Next Article in Journal
Flavonoids and Phenols, the Potential Anti-Diabetic Compounds from Bauhinia strychnifolia Craib. Stem.
Next Article in Special Issue
Polyphenol Extract from “Greco” Grape Canes: Characterization, Antioxidant Capacity, and Antitumor Effects on Cal-33 and JHU-SCC-011 Head and Neck Squamous Cell Carcinoma
Previous Article in Journal
Antibacterial Activity of a Novel Oligosaccharide from Streptomyces californics against Erwinia carotovora subsp. Carotovora
Previous Article in Special Issue
Phytochemical Constituents and Pharmacological Potential of Tamus communis Rhizomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phytoconstituents and Pharmacological Activities of Indian Camphorweed (Pluchea indica): A Multi-Potential Medicinal Plant of Nutritional and Ethnomedicinal Importance

1
Department of Chemistry, Preparatory Year Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
2
Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
3
Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
4
Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(8), 2383; https://doi.org/10.3390/molecules27082383
Submission received: 22 March 2022 / Revised: 3 April 2022 / Accepted: 5 April 2022 / Published: 7 April 2022

Abstract

:
Pluchea indica (L.) Less. (Asteraceae) commonly known as Indian camphorweed, pluchea, or marsh fleabane has gained great importance in various traditional medicines for its nutritional and medicinal benefits. It is utilized to cure several illnesses such as lumbago, kidney stones, leucorrhea, inflammation, gangrenous and atonic ulcer, hemorrhoids, dysentery, eye diseases, itchy skin, acid stomach, dysuria, abdominal pain, scabies, fever, sore muscles, dysentery, diabetes, rheumatism, etc. The plant or its leaves in the form of tea are commonly used for treating diabetes and rheumatism. The plant is a rich source of calcium, vitamin C, dietary fiber, and β-carotene. Various biomolecules have been isolated from P. indica, including thiophenes, terpenes, quinic acids, sterols, lignans, phenolics, and flavonoids. The current review reports detailed information about the phytoconstituents and pharmacological relevance of P. indica and the link to its traditional uses. The reported studies validated the efficacy and safety of P. indica, as well as supported its traditional uses for treating various ailments and promoting health and well-being. Thus, this could encourage the development of this plant into a healthy food supplement or medicine for the prevention and treatment of various diseases. However, further studies on the drug interactions, mechanism of action, pharmacokinetics, toxicology, and metabolism, as well as clinical trials, should be carried out.

1. Introduction

Plants play a remarkable role in medicine development because of their capacity to biosynthesize secondary metabolites with significant bioactivities [1,2,3,4]. Traditionally, plants were utilized to treat various ailments [5,6,7]. It was reported that more than 80% of the population still rely on folk and traditional medicine, most of which are based on plant remedies according to the WHO [1]. The global herbal trade of medicinal plants has been growing exponentially at the rate of 15% annual growth and is likely to reach a scale of five trillion USD by 2050 [8]. Medicinal plants are often cheaper, more easily obtainable, and have fewer side effects than synthetic drugs [9,10,11]. Many promising compounds have been discovered from plants that can be utilized in modifying the existing drugs or designing completely new ones [1,2,3,4]. The Asteraceae (Compositae) family is one of the largest plant families, which includes about 24,000–30,000 species and 1600–1700 genera [12]. Its plants are shrubs and herbs, which have been commonly used since ancient times as herbal medicines and diets all over the world [13]. It contains well-known species, such as sunflower, chicory, coreopsis, lettuce, daisy, and dahlias, as well as several significant medicinal plants, such as chamomile, wormwood, and dandelion [14].
Pluchea is a flowering plants genus of the Asteraceae family, comprising about 80 species [15]. Its members are known as plucheas, camphorweeds, or fleabanes, and some are called sour bushes [16]. Pluchea indica (L.) Less. is an evergreen shrub found abundantly in salt marshes and mangrove swamps with a 1 to 2 m height that has an important role in maintaining the ecological balance in the coastal areas [17]. It is known as Indian camphorweed, pluchea, or marsh fleabane, Khlu (Thai), Kukrakonda, Kakronda, or Munjhu rukha (Bengali), Kuo bao ju (Chinese), luntas (Javanese), Beluntas (Malaysia, Indonesia, Bahasa), and kalapini (Philippines) [18]. This plant is mainly found in the subtropical and tropical zones of Asia, Africa, Australia, and America and in warm temperature regions of countries such as Indonesia, Malaysia, Taiwan, Australia, Mexico, and India [5,17,18]. A chemical investigation of this plant revealed the existence of various phytoconstituents: thiophenes, sesquiterpenes, quinic acids, sterols, lignans, and flavonoids. Additionally, this plant has wide folk uses and diverse bioactivities: anti-inflammatory, anti-cancer, antioxidant, anti-microbial, and insecticidal activities. The current review summarizes the reported literature on the traditional uses, phytoconstituents, and bioactivities of this plant and isolated metabolites to highlight its positive influences on human health.

2. Methodology

The literature search was carried out through Google Scholar, scientific databases (e.g., Scopus, PubMed, and Web of Science), and different publishers (Taylor & Francis, Elsevier, Wiley, Thieme, Bentham, Sage, Springer, and Wiley). This current work reviews the ethnomedicinal uses and phytochemical and pharmacological activities of Pluchea indica. This review contains 140 references dating from 1985 until January 2022.

3. Morphology

The plant is a branching shrub up to 2 m tall. Its leaves are light green, obovate with a sharp acute apex, serrated edge, and subtly fine trichomes decorating its lower and upper surfaces. Moreover, the leaves are alternate, shortly stalked or stalkless with membranous leaf blades [19]. The base is blunted, and leaf bones are pinnate-formed with parchment-like intervenium. An aroma is produced when the leaf blades are crushed [20,21,22]. The stem is strong, rigid, woody, and round with monopodial branches. It has taproot branches with soft-textured root fibers. The inflorescence is a capitulum that grows in dense clusters in the leaf axils and at the branch tips with violet/white corolla [19]. Its flowers have a cup-like structure that bears tiny fruits. The fruit is dry indehiscent, one-seeded, brown, and cylindrical ≈1.0 mm long [19].

4. Nutritional Value

The leaves are a good source of calcium, dietary fiber, and β-carotene. Sudjaroen reported the nutritional values of P. indica, Holy basil, and Indian mulberry leaves. It was found that P. indica leaves contain seven times more calcium (251 mg/100 g) and two times more β-carotene (1225 μg/100 g) than those of Holy basil leaves (32 mg/100 g and 812 μg/100 g, respectively) [23]. One hundred grams of leaves have 251 mg of calcium, whereas nearly 297 mg of calcium are found in one serving (8 oz.) of 2% fat milk [24]. One hundred grams of the leaves contains water (87.53 g), protein (1.79 g), fat (0.49 g), dietary fiber (insoluble 0.89 g, soluble 0.45 g, and total 1.34 g), carbohydrate (8.65 g), calcium (251 mg), β-carotene (1225 μg), and vitamin C (30.17 μg) [23]. P. indica leaves have a sweet and astringent taste and aromatic flavor [23]. Blanched or raw leaves are eaten with Nam Prik (freshly prepared chili paste) as a side dish. In addition, they are utilized as one of the ingredients in several dishes, such as Kang ped (spicy coconut milk soup) and yum (spicy and sour salad) [25].

5. Ethnomedicinal Uses

All parts of P. indica can be used medicinally, and it has a long tradition in alternative medicine for a wide variety of ailments. In Indochina, the roots’ decoction is prescribed for fever as a diaphoretic, and its leaves’ infusion is taken internally in lumbago. The leaves and roots are utilized as an astringent and antipyretic in Patna [26]. It was reported that the overconsumption of the leaves for long periods of time may cause health problems, especially for individuals suffering from cardiovascular disease and hypertension due to its high contents of chloride and sodium [27]. In Indonesia, leave infusion/decoction is utilized as an appetite stimulant, antipyretic, digestive aid, deodorant, diarrhea solution, antitussive, and emollient [28,29]. The root decoction is utilized as an astringent and antipyretic [30]. In Thailand, bark and stem decoctions are utilized to treat kidney stones and hemorrhoids, respectively, and leaves are used to cure leucorrhea, inflammation, and lumbago [31]. Leaf tea is widely consumed in Thailand as a health-promoting drink; however, drinking this tea in large amounts increases the feeling of needing to urinate due to its diuretic effect [26]. Additionally, a poultice of the fresh leaf is used for a gangrenous and atonic ulcer [32]. Arial parts are used as an ointment to treat eye diseases and itchy skin. The plant is used for treating acid stomach, dysuria, hemorrhoids, abdominal pain, stomach cramps, scabies, fever, sore muscles, dysentery, menstruation, and rheumatism [33,34,35,36]. In Malaysia, the leaves are prescribed for leucorrhea, dysentery, rheumatism, bad body odor and breath, boils, and ulcers, while roots are used to treat lumbago, fever, headache, and indigestion [37]. The chopped stem bark cigarettes are smoked to relieve sinusitis pain [38]. Indochina, the young shoots and leaves are crushed, mixed with alcohol, and applied in baths to treat scabies, as well as to the back for lumbago and to relieve rheumatic pains [38]. Currently, dried leaves and their extracts have been commercially available in Thailand as herbal tea due to their blood glucose-lowering potential [39]. In Yogyakarta, Indonesia, the leaves are used as a galactagogue to maintain, induce, and augment maternal milk production [40]. It is used orally as anti-fertility for men [20,21]. In Peninsular Malaysia, it is cultivated in gardens for its young shoots, which can be eaten raw. Its leaf decoction is used to counteract fever, and the sap expressed from leaves is used for dysentery [41]. In Dayak Pesaguan tribes, P. indica leaves are utilized to eliminate bad body odor, increase appetite, and overcome digestive disorders [42].

6. Phytochemistry

Various methods for extraction, separation, and characterization were utilized to purify and identify the chemical metabolites from various parts of P. indica. A total of 122 compounds have been separated and characterized. These metabolites mainly include terpenes (Figure 1), sterols, caffeoylquinic acid derivatives, flavonoids, phenolics, lignans, and thiophenes. These compounds, along with their molecular weights and formulae, plant parts and fraction/extract from which they were obtained, as well as the location of plant collection were summarized.
GC-MS analysis of the essential oil obtained from P. indica leaves collected from Sidoarjo and Surabaya in Indonesia using the hydro-distillation technique revealed the existence of 66 components: aldehydes, alcohols, aliphatic unsaturated hydrocarbons (cyclic, aliphatic, heterocyclic, and aromatic), ketones, esters, sulfoxides, and ethers. (10S,11S)-himachala-3-(12)-4-diene (25) (17.13%) was the major volatile constituent [43]. Ruan et al. investigated the chemical profiling of P. indica aerial parts using OC (orthogonal chromatography)-MS integrated normal-phase SiO2 column with reverse-phase BEH C18 column, revealing the identification of 114 metabolites: sesquiterpenes, monoterpenes, sulphated flavonoids, flavonoids, lignans, thiophenes, caffeoylquinic acid derivatives, and other phenolics. By comparing with standard references, 67 metabolites were identified, whereas 47 compounds were speculated based on MS/MS fragmentation, 10 of them were new metabolites [44]. On the other hand, the LC-MS of the leaves EtOH extract demonstrated major metabolites, including campesteryl ferulate (33), quercetin (44), 6-hydroxykaempferol 7-glucoside (51), 8-hydroxyluteolin 8-glucoside (57), apigenin 7-(2″,3″diacetylglucoside) (54), and trans-trismethoxy resveratrol-d4 (97) [45] (Table 1). It is noteworthy that P. indica cultivation in coastal saline land would improve its medicinal quality and increase the caffeoylquinic acid derivatives concentration [46].

