VSV replication with interfering mitochondrial oxidative phosphorylation in a mPTP dependent manner. Signal Transduct. Target. Ther. 2020, 5, 266. [Google Scholar] [CrossRef] [PubMed]
  111. Shah, S.; Chaple, D.; Arora, S.; Yende, S.; Moharir, K.; Lohiya, G. Exploring the active constituents of Oroxylum indicum in intervention of novel coronavirus (COVID-19) based on molecular docking method. Netw. Model. Anal. Health Informat. Bioinform. 2021, 10, 8. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Oroxylum indicum: (a): whole tree; (b): leaves; (c): stem; (d): stem bark; (e): flowers; (f): fruits.
Figure 1. Oroxylum indicum: (a): whole tree; (b): leaves; (c): stem; (d): stem bark; (e): flowers; (f): fruits.
Sinusitis 05 00009 g001
Figure 2. Mechanisms of action of Sonapatha (Oroxylum indicum). Red: activation/upregulation and blue: downregulation/reduction/deactivation.
Figure 2. Mechanisms of action of Sonapatha (Oroxylum indicum). Red: activation/upregulation and blue: downregulation/reduction/deactivation.
Sinusitis 05 00009 g002
Table 1. Common names of Oroxylum indicum in different languages.
Table 1. Common names of Oroxylum indicum in different languages.
LanguageCommon NamesReferences
EnglishMidnight horror, Indian trumpet flower, broken bones plant, Tree of Damocles, Indian calosanthes, Indian caper, Indian trumpet[1,2,3,5]
HindiShyonak, Sonapatha, Patrorna, Shallaka, Putivriksha, Shuran, Sauna, Son, Aralu, Vatuk, Urru, Bhut-Vriksha, Dirghavrinta, Kutannat[1,2,3,5]
SanskritShyonaka, Nat, Katvanga, Sukanasa, Prithu Simba, Tuntuka, Dirghavrinta, Mandukparana, Kutannata, Patroma, Katammar, Bhalluka, Mayurjangha, Aralu[1,2,3,5]
GujaratiTentu [1,2,3,5]
KannadaAne-mungu Patagani, Bunepaale, Sonepatta, Tigadu, Alangi, and Tattuna[1,2,3,5]
TeluguDundilamu, Manduka-paramu, Chettu, Nemali, Pampena, Pampini, Suka-nasamu [1,2,3,5]
BengaliKhonha, Sona, Kani-Dingi, Paharijora, Hanghoal, Thona Gach, Aklong-Singh, Krong-Sa-Bang (Marma Tribe), Naori Chilana (Chakma Tribe), Kaak-Rakung (Halam Tribe), Tou-Kharung Tripura Tribe), Kanai Dingi (Garo Tribe)[1,2,3,5]
KonkaniDavamadak[1,2,3,5]
TamilAchi, Arandei, Cori-Konnai, Venga maram, Palai-yutaicci, Pana, Puta-Puspam (the flower), Pei-maram, Peiarlanke, Palagaipayani, Peruvaagai or Vangam[1,2,3,5]
MarathiTetu, Tayitu[1,2,3,5]
AssameseBhatghila, Dingdinga, or Toguna [1,2,3,5]
MalayalamVashrppathiri, Palaqapayyani, or Vellapathiri[1,2,3,5]
OriyaArchangkawm, Phanphania, Tatelo[1,2,3,5]
ChineseHandy pinyin, Mud huddle [1,2,3,5]
NepaleseTatelo [1,2,3,5]
SinghaleseThotila, Totila [1,2,3,5]
MalayBonglai [1,2,3,5,6]
Sundanese JavanesePongporang
Kayu lanang, Mungli
[6,7,8]
FilipinoPingka-pingkahan (Tagalog), Abong-abong (Bisaya), Kamkampilan (Iloko)[6,7,8]
Khmer Pi ka[6,7,8]
Lao Lin may, Ung ka [6,7,8]
ThaiPhe kaa (central), Litmai (northern), Lin faa (north-eastern) [6,7,8]
VietnameseNúc nác, Hoàng bá nam, Mộc hồ diệp[6,7,8]
Table 2. Different phytoconstituents detected in the Oroxylum indicum.
Table 2. Different phytoconstituents detected in the Oroxylum indicum.
Plant PartExtract TypePhytoconstituentsReference
Stem barkChloroform, ethanol aqueous
Benzene, chloroform, ethanol
Phenolics, flavonoids, saponins
Alkaloids
[32]
Stem and root bark HydromethanolGlycosides, terpenoids phenols, alkaloids[33]
Seeds Acetone, benzene, methanol, water Alkaloids, flavonoids, tannins, glycosides, sterols, phenols, saponins, fats, oils[34]
Stem bark Acetone, benzene, chloroform, methanol Tannins, saponins, sterols, phenols, fats, oils[34]
Root barkAcetone, benzene, chloroform, methanol, waterFlavonoids, alkaloids, glycosides, tannins, sterols, phenols, lignins, saponins, fats, oils[34]
Stem Bark Ethanol Alkaloids, flavonoids, tannins, glycosides[35]
Stem barkChloroform, ethanol, water
Chloroform, ethanol, water
Alkaloids, flavonoids, tannins
Cardiac glycosides, saponins
[13]
Tender leavesMethanolPhlobatannins, flavonoids, phenols and tannins, glycosides[36]
Table 3. Different active chemicals isolated from various parts of Oroxylum indicum.