7. Biological Activities

The traditional practices and knowledge on P. indica utilization rely exclusively on the observations and practical experience that are passed on verbally from one generation to the next with little documented evidence. Therefore, many studies have been carried out to understand the contemporary relevance of its traditional uses based on biological evaluation (Table 2).

7.1. Anti-Inflammatory Activity

Pulchea indica was assessed for anti-inflammatory activity using various models. The plant showed promising potential, which supported the pharmacological basis of its uses as a traditional herbal medicine for treating inflammation. P. indica roots’ MeOH fraction of the defatted chloroform extract was assessed for its anti-inflammatory capacity utilizing several inflammation models. The extract possessed remarkable inhibitory potential towards histamine-, carrageenin-, serotonin-, sodium urate, and hyaluronidase-induced pedal inflammation. It prohibited cotton pellet- and carrageenin-induced granuloma formation, adjuvant-induced polyarthritis, and turpentine-produced joint edema. Further, it prohibited leucocyte migration and protein exudation. This revealed the extract’s effectiveness in the chronic, proliferative, and exudative stages of inflammation [36]. Sen et al. reported that the root MeOH fraction showed significant anti-inflammatory capacity towards PAF (platelet activation factor), compound 48/80, and AA (arachidonic acid)-produced paw edema with noticeable suppression of spontaneous, as well as compound 48/80-induced histamine release from mast cells [33]. Additionally, it had prominent protective potential towards alcohol-, indomethacin-, and indomethacin-alcohol-caused ulceration, as evident by the significant decrease in acidity and gastric volume in pylorus ligated rats. Additionally, it exhibited a significant protective effect on the gastric mucosa [32]. It was suggested that its dual antiulcer and anti-inflammatory activity could be due to the 5-lipoxygenase inhibition pathway. Another study by Sen et al. demonstrated that the methanol extract of roots (doses 150 and 300 mg/kg) exhibited significant anti-inflammatory potential toward paw edema induced by glucose oxidase [63].
Table 2. Biological activities results of extracts and/or faction of P. indica.
Table 2. Biological activities results of extracts and/or faction of P. indica.
Plant Organ/Tested Extract/FractionActivityModelResultsRef.
Fraction/Extract
(Tested Parameter, Dose)
Control
Root/MeOH fraction of the defatted chloroform extractAnti-inflammatoryCarrageenin-induced paw edema/Charles Foster rats0.103 mL (Paw volume, 100 mg/kg)Phenylbutazone 0.204 mL (Paw volume)[36]
0.046 mL (Paw volume, 300 mg/kg)
75.2 (%Inhibition, 100 mg/kg)Phenylbutazone 50.9 (%Inhibition)[36]
88.9 (%Inhibition, 300 mg/kg)
Carrageenin-induced paw edema/adrenalectomized rats0.113 mL (Paw volume, 300 mg/kg)Phenylbutazone 0.31 mL (Paw volume) [36]
80.7 (%Inhibition, 300 mg/kg)Phenylbutazone 45.4 (%Inhibition)[36]
Mediator-induced edema/Histamine0.030 mL (Paw volume, 300 mg/kg)Control* 0.313 mL (Paw volume)[36]
Mediator-induced edema/Histamine0.180 mL (Paw volume, 300 mg/kg)Control* 0.403 mL (Paw volume)[36]
Mediator-induced edema/Histamine0.030 mL (Paw volume, 300 mg/kg)Control* 0.353 mL (Paw volume)[36]
Carrageenin-induced
pleurisy/Charles Foster rats
46.9 mg (Rear paw weight, 100 mg/kg)Phenylbutazone 24.7 mg (Rear paw weight)[36]
27.4 mg (Rear paw weight, 300 mg/kg) [36]
Cotton pellet-induced granuloma/Charles Foster rats26.2 g (Granuloma weight, 100 mg/kg)Phenylbutazone 15.4 g (Granuloma weight)[36]
21.8 g (Granuloma weight, 300 mg/kg) [36]
41.9 (%Inhibition, 100 mg/kg)Phenylbutazone 65.7 (%Inhibition)[36]
51.8 (%Inhibition, 300 mg/kg) [36]
Carrageenin-induced granuloma/Charles Foster rats1.22 g (Granuloma weight, 100 mg/kg)Phenylbutazone 2.65 g (Granuloma weight)[36]
1.00 g (Granuloma weight, 300 mg/kg) [36]
63.1 (%Inhibition, 100 mg/kg)Phenylbutazone 19.9 (%Inhibition)[36]
69.8 (%Inhibition, 100 mg/kg) [36]
Sodium urate-inducededema/Charles Foster rats0.40 (Rear paw weight, 100 mg/kg)Control* 0.68 (Rear paw weight)
[36]
0.22 (Rear paw weight, 300 mg/kg) [36]
Root/MeOH extractAntiulcerIndomethacin-induced gastric ulcer/Charles Foster rats2.50 mm (Ulcer lesion index, 100 mg/kg)Control* 4.7 1 (Ulcer lesion index)[32]
0.83 (Ulcer lesion index, 300 mg/kg) [32]
46.92 (%Inhibition, 100 mg/kg) [32]
82.37 (%Inhibition, 300 mg/kg)
Alcohol-induced gastric ulcer/Charles Foster rats1.33 (Ulcer lesion index, 100 mg/kg)Control* 4.1 (Ulcer lesion index)[32]
1.16 (Ulcer lesion index, 300 mg/kg) [32]
67.56 (%Inhibition, 100 mg/kg) [32]
71.70 (%Inhibition, 300 mg/kg) [32]
Alcohol-indomethacin-induced gastric ulcer/Charles Foster rats5.72 (Ulcer lesion index, 100 mg/kg)
Control* 19.73 (Ulcer lesion index)[32]
4.36 (Ulcer lesion index, 300 mg/kg) [32]
71.0 (%Inhibition, 100 mg/kg) [32]
78.0 (%Inhibition, 300 mg/kg) [32]
Gastric secretion following pyloric ligation/Charles Foster rats13.0 mEq acid (HCl)/1/h (Total acidity, 100 mg/kg)*Control 16.25 mEq acid (HCl)/1/h[32]
7.5 mEq acid (HCl)/1/h (Total acidity, 300 mg/kg) [32]
9.00 mEq acid (HCl)/1/h (Total acidity, 100 mg/kg)*Control 9.5 mEq acid (HCl)/1/h[32]
5.16 mEq acid (HCl)/1/h (Total acidity, 300 mg/kg) [32]
10.3 mg Tyrosin/mL (Peptic activity, 100 mg/kg)Control* 11.03 Tyrosin/mL (Peptic activity)[32]
9.6 Tyrosin/mL (Peptic activity, 300 mg/kg) [32]
PAF-induced gastric ulcer/Charles Foster rats15.0 mm (Ulcer lesion index, 100 mg/kg)BW 755C 8.33 mm (Ulcer lesion index)[64]
11.66 mm (Ulcer lesion index, 300 mg/kg)BW 755C 8.33 mm (Ulcer lesion index)[64]
35.6 (%Inhibition, 100 mg/kg)BW 755C 64.2 (%Inhibition) [64]
50.2 (%Inhibition, 300 mg/kg)BW 755C 64.2 (%Inhibition)[64]
PAF-induced hematological change/Charles Foster rats41.2 (Haematocrit % change, 300 mg/kg)BW 755C 37.6 (Haematocrit% change)[64]
20.4 (RBC % change)BW 755C 14.3 (RBC % change)[64]
25.2 (Hemoglobin % change)BW 755C 21.2 (Hemoglobin
%change)
[64]
15.8 (WBC % change)BW 755C 16.6 (WBC % change)[64]
Croton oil-induced ear edema/Swiss A mice3.3 mg (Weight of 5 mm ear-punch, 100 mg/kg)*Control 19.6 mg (Weight of 5 mm ear-punch[65]
6.8 mg (Weight of 5 mm ear-punch, 300 mg/kg)-[65]
7.2 mg (Weight of 5 mm ear-punch, 250 µg/ear)-[65]
15.4 mg (Weight of 5 mm ear-punch, 500 µg/ear)-[65]
Root/EtOH extractAntioxidantHydroxyl (OH) radical-scavenging10.77 µg/mL (OH radicals IC50)Vitamin E 33.2 µg/mL (OH radicals, IC50)[63]
CCl4-induced lipid peroxidation/Swiss albino mice54.5 (%Inhibition, 300 µg/mL)vitamin E 46.28 (%Inhibition)[63]
Hydrogen peroxide (H2O2)-scavenging/Charles Foster rats65.2 (% Lysis of erythrocytes, 10 µg/mL)BW 755C 86.9 (% Lysis of erythrocytes)
Phenidone 76.8 (% Lysis of erythrocytes)
[63]
Root/EtOH extractCytotoxicNPC-TW 01 cells/WST-1 colorimetric assay14.07 (%Migration, 80 µg/mL)-[66]
5.65 (%Relative migration rate, 80 µg/mL)-[66]
24.57 (%Colony forming efficiency, 50 µg/mL)-[66]
108.5 µg/mL (24 h) (Growth inhibition 50%)-[66]
NPC-TW04/WST-1 colorimetric assay7.76 (%Migration, 60 µg/mL)-[66]
93.2 (24 h) (Growth inhibition 50%)-[66]
3.47 (%Relative migration rate, 60 µg/mL)-[66]
Leaves/EtOAc fractionAnti-inflammatoryEPP-induced ear edema/Male Sprague Dawley rats8.30 µm (15 min) (ED, 3 mg/ear)PB 3.3 µm (15 min)[67]
18.30 µm (30 min) (ED, 3 mg/ear)PB 25.0 µm (30 min)
45 µm (60 min) (ED, 3 mg/ear)PB 43.3 µm (60 min)
65 µm (120 min) (ED, 3 mg/ear)PB 48.3 µm (120 min)
93.24 (15 min) (%Inhibition)PB 97.3 (15 min) (%Inhibition)[67]
89 (30 min) (%Inhibition)PB 85 (30 min) (%Inhibition)
73 (60 min) (%Inhibition)PB 74 (60 min) (%Inhibition)
54.65 (120 min) (%Inhibition)PB 66.28 (120 min) (%Inhibition)
Carrageenan-induced hind paw edema/Male Sprague Dawley rats0.12 mL (1 h) (EV, 600 mg/kg)DC 0.16 (1 h) (EV)[67]
0.20 mL (3 h) (EV, 600 mg/kg)DC 0.21 (3 h) (EV)
66.2 (1 h) (%Inhibition, 600 mg/kg)DC 55.09 (1 h) (%Inhibition)[67]
56.74 (3 h) (%Inhibition, 600 mg/kg)DC 54.61 (3 h) (%Inhibition)
Leaves/EtOH extractAnti-inflammatoryCarrageenan-induced hind paw edema/Male Sprague Dawley rats5.75 (180 min) (%Oedema, 300 mg/kg)Indo 3.46 (180 min) (%Oedema)[37]
87.9 (%Inhibition, 300 mg/kg)92.7 Indo (180 min) (%Inhibition)[37]
AnalgesicAcetic acid-induced writhing/Adult Balb/calbino mice20.3 (15 min) (Mean of writhings)19.0 (15 min) (Mean of writhings)[37]
60.7 (15 min) (%Inhibition)63.2 (15 min) (%Inhibition)[37]
CytotoxicityEA.hy926 cells/MTT assay54.9 (%Cell viability, 100 µg/mL)-[68]
27.5 (%Cell viability, 200 µg/mL)-[68]
26.4 (%Cell viability, 400 µg/mL)-[68]
Lipase inhibitory3T3-L1 adipocytes/Pancreatic lipase assay1708.35 µg/mL (IC50, 250–1000 µg/mL)Orlistat 68.23 µg/mL (IC50) [69]
Antihyperlipidemic3T3-L1 adipocytes/Lipid accumulation76.87 (%Inhibition, 750 µg/mL)-[69]
71.93 (%Inhibition, 1000 µg/mL)-[69]
Leaves/H2O extractAnti-dyslipidemiaPrediabetic patients/Clinical trials109.22 mg/dL (TG, 1.5 g/once/12 weeks)Placebo 145.56 mg/dL (TG)[70]
122.20 mg/dL (LDL-C, 1.5 g/once/12 weeks)Placebo 142.07 mg/dL (LDL-C) [70]
57.56 mg/dL (HDL-C, 1.5 g/once/12 weeks)Placebo 46.44 mg/dL (HDL-C) [70]
Fibroblast hyperproliferation inhibitionFlowcytometry analysis/
Fibroblast cultures
1.3 (%Fibroblast density, 20 µmol/L)Control 1.5 (%Fibroblast density)[5]
1.0 (%Fibroblast density, 40 µmol/L)-[5]
0.7 (%Fibroblast density, 80 µmol/L)-[5]
Leaves/MeOH extractAntidiabeticNormoglycemic/Wistar albino rats72.67 mg/dL (24 h) (Plasma glucose, 200 mg/kg)Glibenclamide 73.67 mg/dL (24 h) (Plasma glucose)[71]
71.67 mg/dL (24 h) (Plasma glucose, 400 mg/kg) [71]
STZ-induced hyperglycemia/Wistar albino rats190.0 mg/dL (24 h) (Plasma glucose, 200 mg/kg)Glibenclamide 168.0 mg/dL (24 h) (Plasma glucose)[71]
172.83 mg/dL (24 h) (Plasma glucose, 400 mg/kg)-[71]
Milk productionTotal milk yield/lactating rats6.82 g/L/day (23 h) (Milk yield, 750 mg/kg) Domperidone 7.17 g/L/day
(23 h) (Milk yield)
[72]
Growth hormone promotion 1963.25 pg/µL (Serum growth hormone level, 750 mg/kg)Domperidone 409.46 pg/µL (Serum growth hormone level)[72]
Weight gainBodyweight gain/Wistar rat dams8.44 (%Dams body weight gain)Domperidone 0.66 (%Dams body weight gain)[72]
Leaves/Et OH extractAntioxidantDPPH scavenging/DPPH assay96.4 µmol TE/g fw (%DPPH scavenging, 100 mg/mL)-
Quercetin 6.70 µg/mL (IC50, DPPH scavenging)
[28]
24.45 µg/mL (IC50, DPPH scavenging)[41]
DPPH scavenging/DPPH assay42.24 µg/mL (IC50, DPPH scavenging)α-Tocopherol 35.57 µg/mL (IC50, DPPH scavenging)[73]
ABTS scavenging/TEAC method3.75 µmol TE/g fw, ABTS scavenging-[28]
Ferric-reducing power/FRAP assay81.1 (µmol TE/g fw, Ferric-reducing-[28]
Inhibition of lipid oxidation/TBA method98.5 (%Inhibition)-[28]
β-Carotene-linoleic scavenging activity/β-carotene bleaching method59.8 (%Antioxidant activity)BHA 93.5 (% Antioxidant activity)[73]
Control*: inflamed control; Phenylbutazone: PB; Diclofenac: DC; Indomethacin: Indo; ED: edema thickness; EV: edema volume; AA: antioxidant activity.
Sen et al. reported that pretreatment with root MeOH extract remarkably attenuated severe gastric mucosal vaso-congestion, hematological alteration, inflammation, necrosis, and ulceration induced by PAF administration in rats [64]. It significantly suppressed edema and decreased the hydroperoxides formation induced by croton oil in mice. It also significantly reduced lipid peroxidation induced by CC14/NADPH and Fe3+/ADP/NADPH systems [64]. The significant anti-inflammatory activity of the EtOH extract was attributed to taraxasterol acetate that was separated from the hexane fraction (Figure 2 and Figure 3) [36].
The root MeOH extract had significant inhibitory potential on carrageenin-caused edema in rats. It remarkably prohibited granuloma formation induced by a cotton pellet in rats. It reduced the paw diameter in formaldehyde-induced arthritic rats. Additionally, it exhibited antipyretic potential on the pyrexia induced by yeast in rats [74].
The EtOAc fraction of ethanol extract of leaves exhibited potent inhibition of NO (nitric oxide) production induced by LPS (lipopolysaccharide) in RAW 264.7 macrophages and prohibited PGE2 release. It decreased iNOS (inducible nitric oxide synthase) mRNA and protein expression through suppressed iNOS promoter activity and nuclear translocation of subunit p65 of nuclear factor-kB. Moreover, it possessed anti-inflammatory potential in the inflammation acute phase, as observed in EPP (ethylphenylpropiolate)-induced ear edema and carrageenan-induced paw edema in rats. Therefore, its anti-inflammatory effectiveness was proposed to be due to iNOS suppression and NO production inhibition, which mediated via the suppression of NF-kB activation [67].
The leaf EtOH extract (dose 300 mg/kg, orally) demonstrated a remarkable inhibition of edema induced by carrageenan similar to indomethacin (dose 10 mg/kg). It noticeably prohibited (doses 10, 30, 100, and 300 mg/kg, ip) the abdominal constriction induced with acetic acid (0.6% v/v) in mice with a maximal effect at a dose of 300 mg/kg (60.7%), which was comparable to that of indomethacin (63.2%). These effects were attributed to the inhibition of PG synthesis [37]. Additionally, the EtOH extract possessed anti-inflammation potential towards TNF-α-boosted vascular inflammation on EAhy926 (human vascular endothelial) cells by reducing ROS production and lessening the ICAM-1 (intercellular adhesion molecule-1) and VCAM-1 (vascular cell adhesion molecule-1), which was partly mediated via the HO-1 up-regulation [68].
Additionally, 3,4,5-tri-O-caffeoylquinic acid (41), 1,3,4,5-tetra-O-caffeoylquinic acid (43), and quercetin (44) identified from the EtOAc fraction of the leaves were assayed for their collagenase and metalloproteinase (MMP-2 and -9) inhibitory activities in the fluorometric assay. Compounds 41 and 43 showed remarkable collagenase inhibitor potential (IC50 values 1.5 and 6.3 μM, respectively) compared to phosphoramidon (IC50 7.4 µM); however, compound 44 had moderate potential (IC50 16.9 µM). Additionally, 41 had higher MMPs’ inhibitory activities (IC50 2.5 μM/MMP-2 and 6.4 μM/MMP-9) than 43 (IC5018.4 µM/MMP-2 and 16.8 µM/MMP-9) in comparison to chlorhexidine (IC50 7.3 µM/MMP-2 and 25.2 µM/MMP-9). It was found that the number of caffeoyl groups in the quinic acid moiety and connecting position had a noticeable role in the inhibitory activity [58] (Figure 4 and Figure 5, Table 3).
New thiophenes 108111, along with the known ones 2023, 29, 6269, 74, 85, 98101, 111, 114, 115, 117, 120, and 121, were obtained from 70% ethanol–water (EtOH) extract of P. indica aerial parts. Compounds 68, 74, 101, 108111, 114, 117, and 120 possessed significant inhibitory potential towards NO production boosted by LPS in RAW 264.7 macrophages (concentration 40 µM with %inhibition ranging from 52.1 to 91.1%) compared to dexamethasone (62.2%) [51].