Table 3. Different active chemicals isolated from various parts of Oroxylum indicum.
PartsIsolated ChemicalReference
LeavesChrysin, oroxylin-A, scutellarin, scutellarin, baicalein, baicalein-6-O-glucuronide, baicalein, baicalein-7-O-glucoside, baicalein-7-O-diglucoside, baicalein-7-O-glucuronide, chrysin, chrysin-diglucoside, chrysin-7-O-glucuronide, ar-tumerone, methyl hexadecanoate, laurenan-2-one, isopropyl butanoate.[37,38]
StemOroxylin A, scutellarin, baicalein, baicalein-6-O- glucuronide, dihydrobaicalein, ellagic acid, dihydrooroxylin-A, methyl-3,-4,5-trihydroxy-6-(5-hydroxy-6-methoxy-4-oxo-2-phenylchroman-7-yloxy)-tetrahydro-2H-pyran-2-carboxylate, 5-hydroxyl-7-methoxy-2-(2-methoxy-6-(3,4,5-trihydroxy-6-(hydroxymethyl)-tetrahydro-2H-pyran-2-yoloxy)phenyl)-4H-chromen-4-one, chrysin, oroxylin A, oroxyloside, hispidulin, apigenin, ficusal, balanophonin, 2-(1-hydroxy- methylethyl)-4H,9H-naphtho[2,3-b]furan-4,9-dione, salicylic acid, ρ-hydroxybenzoic acid, protocatechuic acid, isovanillin, β-hydroxypropiovanillon, baicalein, oroxylin, pinostrobin, stigmast-7-en-3-ol, 3,4′,5,7 tetrahydroxy flavonol (kaempferol), 5,6,7-trimethoxyflavone-8-Odglucopyranoside, oroxylin A-7-Odglucuronide butyl ester, 6-methoxy-baicalein, oroxylin-A-7-O-glucoside, 5,7-dihydroxyflavone, baicalein 6-methoxy-7-glucuronide, dehydro-iso-α-lapachone, lapachol.[6,14,39,40,41,42,43]
Root Ellagic acid, baicalein, chrysin, biochanin-A, oroxylin A, baicalein, hispidulin, 2,5-dihydroxy-6,7-dimethoxy flavone, 3,7,3′,5′-tetra- methoxy-4-hydroxy flavone, lapachol, β-sitosterol[6,44,45,46]
SeedsBaicalein-6-glucoside (tetuin), oroxindin, oroxylin-A, scutellarin, baicalein, oroxin A, oroxin B, chrysin, baicalin, quercetin, apigenin, kaempferol, quercetin-3-O-arabinopyranoside, lupeol, lup-20 (29)-ene-2α,3β-diol, pinosylvin, dihydropinosylvin, cholest-5-ene-3, 7-diol, rengyol, isorengyol, zarzissine, (E)-pino-sylvin-3-Odglucopyranoside, adenosine, sitosterol, daucosterol, chrysin 6-C-βdglucopyranosyl-8-Od glucuronopyranoside, baicalein 7-Odglucuronopyranosyl(1→3)-[βdglucopyranosyl-(1→6)]-βdglucopyranoside, scutellarein 7-Odglucopyranosyl-(1-6)-βdglucopyranoside, chrysin-7-Ogentiobioside, baicalein-7-O-diglucoside, baicalein-7-O-glucoside, scutellarein-7-O-glucopyranoside, chrysin-7-O-glucuronide, chrysin-6-C-βdglucopyranosyl-8-C-α-L-arabinopyranoside, baicalein, oroxylin-A, pinocembrin, pinobanksin, 2-methyl-6-phenyl-4H-pyran-4-one, lupeol, 2α-hydroxyllupeol, echinulin, adenosine, dimethyl sulfone, β-sitosterol, baicalein-7-O-gentiobioside, baicalein-7-O-glucoside, baicalein-7-O-glucuronide, oroxin C, oroxin D[47,48,49,50,51,52,53,54]
FlowerBaicalein, chrysin[55]
FruitOroxylin A, orlumin A, chrysin, ursolic acid[56,57]
Table 4. Free radical scavenging and antioxidant activity of Oroxylum indicum.
Table 4. Free radical scavenging and antioxidant activity of Oroxylum indicum.
Plant PartActivity TypeReference
Stem barkAntioxidant[32]
Root/root bark, stem/stem bark, leaf, fruitHydroxyl, nitric oxide, superoxide, DPPH radical scavenging[58]
Leaf DPPH, nitric oxide, superoxide, hydroxyl radical scavenging, reducing power [59,60]
Stem Bark DPPH, nitric oxide, superoxide, hydroxyl radical scavenging, reducing power [32,61,62,63]
Seed DPPH [64]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jagetia, G.C. A Review on the Medicinal and Pharmacological Properties of Traditional Ethnomedicinal Plant Sonapatha, Oroxylum indicum. Sinusitis 2021, 5, 71-89. https://doi.org/10.3390/sinusitis5010009

AMA Style

Jagetia GC. A Review on the Medicinal and Pharmacological Properties of Traditional Ethnomedicinal Plant Sonapatha, Oroxylum indicum. Sinusitis. 2021; 5(1):71-89. https://doi.org/10.3390/sinusitis5010009

Chicago/Turabian Style

Jagetia, Ganesh Chandra. 2021. "A Review on the Medicinal and Pharmacological Properties of Traditional Ethnomedicinal Plant Sonapatha, Oroxylum indicum" Sinusitis 5, no. 1: 71-89. https://doi.org/10.3390/sinusitis5010009

Article Metrics

Back to TopTop