7.2. Anti-Obesity and Anti-Hyperlipidemic Activities

The incidence of dyslipidemia and obesity is currently growing at a dramatic rate throughout the world. Hyperlipidemia and obesity have resulted from energy expenditure/intake energy imbalance that leads to several health complications, including diabetes, stroke, cancer, and ischemic heart diseases [75]. The conventional anti-obesity and anti-hyperlipidemic therapy, such as orlistat and simvastatin, have severe adverse effects and limited effectiveness, often accompanied by weight gain after drug cessation [75,76]. Hence, the focus has been directed to traditional medicine as an alternative treatment for alleviating these diseases that may possess fewer undesirable influences in comparison with the conventional treatment [77].
The leaf H2O extract (concentrations of 750 to 1000 µg/mL) markedly reduced the accumulation of lipids, suppressed adipogenesis, and modified protein, carbohydrate, glycogen, and nucleic acid concentrations in the 3T3-L1 adipocytes. Additionally, it possessed lipase inhibitory potential (concentration 250 to 1000 µg/mL) [69]. In another study, the leaf extract was found to prohibit pancreatic lipase with % inhibition ranging from 11.7% to 18.4% at a concentration ranging from 625 to 1000 ppm compared to orlistat (% inhibition from 26.6 to 36.6%) and epicatechin (% inhibition from 10.7 to 18.6%) at the same concentrations [73]. Therefore, it could be further developed into an herbal supplement for managing obesity or overweight [69]. P. indica tea (herb, 400 and 600 mg/kg, orally) ameliorated hyperglycemia, dyslipidemia, and obesity in high-fat diet-induced (HFD) mice. Moreover, it significantly lowered TG, TC, LDL-C, and perigonadal fat weight in HFD-treated mice; however, it increased HDL-C. This was related to its total phenolic and flavonoid contents (TFC), caffeoylquinic acid derivatives, betacaryophyllene, and gamma-gurjunene (Figure 6, Figure 7, Figure 8, Figure 9, Figure 10 and Figure 11) [70].

7.3. Antidiabetic Activity

Diabetes mellitus (DM) is a complicated metabolic illness that is characterized by the destruction of insulin-secreting β-cells or by insulin resistance, whereby the insulin-target organs are unresponsive to insulin action [78,79]. One of the effective strategies in diabetes management is to decrease postprandial hyperglycemia through the inhibition of α-glucosidase in the digestive system [80,81,82,83]. The available antidiabetic agents of chemical origin cause harmful side effects. Hence, developing safe and effective alternative natural antidiabetic agents is an important era of research.
Nopparat et al. reported that the pretreatment with the leaf EtOH extract (dose 100 mg/kg for 2 weeks) effectively alleviated β-cell injury produced by cytokine in STZ (streptozotocin) mice as it minimized the levels of inflammatory markers IFN-γ (interferon-γ), TNF-α (tumor necrosis factor-α), and IL-1β (interlukin1-β), along with prohibiting caspases; 3, 8, and 9, pSTAT1 phosphorylation (signal transducer and activator of transcription 1), NF-κBp65 (nuclear factor-κBp65), and iNOS. Further, it protected β-cells by boosting cell proliferation and suppressing apoptosis. The blood glucose-lowering potential of the leaf extract was attributed to the antioxidant capacities of the extract’s constituents, particularly resveratrol derivative and quercetin [45]. Additionally, it was noted that the P. indica leaf extracts possessed α-glucosidase and maltase inhibitory capacities that could delay the breakdown of starch into glucose and increase the glucose uptake, thereby lowering the blood glucose level [57,84].
Suriyah et al. carried out an in vitro study to assess the potential of P. indica’s various leaf extracts: n-hexane, CH2Cl2, EtOAc, MeOH, and H2O in stimulating glucose consumption in a human liver CCL-13 cell line model. It was found that P. indica markedly increased glucose consumption of the cells, which revealed that the antidiabetic potential of the extract was due to the stimulation of glucose uptake in the liver cells [85]. Additionally, Widyawati et al. established that the leaf H2O extract had higher blood glucose-reducing capability (56.37%) than glibenclamide (49.59%) and other extracts (EtOAc 19.11% and MeOH 24.27%) in rats [39]. Conversely, the leaf MeOH extract (200 and 400 mg/kg, per os/orally (p.o.)) was found to reduce blood glucose levels in both STZ-mediated diabetic (36.1 and 41.87%) and normal rats (35.12 and 36.1%) [71].
α-Glucosidase inhibitory assay-directed fractionation of the leaves’ EtOAc fraction yielded caffeoylquinic acid derivatives, 38 and 4043, which were isolated using SiO2, RP-18 CC, and HPLC and elucidated by MS and NMR analyses. Compound 42 had the highest α-glucosidase inhibitory effectiveness among the separated caffeoylquinic acid derivatives (IC50 2.0 µM) compared to acarbose (IC50 0.5 µM), while the other compounds displayed moderate to weak activity compared to the activity of acarbose. It was found that increasing numbers of caffeoyl groups attached to the quinic acid moiety and methyl esterification of the carboxylic group in quinic acid promoted the α-glucosidase inhibitory capacity [57].

7.4. Insecticidal and Herbicidal Activity

Weeds and pests that affect the plants are very harmful and represent a problem because they can prohibit the growth of cultivated plants and decrease the yield of various crops [86]. Weeds and pests can be controlled utilizing synthetic pesticides and herbicides. Nevertheless, these synthetic agents cause diverse side effects due to the toxic residual active matter that remains in the soil. Additionally, these synthetic agents can affect non-target organisms such as plants or animals. Other undesirable effects are pest resurgence and pest resistance, as well as environmental pollution [87]. The traditional methods of pest and weed control using plants or their derived metabolites are commonly used because they are safe and eco-friendly.
Yuliani and Rahayu reported that biopesticides derived from P. indica leaf extract caused optimal mortality (81.90% at concentration 12%) of Spodoptera litura larva with LC80 9.88% and LC50 4.00%. It also markedly prohibited the seed germination of the plant weed, Amaranthus spinosus [88]. Therefore, the leaf extract could be a potential bioinsecticide and bioherbicide.

7.5. Cytotoxicity Activity

Cancer is one of the most prevalent health problems with wide epidemiology and great mortality [89]. Its current remedies include chemotherapy, surgery, and radiation therapy [90]. These treatment strategies can result in a high recurrence rate in the case of surgical treatment, whereas chemo- and radiation therapy are lacking specificity and may harm normal cells, causing countless side effects [91].
It was found that the root EtOH extract markedly prohibited NPC (nasopharyngeal carcinoma)-TW04 and NPC-TW01 cell viability and migrations, whereas the NPC-TW04 cells had more susceptibility [92]. It noticeably increased NPC apoptosis that was triggered by up-regulation of pro-apoptotic protein Bax and suppression of anti-apoptotic protein Bcl-2 expression. Therefore, it induced the NPC cells’ apoptosis by activating p53 and regulating apoptosis-related proteins. Hence, it could be further assessed as an alternative chemotherapeutic agent for NPC [66,92]. It also prohibited K562 (human leukemia cells) proliferation, induced cell cycle arrests in the G2/M phase, and caused apoptosis [93].
The leaf and root aqueous extracts demonstrated prominent anti-migratory and anti-proliferative potential on HeLa and GBM8401 cells. They induced critical tumor suppressor molecules: phosphorylated-p53 and p21 and down-regulated phosphorylated-AKT. This effect was attributed to tannins, flavonoids, and the total phenolic contents of these extracts [91].
Moreover, the root hexane fraction exhibited a potent suppressive potential versus GBM cell growth, apparently by inducing G0/G1-phase cell cycle arrest. Further, it enhanced autophagy by increasing the formation of AVOs (acidic vesicular organelles), LC3-II expression (microtubule-associated light chain 3-II), and p38 and JNK phosphorylation [94].
Goswami et al. prepared SLN (solid lipid nanoparticle), utilizing compound 102 purified from P. indica, which displayed anticancer potential on EAC (Ehrlich ascites carcinoma) cells in Swiss albino mice. It prolonged the lifespan of cancer-bearing mice and reduced tumor volume compared to PITC-2-treated groups, and it promoted the apoptosis of tumor cells [95] ((Figure 12).
The human liver cytochrome P450 (CYP)-2A13 and -2A6 enzymes demonstrated a crucial function in the activation of tobacco-specific nitrosamine carcinogens and nicotine metabolism. Their prohibition could represent a strategy for smoking abstinence and decreasing the risk of lung cancer and respiratory complaints. Flavonoids 44, 53, 55, 56, 102, 106, and 107 isolated from the EtOAc fraction of the aerial parts by SiO2 CC and HPLC were evaluated for their inhibition capacity towards CYP2A6- and CYP2A13-mediated coumarin 7-hydroxylation in enzymatic reconstitution spectrofluorometric assay (Figure 12 and Figure 13). Flavonoids were the most effective metabolites at inhibiting CYP2A6 and CYP2A13 (IC50 ranging from 0.90 to 2.66 µM for CYP2A6 and from 0.05 to 0.80 µM for CYP2A13), followed by thiophenes (IC50 values 3.90 to 6.43 for CYP2A6 and 2.40 µM to 6.18 µM for CYP2A13) compared to methoxsalen (IC50 0.19 and 0.43 µM, respectively). Thiophenes exhibited irreversible inhibition towards both enzymes with inactivation kinetic KI values of 0.11–1.01 and 0.67–0.97 µM, respectively). These metabolites could have application in smoking stoppage and lessened risks of lung cancer and respiratory illnesses [59] (Figure 13).
Further, 102 purified from the EtOAc soluble fraction of the root extract of tissue cultured P. indica induced apoptosis and suppressed sarcoma-180 cancer cell proliferation, alongside G1 cell cycle arrest through cyclin D1, Bcl-2, and Ki-67 down-regulation, revealing its apoptotic and antiproliferative capacities versus sarcoma-180 solid tumor in vivo in mice [61].

7.6. Venom Neutralizing Activity

Death and injury due to snakebite are important sociomedical concerns in tropical countries [96,97]. In some hospitals, the antiserum is the only available antidote for a snakebite, which does not show complete protection against venom-induced necrosis, nephrotoxicity, or hemorrhage, and often produces adverse hypersensitivity reactions [98,99]. People in rural areas tend to choose herbal medicine from Traditional Healers because of limited or lacking hospitals [100].
P. indica root methanolic extract was reported to have the potential to neutralize viper venom and counter venom-induced hemorrhagic and lethality influences [97]. Further, 29 and 31 purified from the root extract were able to neutralize cobra and viper venom and antagonize cobra venom-induced cardiotoxicity, lethality, respiratory changes, and neurotoxicity, as well as potentiating the action of equine polyvalent snake venom antiserum in mice [96]. Therefore, these studies give scientific evidence for the folk use of the plant versus snakebite.

7.7. Hepatoprotective and Neurological Activities

Some reported findings validated the ethnomedicinal uses of P. indica for managing diabetic liver injury. P. indica leaf EtOH extract (doses 50 and 100 mg/kg for 2 weeks) ameliorated hyperglycemia-induced liver damage in STZ mice through the modulation of inflammatory responses and oxidative stress by inhibiting IL-6, TNF-α, TGF-β1, NF-κB p65, and PKC (protein kinase C), resulting in a reduction in hepatic apoptosis and improvement of hepatic architecture [101]. Further, the roots MeOH fraction displayed remarkable protective potential toward CCl4-induced hepatotoxicity in rats and mice. It reduced the elevated serum enzyme levels and bilirubin content. It also demonstrated a significant reduction in the prolonged pentobarbitone-produced sleeping time and plasma prothrombin time as well as a reduction in the increased bromo-sulphthalein retention produced by CCl4 treatment [32].
Another study reported that the root extract (doses 50–100 mg/kg, p.o.) remarkably prolonged pentobarbital sleep and reduced locomotor activity in isolated mice but not in group-housed mice. However, at high doses (dose 400 mg/kg, p.o.), it reduced locomotor activity and prolonged pentobarbital sleep in group-housed mice in comparison to diazepam (doses 0.1 and 0.5 mg/kg), which noticeably prolonged pentobarbital sleep in both isolated and group-housed mice. The effect of root extract and diazepam on pentobarbital sleep was significantly attenuated by flumazenil (1 mg/kg, i.v.). It suppressed the isolated mice’s aggressive behavior induced by social isolation. It was suggested that the GABAergic system was partly implicated in pentobarbital-caused sleep [34]. In another study, P. indica root extract reduced spontaneous motility, altered behavior patterns, prolonged pentobarbitone-induced sleep, and suppressed aggressive behavior patterns. Therefore, it possessed a potent CNS depressing potential [102].

7.8. Antifertility Activity

The population growth has raised remarkable economic and social concerns. The current strategies for controlling fertility have been debated for a long time [103]. Recently, male fertility regulation has been widely studied, aiming at developing a new male contraceptive for further inclusion of men in family planning [104]. Based on traditional methods of birth rates control, natural products represent a promising source for developing a male contraceptive. Many plants and their constituents have been investigated for their antifertility potential [105]. Interestingly, P. indica has been used orally in men as an antifertility alternative medicine [20,21,106].
Spermiogenesis is the last phase of spermatogenesis in which spermatids are converted to spermatozoa, which play a substantial function in the fertilization process [107]. It was reported that the leaf extract (doses 1.4, 2.8, and 5.6 mg/kg for 10 days) caused a decrease in the number of spermatid cells in male mice when the dose increased [106]. It was found that the treatment of white male adult rats with leaf tannin reduced the number of spermatozoa and spermatids [20,21]. Further, tannin remarkably reduced glutamic acid levels with an average decrease ranging from 494.43 to 341.40 mg/100 g in the semen of male white rats in comparison to the control group (547.48 mg/100 g glutamic acid level) [21,108]. Glutamic acid is needed for spermatozoa metabolism, and decreasing its level leads to the reduction in spermatozoa motility [109].

7.9. Wound-Healing Activity

Wound healing is a complex process that involves a variety of molecular, cellular, and biochemical reactions, which play a role in hemostasis, proliferation, inflammation, re-epithelialization, growth, and remodeling phases [110]. The healing ability is greatly influenced by systemic and local factors such as adequate nutrition, depth and location of the wound, infection, oxygenation, stress, age, obesity, diabetes, medications, and smoking [111].
In Thai traditional medicines, P. indica leaf was used as stringent to heal wounds and ulcers [112]. Fibroblasts represent the major type of connective tissue cells that produce an extracellular matrix accountable for maintaining tissue structural integrity [113,114]. They have a substantial function in the wound-healing proliferative phase, causing deposition of extracellular matrix [115]. Over proliferation of fibroblast during wound healing leads to the production of abnormally large amounts of collagens and extracellular matrix, resulting in scar formation and functional impairment that may further trigger permanent fibrosis [116]. The EtOH extract of leaves that was rich in flavonoids (19.44 mg/gram) exhibited remarkable antioxidant potential (IC50 21.53 μg/mL) and had no effect on fibroblast 3T3 BALB C viability (IC50 311.776 μg/mL) in the prestoblue cytotoxicity test. This supported the use of leaf extract as a nutrient to accelerate wound healing in the oral cavity injury [112]. Buranasukhon et al. assessed the influence of P. indica leaf ethanol extract and nanoparticles prepared from the leaf extract on enhancing oral wound healing using a human oral squamous carcinoma cell line. It was found that the NPs possessed wound-healing potential (concentration 62.5 µg/mL), as evident by increased cell survival and migration, as well as increased the extract’s colloidal stability in the oral spray formulation. Therefore, P. indica leaf extract NPs could relieve oral mucositis [113].
It was reported that the leaf extract (concentration 80 µmol/L) prevented the fibroblasts’ hyperproliferation by its major flavonoid quercetin, which prohibited collagen synthesis by interfering with the biosynthesis of procollagen (collagen precursor molecule) [5]. Increasing the synthesis of collagen is a crucial problem, particularly in the fibrotic tissue repair response due to chronic inflammation/cellular damage [117].

7.10. Anti-Hemorrhoidal Activity

Hemorrhoids are the major reason for rectum bleeding. They can be treated with nonsurgical or surgical treatments. Generally, initial hemorrhoid therapy is a nonsurgical treatment, which includes dietary modifications, medical management, and behavioral therapies [118].
The leaves of P. indica are traditionally utilized for treating hemorrhoids. This was supported by a study performed by Senvorasinh et al. They reported that the oral administration of hot P. indica tea H2O extract remarkably attenuated (dose 50 mg/kg/day) the rectal damage and hemorrhoids induced by croton oil in rats, as evident by no noticeable reduction in rectum and spleen weights. Conversely, it had no effect on gastrointestinal movement in mice, indicating it did not reduce constipation [119].

7.11. Antimicrobial Activity and Pharmaceutical Preparations

The leaf extract prohibited the growth of Streptococcus mutans, the causative organism of dental caries (MIC 10%), in the disk diffusion assay [120,121]. Further, the leaf extract toothpaste prevented dental caries in rats based on Rontgen examination results. Additionally, it reduced the virulence of mouth bacteria that initiated dental caries. Therefore, P. indica herbal toothpaste could treat caries in rat teeth [122]. P. indica leaf EtOH extract had inhibitory effects on C. albicans (MIC 100 mg/mL) [123]. Further, Alvionida et al. prepared different gel formulae using HPMC (hydroxypropyl methylcellulose) and carbopol 940. It was found that the gel formula with 1% carbopol 940 and 1.5% HPMC possessed antibacterial potential toward P. aeruginosa and S. aureus in the cup-plate diffusion assay [124]. Komala et al. stated that the deodorant rolls with 3 to 5% P. indica leaf extracts exhibited antibacterial potential towards S. epidermidis without causing skin irritation in both women and men. The 3% extract deodorant roll was most effective against S. epidermidis than the other formulae for removing the bad body odor [125], which proved its traditional use to eliminate bad odor. The leaf extract was evaluated for antibacterial potential (concentration 2.5 to 6.5%) towards E. faecalis, P. gingivalis, F. nucleatum, and S. mutans, which are accountable for root canal infections, periodontal disease, and caries. It showed significant growth inhibition towards E. faecalis (inhibition zone diameter (IZD) 12.6 mm), followed by S. mutans (IZD 12.2 mm) and P. gingivalis (IZD 12.2 mm) at a concentration of 6.5%, compared to chlorhexidine (IZDs 10.9, 11.4, and 10.6 mm, conc 2%) [126]. It also prohibited biofilm formation and decreased adhesion of E. faecalis and F. nucleatum in the micro-titter plate and auto-aggregation assays, respectively [127]. Hence, it could be utilized as an alternative to root canal sterilization dressing [127]. It had activity versus S. epidermidis (IZD 21.73 mm) and P. aeruginosa (IZD 21.44 mm) at 1 mg/mL [128]. Sittiwet suggested the possible urinary tract infection treatment potential of the aerial parts aqueous extract through its inhibitory effect on E. coli and Klebsiella pneumoniae [129]. Further, the root MeOH extract of P. indica (doses 0.5 and 1.0 mg/kg body weight) remarkably alleviated S. typhimurium caused typhoid fever in mice in vivo [130]. Fresh stems, roots, and twigs prohibited the growth of S. aureus, P. fluorescens, B. cereus, S. typhimurium, and E. coli, whereas fresh samples had more potent inhibitory potential than dried samples [131]. Farhamzah et al. prepared foot spray from the leaf EtOH extract that possessed antibacterial activity versus B. subtilis, which causes foot odor [132].

7.12. Antioxidant Activity

Oxygen radicals and other oxygen-related species are considered a cause of diverse diseases and health disorders such as degenerative and cardiovascular diseases, cancer, and immune-system and brain dysfunction [133]. The endogenous antioxidant defenses are not effective enough to alleviate or prohibit oxygen-radical-related diseases [134]. Thus, free radicals’ scavengers or exogenous antioxidants are required. The protection against free radicals or oxidizing agents can be enhanced with adequate dietary antioxidants intake.
P. indica fresh leaves are used in many kinds of foods, including soups, salads, and side dishes. Additionally, leaf extracts and dried leaves are commonly consumed as food supplements and tea in Thailand. Several studies confirmed the significant antioxidant potential of various extracts of P. indica that is highly correlated to its high total phenols and flavonoids contents. This current work summarizes these studies.
In 2002, Sen et al. reported that the MeOH fraction of P. indica root extract prohibited superoxide and HO. radical generation, H2O2-produced lysis of erythrocytes, and CCl4-caused lipid peroxidation, in addition to lipoxygenase’s dioxygenase activity in the absence and presence of H2O2 [63]. Additionally, the EtOH extract of the leaves prohibited oxidation of linoleic acid and displayed ferric cyanide, ABTS, and DPPH antioxidant potential [28]. Noridayu et al. mentioned that the leaf MeOH extract had remarkable antioxidant potential (IC50 24.45 µg/mL) in the DPPH assay, compared to quercetin (IC50 6.70 µg/mL); however, the MeOH extract of the stem and hexane extracts of stems and leaves exhibited moderate to weak DPPH radical-scavenging capacity with IC50 ranging from 83.74 to 401.68 µg/mL [41]. Abdul Rahman et al. further reported the DPPH and β-carotene-linoleic scavenging potential of the P. indica leaf EtOH extract (IC50 42.24 and 59.8 µg/mL, respectively) [73].
Interestingly, the hot water leaf extract possessed marked DPPH scavenging potential (EC50 23.8 μg/mL) greater than BHT (EC50 44.4 µg/mL) [135]. In 2018, Widyawati et al. evaluated the antioxidant capacity of black tea-Pluchea leaves drink using different proportions (0:100, 25:75, 50:50, 75:25, and 100:0% w/w). It was found that increased black tea proportion remarkably reduced iron ion-reducing power and DPPH free radical scavenging activity, except for 100% black tea proportion [136]. The leaf MeOH extracts potentially scavenged superoxide radical, reduced iron ion, and inhibited linoleic acid-β-carotene bleaching. Conversely, the EtOAc fraction exhibited a remarkable potency to scavenge superoxide radicals, reduce iron ions and TBARS, and chelate iron ions and hemoglobin. Additionally, the EtOAc fraction had an anti-warmed-over flavor (WOF) effect on duck meat, where it prevented the oxidation of linoleic acid to hexanal. Hexanal can result in a warmed-over flavor (WOF) of meat and its products [137]. Further, the antioxidant capacity of leaf extracts from mature leaves in the flowering and pre-flowering stage, as well as juvenile leaf shoots for P. indica samples obtained from various locations in Thailand, was estimated. Regardless of origin, the juvenile leaf shoots displayed potent antioxidant potential [84].
Sirichaiwetchakoon et al. assessed the antioxidant potential of P. indica aqueous tea using DPPH, ABTS, HOCl (hypochlorous acid), and peroxynitrite scavenging assays. Further, its protective potential on copper (Cu2+), AAPH (2,2′-azobis(2-amidinopropane) dihydrochloride), and SIN-1 (3-morpholinosydnonimine hydrochloride)-induced human LDL (low-density lipoproteins) oxidation was evaluated using TBARS assay in comparison to the known commercial green tea (CST). The tea exhibited stronger antioxidant capacity (%DPPH scavenging 51.19%, IC50 245.85 μg/mL at concentrations of 75–300 μg/mL) than CST (%DPPH scavenging 41.46%, IC50 315.41 μg/mL) in the DPPH, NO (nitric oxide), and peroxynitrite scavenging assays. Further, it possessed protective effects such as CST towards Cu2+, AAPH, and SIN-1-produced LDL oxidation that was attributed to its caffeoyl derivatives [138].

7.13. Other Activities

The leaf extract (dose 750 mg/kg, orally) increased growth hormone serum levels, milk production, and body weight in lactating rats [72]. The hexane extracts of stems and leaves exhibited AChE inhibitory potential that was correlated to its terpenoids content [41]. Compound 102 was purified and characterized from the EtOAc fraction of the root MeOH extract by SiO2 CC and spectroscopic analyses. This metabolite possessed marked anti-proliferative potential (MIC 50 µg/mL) and lytic activity on trophozoites within 4 h administration compared with metronidazole that produced complete lysis (concentration 5 µg/mL) within 2 h towards Entamoeba histolytica HM1 in the cell count assay [60].

8. Toxicity and Safety of P. indica

Many studies have been conducted to evaluate the safety of P. indica. The acute toxicity assessment was carried out using different doses.
Pramanik et al. proved that the MeOH extract of the leaves did not display any sign of toxicity in rats at a dose of 3200 mg/kg orally [71]. Further, the toxicity assessment of P. indica tea in randomized clinical trials on prediabetic humans revealed no toxicity to the liver, kidney, and blood, as evident by the lack of any remarkable variation in ALT, BUN, creatinine, CBC, and ALP of the P. indica-received group compared to the placebo [70]. In another study, Sirichaiwetchakoon et al. stated that P. indica tea (dose 600 mg/kg/day) was non-toxic to the kidney, liver, and blood in mice [139]. Nopparat et al. carried out a study to validate the toxicity of P. indica administration on experimental animals. The results revealed no toxic effect on kidney and liver functions, as evident by the lack of any significant variation in serum TC, TG, AST, BUN, creatinine, ALP, albumin, and ALT among all experimental groups, indicating its safety [45]. P. indica leaf H2O extract (concentration 25 to 400 µg/mL) exhibited no cytotoxic effectiveness on RAW 264.7 macrophage cells [135]. Conversely, the leaf EtOH extract did not have a cytotoxic influence (concentration 12.5–50 µg/mL) on EA.hy926 cells, while it reduced cell viability at concentrations ≥ 100 µg/mL in the MTT assay [68]. It was found that P. indica fresh herb H2O extract was safe at a concentration ≤ 1000 µg/mL on 3T3-L1 cells [69]. A sub-chronic toxicity study revealed that the oral administration of water extract was safe for mice (dose 2.6 mg/20 g b.w.); however, EtOAc and MeOH extracts (doses 1.3 and 2.6 mg/20 g b.w.) caused mortality in mice [39]. Thongpraditchote et al. reported that the root extract did not cause any mortality at a dose of up to 5 g/kg orally [34]. Additionally, it did not produce mortality in mice at a dose up to 1100 mg/kg [102].

9. Clinical Traits

Werdani and Widyawati stated that the regular administration of P. indica twice daily (2 g of tea of P. indica with 100 mL hot H2O without sugar for two months) reduced random blood glucose levels and eliminated subjective patient complaints, such as tingling in the extremities, and improved physical fatigue [140]. A randomized clinical trial in 45 prediabetic patients who received P. indica tea (1.5 g, daily for 12 weeks) revealed that the tea alleviated dyslipidemia and hyperglycemia, where it noticeably lowered TG and LDL-C and raised HDL-C. This supports its utilization as herbal medicine for dyslipidemia and hyperglycemia prevention [70].

10. Conclusion and Recommendations

Medicinal plants and their phytoconstituents have been proven to have a significant contribution to the prevention and therapy of several diseases and to have diverse pharmacological properties. Encouragingly, the current work reviewed several promising bioactivities that are induced by a variety of phytoconstituents of P. indica. This plant is widely used by various traditional medicines for treating various diseases, especially diabetes and its related complications. Many studies have been carried out to prove these traditional uses. The plant studied from various regions has terpenoids and caffeoylquinic acid derivatives, as well as high phenolics and flavonoid contents (TFC), which confer most of the characteristic bioactivities of this plant (Figure 14 and Figure 15). The majority has been purified from aerial parts (Figure 16).
It is noteworthy that the reported studies in this work validate the efficacy and safety of P. indica and support its traditional uses for treating various ailments and promoting health and well-being, particularly for people for whom modern health care is not easily accessible. Therefore, reported results could encourage the food supplement manufacturers to develop this plant into a healthy food supplement or medicine for the prevention and treatment of various diseases.
P. indica root and leaf extracts revealed anti-inflammatory potential towards different types of induced inflammations using various models. The suppression of NF-κB, attenuation of antioxidant enzymes, up-regulation of HO-1, and inhibition of PGE2 and 5-LOX are the reported molecular mechanisms for the anti-inflammatory potential of P. indica.
P. indica leaves/tea could have the potential to prevent and treat metabolic syndrome because they possess blood glucose and lipid-lowering effects. The antihyperlipidemic effect was due to the suppression of adipogenesis and lipase inhibition, whereas the antidiabetic effect was due to the protection of β-cell towards inflammatory responses and apoptosis, α-glucosidase and maltase inhibition, and stimulation of glucose uptake. Further, caffeoylquinic acid derivatives of the leaves were proven to have α-glucosidase inhibitory potential that was promoted by increased numbers of caffeoyl groups linked to quinic acid moiety and methyl esterification of quinic acid’s carboxylic group. The discovery of natural anticancer agents is a major concern of scientists. The reviewed plant was established to have cytotoxic potential, which is most often linked to chemoprevention. The plant was found to promote apoptosis and inhibit the migration and proliferation of diverse cancer cell lines. Further studies are warranted to specify the bioactive compound(s) accountable for the induction of these effects and to elucidate the mechanisms of P. indica on in vivo models.
Additionally, the venom-neutralizing potential of P. indica root was attributed to its phytoconstituents: stigmasterol and beta-sitosterol. The plant showed liver injury, hemorrhoids, and wound-healing capacity.
Several studies confirmed the significant antioxidant potential of various extracts of P. indica that is highly correlated to its high total phenols and flavonoids contents. Therefore, the plant may positively contribute to dietary antioxidant intake. Moreover, P. indica could be further developed into functional food and nutraceutical products.
P. indica extracts and their pharmaceutical preparations possessed antibacterial capacity towards the causative organisms of dental caries, bad body odor, and urinary tract infections.
Despite the diverse studies carried out on P. indica, a limited number of studies focus on the isolation of their chemical constituents and evaluation of their activities. Further, the clinical and toxicity studies suggested the safety of this plant for human use. Based on these facts, the authors hope that this review highlights the role of P. indica in various treatments and recommend that further phytochemical and clinical research should be conducted on this traditional medicinal plant for the discovery of safer drugs.

Author Contributions

Conceptualization, S.R.M.I., A.O.N., and G.A.M.; resources, G.A.M., A.A.B., R.M.D., and S.R.M.I.; data curation and funding acquisition: A.A.B., A.O.N., R.M.D., and H.T.B.; Discussion of the contents, G.A.M., R.M.D., and S.R.M.I.; writing—original draft preparation, S.R.M.I., A.O.N., G.A.M., and H.T.B.; writing—review and editing, S.R.M.I., A.A.B., H.T.B., and G.A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AA: Arachidonic acid; AAPH: 2,2′-azobis(2-amidinopropane) dihydrochloride; ABTS: 2,2′-Azino-bis-(3-ethylbenzthiazoline-6-sulphonic acid; ADP: Adenosine di-phosphate; ALP: Alkaline phosphatase; ALT: Alanine transaminase; AVO: Acidic vesicular organelle; Bcl-2: β-Cell lymphoma 2; BUN: Blood urea nitrogen; CBC: Complete blood count; CH2Cl2: Dichloromethane; CHCl3: Chloroform; DPPH: 2,2-Diphenyl-1-picrylhydrazyl; EA.hy926: Human vascular endothelial cells; EAC: Ehrlich ascites carcinoma; CYP 2A13: Cytochrome P450 2A13; CYP 2A6: Cytochrome P450 2A6; EC50: Effective Concentration at 50% of the total effect; EPP: Ethylphenylpropiolate; EV: Edema volume; EtOAc: Ethyl acetate; EtOH: Ethanol; FRAP: Ferric Antioxidant Power; GABA: gamma-Aminobutyric acid GAE/g: Gallic acid equivalent/g: GBM: Human glioblastoma cells; GBM8401: Human glioma cells; GC-MS: Gas chromatography–mass spectrometry; HDL-C: High-density lipoprotein cholesterol; HO-1: Heme oxygenase-1; HFD: High fat diet; HeLa: Cervical carcinoma cells; HOCl: Hypochlorous acid; HPLC: High-pressure liquid chromatography; IC50: concentration of inhibitor at 50% of the total inhibition; ; ICAM-1: Intercellular adhesion molecule-1; IFN-γ: Interferon-γ; IL-1β: Interlukin1-β; iNOS: Inducible nitric oxide synthase; Ki-67: Nuclear protein; LC3-II: Microtubule-associated light chain 3-II; LC-MC: Liquid chromatography–mass spectrometry; LDL: Low-density lipoproteins; LDL-C: Low-density lipoprotein cholesterol; LPS: Lipopolysaccharide; MeOH: Methanol; MIC: The minimum inhibitory concentration; MMP-2: Matrix metalloproteinase-2; MMP-9: Matrix metalloproteinase-9; MTT: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; n-BuOH: NADPH: Nicotinamide adenine dinucleotide phosphate; NMR: Nuclear magnetic resonance; NF-κBp65: Nuclear factor-κBp65; NO: Nitrous oxide; NRC: Nitrite relative concentration; OC: Orthogonal chromatography: PAF: Platelet activation factor; PGE2: Prostaglandin E2; PKC: Protein kinase C; PITC 2: 2-(Prop-1-ynyl)-5(5,6-dihydroxyhexa-1,3-diynyl)-thiophene; pSTAT1: Signal transducer and activator of transcription 1; ROS: Reactive oxygen species; RP-18: Reversed phase C18 silica gel; SIN-1: 3-Morpholinosydnonimine hydrochloride; pSTAT1: Signal transducer and activator of transcription 1; SiO2 CC: Silica gel column chromatography; SLN: Solid lipid nanoparticle; STZ: Streptozotocin; TBA: 2-Thiobarbituric acid; TBARS: Thiobarbituric Acid Reactive Substances; TE/g: Total extract/gram; TEAC: Trolox equivalent antioxidant capacity; TFC: Total flavonoid contents; TG: Triglyceride; TNF-α: Tumor necrosis factor-α; VCAM-1: Vascular cell adhesion molecule-1; WOF: Warmed-over flavor.

References

  1. Kesharwani, R.K.; Misra, K.; Singh, D.B. Perspectives and challenges of tropical medicinal herbs and modern drug discovery in the current scenario. Asian Pac. J. Trop. Med. 2019, 12, 1. [Google Scholar] [CrossRef]
  2. Abdallah, H.M.; Mohamed, G.A.; Ibrahim, S.R.M. Lansium domesticum—A fruit with multi-benefits: Traditional uses, phytochemicals, nutritional value, and bioactivities. Nutrients 2022, 14, 1531. [Google Scholar]
  3. Lautié, E.; Russo, O.; Ducrot, P.; Boutin, J.A. Unraveling Plant Natural Chemical Diversity for Drug Discovery Purposes. Front. Pharmacol. 2020, 11, 397. [Google Scholar] [CrossRef] [PubMed]
  4. Katiyar, C.; Kanjilal, S.; Gupta, A.; Katiyar, S. Drug discovery from plant sources: An integrated approach. Ayu 2012, 33, 10. [Google Scholar] [CrossRef] [PubMed]
  5. Maharani, S.C.; Julianto, I.; Widhiati, S. The role of beluntas (Pluchea indica L.) leaf extract in preventing the occurrence of fibroblasts hyperproliferation: An in vitro preliminary study. Dermatol. Rep. 2019, 11, 8019. [Google Scholar] [CrossRef] [Green Version]
  6. Ibrahim, S.R.; Mohamed, G.A. Litchi chinensis: Medicinal uses, phytochemistry, and pharmacology. J. Ethnopharmacol. 2015, 174, 492–513. [Google Scholar] [CrossRef]
  7. Ibrahim, S.R.M.; Mohamed, G.A.; Khedr, A.; Zayed, M.; El-Kholy, A.A.-E.S. Genus Hylocereus: Beneficial phytochemicals, nutritional importance, and biological relevance-A review. J. Food Biochem. 2018, 42, e12491. [Google Scholar] [CrossRef]
  8. Riaz, U.; Iqbal, S.; Sohail, M.I.; Samreen, T.; Ashraf, M.; Akmal, F.; Siddiqui, A.; Ahmad, I.; Naveed, M.; Khan, N.I.; et al. A Comprehensive Review on Emerging Importance and Economical Potential of Medicinal and Aromatic Plants (MAPs) in Current Scenario. Pak. J. Agric. Res. 2021, 34, 381–392. [Google Scholar] [CrossRef]
  9. Ekor, M. The growing use of herbal medicines: Issues relating to adverse reactions and challenges in monitoring safety. Front. Pharmacol. 2014, 4, 177. [Google Scholar] [CrossRef] [Green Version]
  10. Taylor, J.; Rabe, T.; McGaw, L.; Jäger, A.; Van Staden, J. Towards the scientific validation of traditional medicinal plants. Plant Growth Regul. 2001, 34, 23–37. [Google Scholar] [CrossRef]
  11. Setorki, M. Medicinal herbs with anti-depressant effects. J. Herbmed Pharmacol. 2020, 9, 309–317. [Google Scholar] [CrossRef]
  12. Abraham, J.; Thomas, T.D. Recent Advances in Asteraceae Tissue Culture. In Plant Tissue Culture: Propagation, Conservation and Crop Improvement; Anis, M., Ahmad, N., Eds.; Springer: Singapore, 2016. [Google Scholar] [CrossRef]
  13. Rolnik, A.; Olas, B. The Plants of the Asteraceae Family as Agents in the Protection of Human Health. Int. J. Mol. Sci. 2021, 22, 3009. [Google Scholar] [CrossRef] [PubMed]
  14. Nikolic, M.; Stevovic, S. Family Asteraceae as a sustainable planning tool in phytoremediation and its relevance in urban areas. Urban For. Urban Green. 2015, 14, 782–789. [Google Scholar] [CrossRef]
  15. Anderberg, A.A. Asteraceae. In Cladistics & Classification; Bremer, K., Ed.; Timber Press: Portland, OR, USA, 1994; pp. 292–303. [Google Scholar]
  16. Aggarwal, P. A review on phytochemical and biological investigation of plant genus Pluchea. Indo Am. J. Pharm. Res. 2013, 3, 3373–3392. [Google Scholar]
  17. Wang, J.; Pei, Y.H.; Lin, W.H.; Deng, Z.W.; Qiao, L. Chemical constituents from the stems and leaves of marine mangrove plant Pluchea indica (L.) Less. Shenyang Yaoke Daxue Xuebao 2008, 25, 960–963. [Google Scholar]
  18. Rodsom, T. A Study on the Diuretic Effects of Pluchea Indica in Healthy Subjects and Patients; Mahidol University: Bangkok, Thailand, 1993. [Google Scholar]
  19. Susetyarini, E.; Wahyono, P.; Latifa, R.; Nurrohman, E. The Identification of Morphological and Anatomical Structures of Pluchea indica. J. Phys Conf. Ser. 2020, 1539, 012001. [Google Scholar] [CrossRef]
  20. Susetyarini, E. Jumlah sel spermiogenesis tikus putih yang diberi tanin daun Beluntas (Pluchea indica) sebagai sumber belajar. Proc. Biol. Educ. Conf. 2015, 12, 462–466. [Google Scholar]
  21. Susetyarini, E. The level of glutamic acid in the semen of male white rat (Ratus norwegicus) after being treated with tannin of Pluchea indica. Procedia Chem. 2015, 14, 152–156. [Google Scholar] [CrossRef] [Green Version]
  22. Polsiri, K. Antioxidant Activity of Pluchea Indica Less. Extract After In Vitro Digestion and Absorption by Caco–2 Cell Line. Ph.D. Thesis, Burapha University, Chon Buri, Thailand, 2015. [Google Scholar]
  23. Sudjaroen, Y. Evaluation of ethnobotanical vegetables and herbs in Samut Songkram province. Procedia Eng. 2012, 32, 160–165. [Google Scholar] [CrossRef] [Green Version]
  24. US Department of Agriculture. Handbooks 8–1 to 8–21: Composition of Food Raw, Processed, Prepared; Government Printing Office: Washington, DC, USA, 1972–1991. [Google Scholar]
  25. Neamsuwan, A.; Sengnon, N.; Yingcharoen, K. Ethnobotany of edible plants from mangrove and beach forest in Sating Phra Peninsula, Songkhla province. KKU Sci. J. 2012, 40, 981–991. [Google Scholar]
  26. Pramanik, K.C.; Biswas, R.; Mitra, A.; Bandyopadhyay, D.; Mishra, M.; Chatterjee, T.K. Tissue culture of the plant Pluchea indica (L.) and evaluation of diuretic potential of its leaves. Adv. Tradit. Med. 2007, 7, 197–204. [Google Scholar] [CrossRef]
  27. Shannon, M.; Grieve, C. Tolerance of vegetable crops to salinity. Sci. Hortic. 1998, 78, 5–38. [Google Scholar] [CrossRef]
  28. Andarwulan, N.; Batari, R.; Sandrasari, D.A.; Bolling, B.; Wijaya, C.H. Flavonoid content and antioxidant activity of vegetables from Indonesia. Food Chem. 2010, 121, 1231–1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Andarwulan, N.; Kurniasih, D.; Apriady, R.A.; Rahmat, H.; Roto, A.V.; Bolling, B. Polyphenols, carotenoids, and ascorbic acid in underutilized medicinal vegetables. J. Funct. Foods 2012, 4, 339–347. [Google Scholar] [CrossRef]
  30. Kirtikar, K.R.; Basu, B.D. Indian Medicinal Plants; International Book Distributors: Dehradun, India, 1999; pp. 1344–1345. [Google Scholar]
  31. Ahem, S.A.; Kamel, E.M. Phenolic constituents and biological activity of the genus Pluchea. Pharma Chem. 2014, 5, 109–114. [Google Scholar]
  32. Sen, T.; Basu, A.; Ray, R.N.; Chaudhuri, A.K.N. Hepatoprotective effects of Pluchea indica (L.) extract in experimental acute liver damage in rodents. Phytother. Res. 1993, 7, 352–355. [Google Scholar] [CrossRef]
  33. Sen, T.; Ghosh, T.K.; Chaudhuri, A.K. Studies on the mechanism of anti-inflammatory and anti-ulcer activity of Pluchea indica–probable involvement of 5–lipooxygenase pathway. Life Sci. 1993, 52, 737–743. [Google Scholar] [CrossRef]
  34. Thongpraditchote, S.; Matsumoto, K.; Temsiririrkkul, R.; Tohda, M.; Murakami, Y.; Watanabe, H. Neuropharmacological Actions of Pluchea indica L. Root Extract in Socially Isolated Mice. Biol. Pharm. Bull. 1996, 19, 379–383. [Google Scholar] [CrossRef] [Green Version]
  35. Locher, C.; Burch, M.; Mower, H.; Berestecky, J.; Davis, H.; Van Poel, B.; Lasure, A.; Berghe, D.; Vlietinck, A. Anti-microbial activity and anti-complement activity of extracts obtained from selected Hawaiian medicinal plants. J. Ethnopharmacol. 1995, 49, 23–32. [Google Scholar] [CrossRef]
  36. Sen, T.; Chaudhuri, A. Antiinflammatory evaluation of a Pluchea indica root extract. J. Ethnopharmacol. 1991, 33, 135–141. [Google Scholar] [CrossRef]
  37. Roslida, A.H.; Erazuliana, A.K.; Zuraini, A. Anti-inflammatory and antinociceptive activities of the ethanolic extract of Pluchea indica (L.) Less leaf. Pharmacologyonline 2008, 2, 349–360. [Google Scholar]
  38. Bandaranayake, W. Bioactivities, bioactive compounds and chemical constituents of mangrove plants. Wetl. Ecol. Manag. 2002, 10, 421–452. [Google Scholar] [CrossRef]
  39. Widyawati, P.S.; Budianta, T.D.W.; Gunawan, D.I.; Wongso, R.S. Evaluation antidiabetic activity of various leaf extracts of Pluchea indica L. Int. J. Pharmacog. Phytochem. Res. 2015, 7, 597–603. [Google Scholar]
  40. Sumanth, M.; Narasimharaju, K. Evaluation of galactagogue activity of lactovedic: A polyherbal formulation. Int. J. Green Pharm. 2011, 5, 61–66. [Google Scholar] [CrossRef]
  41. Noridayu, A.R.; Hii, Y.F.; Faridah, A.; Khozirah, S.; Lajis, N. Antioxidant and antiacetylcholinesterase activities of Pluchea indica Less. Int. Food Res. J. 2011, 18, 925–929. [Google Scholar]
  42. Due, R.; Symaswisna, M.R. Ethnobotany of dayak pesaguan medicinal plant and its implementation in the making of biodiversity flash cards. Biol. Study Fac. Teach. Train. Educ. Untan Pontianak 2013, 3, 1–15. [Google Scholar]
  43. Widyawati, P.S.; Wijaya, C.H.; Hardjosworo, P.S.; Sajuthi, D. Volatile compounds of Pluchea indica less and Ocimum basillicum linn essential oil and potency as antioxidant. HAYATI J. Biosci. 2013, 20, 117–126. [Google Scholar] [CrossRef] [Green Version]
  44. Ruan, J.; Yan, J.; Zheng, D.; Sun, F.; Wang, J.; Han, L.; Zhang, Y.; Wang, T. Comprehensive Chemical Profiling in the Ethanol Extract of Pluchea indica Aerial Parts by Liquid Chromatography/Mass Spectrometry Analysis of Its Silica Gel Column Chromatography Fractions. Molecules 2019, 24, 2784. [Google Scholar] [CrossRef] [Green Version]
  45. Nopparat, J.; Nualla–Ong, A.; Phongdara, A. Ethanolic extracts of Pluchea indica (L.) leaf pretreatment attenuates cytokine–induced β–cell apoptosis in multiple low–dose streptozotocin–induced diabetic mice. PLoS ONE 2019, 14, e0212133. [Google Scholar] [CrossRef]
  46. Chewchida, S.; Vongsak, B. Simultaneous HPTLC quantification of three caffeoylquinic acids in Pluchea indica leaves and their commercial products in Thailand. Rev. Bras. Farmacogn. 2019, 29, 179–181. [Google Scholar] [CrossRef]
  47. Uchiyama, T.; Miyase, T.; Ueno, A.; Usmanghani, K. Terpenic glycosides from Pluchea indica. Phytochemistry 1989, 28, 3369–3372. [Google Scholar] [CrossRef]
  48. Uchiyama, T.; Miyase, T.; Ueno, A.; Usmanghani, K. Terpene and lignan glycosides from Pluchea indica. Phytochemistry 1991, 30, 655–657. [Google Scholar] [CrossRef]
  49. Mukhopadhyay, S.; Cordel, G.A. Traditional medicinal plants of Thailand. IV. 3-(2’,3’-diacetoxy-2’-methyl butyryl)-cuauhtemone from Pluchea indica. J. Nat. Prod. 1983, 46, 671–674. [Google Scholar] [CrossRef]
  50. Wang, H.M.; Kao, C.L.; Liu, C.M.; Li, W.J.; Yeh, H.C.; Li, H.T.; Kuo, C.N.; Chen, C.Y. Chemical Constituents of the Roots of Pluchea indica. Chem. Nat. Compd. 2017, 53, 736–737. [Google Scholar] [CrossRef]
  51. Ruan, J.; Li, Z.; Yan, J.; Huang, P.; Yu, H.; Han, L.; Zhang, Y.; Wang, T. Bioactive Constituents from the Aerial Parts of Pluchea indica L. Molecules 2018, 23, 2104. [Google Scholar] [CrossRef] [Green Version]
  52. Qiu, Y.Q.; Qi, S.H.; Zhang, C.; Li, Q.X. Chemical constituents of Pluchea indica (II). Zhongcaoyao 2010, 41, 24–27. [Google Scholar]
  53. Huong, D.T.V.; Giang, P.M.; Sim, H.T.; Chinh, T.T.T. Triterpenoids and Phytosterols Isolated from Pluchea indica L. Leaves. VNU J. Sci. Nat. Sci. Technol. 2019, 35, 106–111. [Google Scholar] [CrossRef]
  54. Huong, D.T.V.; Giang, P.M. Sterol, glycerol ester, and thiophene constituents from the twigs of Pluchea indica L. of Vietnam. VNU J. Sci. Nat. Sci. Technol. 2018, 34, 78–82. [Google Scholar] [CrossRef]
  55. Shukri, M.A.M.; Alan, C.; Noorzuraini, A.R.S. Polyphenols and antioxidant activities of selected traditional vegetables. J. Trop. Agric. Food Sci. 2011, 39, 69–83. [Google Scholar]
  56. Kongkiatpaiboon, S.; Chewchinda, S.; Vongsak, B. Optimization of extraction method and HPLC analysis of six caffeoylquinic acids in Pluchea indica leaves from different provenances in Thailand. Rev. Bras. Farm. 2018, 28, 145–150. [Google Scholar] [CrossRef]
  57. Arsiningtyas, I.S.; Gunawan–Puteri, M.D.; Kato, E.; Kawabata, J. Identification of α–glucosidase inhibitors from the leaves of Pluchea indica (L.) Less., a traditional Indonesian herb: Promotion of natural product use. Nat. Prod. Res. 2014, 28, 1350–1353. [Google Scholar] [CrossRef] [PubMed]
  58. Ohtsuki, T.; Yokosawa, E.; Koyano, T.; Preeprame, S.; Kowithayakorn, T.; Sakai, S.; Toida, T.; Ishibashi, M. Quinic acid esters from Pluchea indica with collagenase, MMP-2 and MMP-9 inhibitory activities. Phytother. Res. 2008, 22, 264–266. [Google Scholar] [CrossRef] [PubMed]
  59. Boonruang, S.; Prakobsri, K.; Pouyfung, P.; Srisook, E.; Prasopthum, A.; Rongnoparut, P.; Sarapusit, S. Inhibition of human cytochromes P450 2A6 and 2A13 by flavonoids, acetylenic thiophenes and sesquiterpene lactones from Pluchea indica and Vernonia cinerea. J. Enzym. Inhib. Med. Chem. 2017, 32, 1136–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Biswas, R.; Dutta, P.; Achari, B.; Bandyopadhyay, D.; Mishra, M.; Pramanik, K.; Chatterjee, T. Isolation of pure compound R/J/3 from Pluchea indica (L.) Less. and its anti-amoebic activities against Entamoeba histolytica. Phytomedicine 2007, 14, 534–537. [Google Scholar] [CrossRef]
  61. Goswami, S.; Debnath, S.; Karan, S.; Chatterjee, T.K. In vivo antitumor activity of phytochemical pitc-2 obtained from tissue cultured plant Pluchea indica on sarcoma-180 solid tumor mice model. Asian J. Pharm. Clin. Res. 2018, 11, 211–218. [Google Scholar] [CrossRef] [Green Version]
  62. Qiu, Y.-Q.; Qi, S.-H.; Zhang, S.; Tian, X.-P.; Xiao, Z.-H.; Li, M.-Y.; Li, Q.-X. Thiophene derivatives from the aerial part of Pluchea indica. Heterocycles 2008, 75, 1757–1764. [Google Scholar]
  63. Sen, T.; Dhara, A.K.; Bhattacharjee, S.; Pal, S.; Chaudhuri, A.K.N. Antioxidant activity of the methanol fraction of Pluchea indica root extract. Phytother. Res. 2002, 16, 331–335. [Google Scholar] [CrossRef]
  64. Sen, T.; Ghosh, T.K.; Bhattacharjee, S.; Nag Chaudhuri, A.K. Action of Pluchea indica methanol extract as a dual inhibitor on PAF–induced paw oedema and gastric damage. Phytother. Res. 1996, 10, 74–76. [Google Scholar] [CrossRef]
  65. Sen, T.; Pal, S.; Izzo, A.A.; Capasso, F.; Nag Chaudhuri, A.K. Studies on the methanolic fraction of Pluchea indica on croton oil–induced mouse ear oedema and lipid peroxidation. Pharm. Sci. 1996, 2, 433–435. [Google Scholar]
  66. Kao, C.-L.; Cho, J.; Lee, Y.-Z.; Cheng, Y.-B.; Chien, C.-Y.; Hwang, C.-F.; Hong, Y.-R.; Tseng, C.-N.; Cho, C.-L. Ethanolic Extracts of Pluchea indica Induce Apoptosis and Antiproliferation Effects in Human Nasopharyngeal Carcinoma Cells. Molecules 2015, 20, 11508–11523. [Google Scholar] [CrossRef] [Green Version]
  67. Buapool, D.; Mongkol, N.; Chantimal, J.; Roytrakul, S.; Srisook, E.; Srisook, K. Molecular mechanism of anti-inflammatory activity of Pluchea indica leaves in macrophages RAW 264.7 and its action in animal models ofinflammation. J. Ethnopharmacol. 2013, 146, 495–504. [Google Scholar] [CrossRef] [PubMed]
  68. Srisook, K.; Jinda, S.; Srisook, E. Anti-inflammatory and Antioxidant Effects of Pluchea indica Leaf Extract in TNF-α-Induced Human Endothelial Cells. Walailak J. Sci. Technol. 2021, 18, 10271. [Google Scholar] [CrossRef]
  69. Sirichaiwetchakoon, K.; Lowe, G.M.; Thumanu, K.; Eumkeb, G. The Effect of Pluchea indica (L.). Tea on Adipogenesis in 3T3-L1 Adipocytes and Lipase Activity. Evid. Based Complement. Altern. Med. 2018, 2018, 4108787–4108813. [Google Scholar] [CrossRef] [PubMed]
  70. Sirichaiwetchakoon, K.; Churproong, S.; Kupittayanant, S.; Eumkeb, G. The Effect of Pluchea indica (L.). Tea on Blood Glucose and Lipid Profile in People with Prediabetes: A Randomized Clinical Trial. J. Altern. Complement. Med. 2021, 27, 669–677. [Google Scholar] [CrossRef] [PubMed]
  71. Pramanik, K.C.; Bhattacharya, P.; Biswas, R.; Bandyopadhyay, D.; Mishra, M.; Chatterjee, T. Hypoglycemic and antihyperglycemic activity of leaf extract of Pluchea indica L. Orient. Pharm. Exp. Med. 2006, 6, 232–236. [Google Scholar]
  72. Syarif, R.A.; Anggorowati, N.; Yuniyanti, M.M.; Wahyuningsih, M.S.H. Ethanolic extract of Pluchea indica L. leaf increases serum growth hormone in lactating rats. Trad. Med. J. 2021, 26, 111–116. [Google Scholar] [CrossRef]
  73. Abdul Rahman, H.; Saari, N.; Abas, F.; Ismail, A.; Mumtaz, M.W.; Abdul Hamid, A. Anti-obesity and antioxidant activities of selected medicinal plants and phytochemical profiling of bioactive compounds. Int. J. Food Prop. 2017, 20, 2616–2629. [Google Scholar] [CrossRef]
  74. Chaudhuri, A.K.N.; Mahapatra, P.K. Preliminary studies on anti inflammatory actions of Pluchea indica less roots. Med. Sci. Res. 1987, 15, 487–488. [Google Scholar]
  75. Mohamed, G.A.; Ibrahim, S.R.; Elkhayat, E.S.; El Dine, R.S. Natural anti-obesity agents. Bull. Facu. Pharm. Cairo Univ. 2014, 52, 269–284. [Google Scholar] [CrossRef] [Green Version]
  76. Filippatos, T.D.; Derdemezis, C.S.; Gazi, I.F.; Nakou, E.S.; Mikhailidis, D.P.; Elisaf, M.S. Orlistat–associated adverse effects and drug interactions: A critical review. Drug Safety 2008, 31, 53–65. [Google Scholar] [CrossRef]
  77. Payne, C.; Wiffen, P.J.; Martin, S. Interventions for fatigue and weight loss in adults with advanced progressive illness. Cochrane Database Syst. Rev. 2012, 1, CD008427. [Google Scholar] [PubMed]
  78. Tomita, T. Apoptosis in pancreatic beta–islet cells in type 2 diabetes. Bosn. J. Basic Med. Sci. 2016, 16, 162–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Thomas, H.E.; Kay, T.W. Intracellular pathways of pancreatic beta–cell apoptosis in type 1 diabetes. Diabetes Metab. Res. Rev. 2011, 27, 790–796. [Google Scholar] [CrossRef] [PubMed]
  80. Ibrahim, S.R.; Mohamed, G.A.; Khayat, M.T.; Ahmed, S.; Abo-Haded, H.; Alshali, K.Z. Mangostanaxanthone VIIII, a new xanthone from Garcinia mangostana pericarps, α-amylase inhibitory activity, and molecular docking studies. Rev. Bras. Farm. 2019, 29, 206–212. [Google Scholar] [CrossRef]
  81. Ibrahim, S.R.M.; Mohamed, G.A.A.; Khayat, M.T.A.; Ahmed, S.; Abo-Haded, H. Garcixanthone D, a New Xanthone, and Other Xanthone Derivatives from Garcinia mangostana Pericarps: Their α-Amylase Inhibitory Potential and Molecular Docking Studies. Starch-Stärke 2019, 71, 1800354. [Google Scholar] [CrossRef]
  82. Ibrahim, S.R.M.; Mohamed, G.A.; Khayat, M.T.; Ahmed, S.; Abo–Haded, H. α–Amylase inhibitors xanthones from Garcinia mangostana pericarps and its possible use for the treatment of diabetes with their molecular docking studies. J. Food Biochem. 2019, 43, e12844. [Google Scholar] [CrossRef]
  83. Ibrahim, S.R.; Mohamed, G.A.; Zayed, M.; Ross, S.A. 8-Hydroxyirilone 5-methyl ether and 8-hydroxyirilone, new antioxidant and α-amylase inhibitors iso flavonoids from Iris germanica rhizomes. Bioorg. Chem. 2017, 70, 192–198. [Google Scholar] [CrossRef]
  84. Vongsak, B.; Kongkiatpaiboon, S.; Jaisamut, S.; Konsap, K. Comparison of active constituents, antioxidant capacity, and α-glucosidase inhibition in Pluchea indica leaf extracts at different maturity stages. Food Biosci. 2018, 25, 68–73. [Google Scholar] [CrossRef]
  85. Suriyah, W.H.; Ichwan, S.J.A.; Kasmuri, A.R.; Taher, M. In vitro Evaluation of the Effect of Pluchea indica Extracts in Promoting Glucose Consumption Activity on A Liver Cell Line. Makara J. Health Res. 2019, 23, 48–52. [Google Scholar] [CrossRef]
  86. Reddy, P.P. Crop Residue Management and Organic Amendments; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2017; pp. 29–41. [Google Scholar]
  87. Metcalf, R.L. The Ecology of Insecticides and the Chemical Control of Insects Ecological Theory an Integrated Pest Management Practice; Kogan, M., Ed.; Wiley: New York, NY, USA, 1986. [Google Scholar]
  88. Rahayu, Y.S. The Using of Fenolic Compounds of Pluchea indica (L.). Leaves Extracts as A Bioinsecticide and Bioherbicide; In Proceedings of the Journal of Physics: Conference Series; IOP Publishing: Bristol, UK, 2018; Volume 953, p. 012206. [Google Scholar]
  89. Zhang, S.; Xu, H.; Zhang, L.; Qiao, Y. Cervical cancer: Epidemiology, risk factors and screening. Chin. J. Cancer Res. 2020, 32, 720–728. [Google Scholar] [CrossRef]
  90. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.-W. Cancer and Radiation Therapy: Current Advances and Future Directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [Green Version]
  91. Cho, J.; Cho, C.-L.; Kao, C.-L.; Chen, C.-M.; Tseng, C.-N.; Lee, Y.-Z.; Liao, L.-J.; Hong, Y.-R. Crude aqueous extracts of Pluchea indica (L.) inhibit proliferation and migration of cancer cells through induction of p53-dependent cell death. BMC Complement. Altern. Med. 2012, 12, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Kao, C. Antiproliferation and Apoptosis Induction of Ethanolic Extracts of Pluchea indica Root on Human Nasopharyngeal Carcinoma Cell Lines. Ph.D. Thesis, National Sun Yat–Sen University, Kaohsiung City, Taiwan, 2015. [Google Scholar]
  93. Ko, H.J. Effect of Crude Ethanol Extracts of Pluchea Indica on Human Leukemia K562 Cells. Master’s Thesis, National Sun Yat–Sen University, Kaohsiung City, Taiwan, 2015. [Google Scholar]
  94. Cho, C.-L.; Lee, Y.-Z.; Tseng, C.-N.; Cho, J.; Cheng, Y.-B.; Wang, K.; Chen, H.-J.; Chiou, S.-J.; Chou, C.-H.; Hong, Y.-R. Hexane fraction of Pluchea indica root extract inhibits proliferation and induces autophagy in human glioblastoma cells. Biomed. Rep. 2017, 7, 416–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Goswami, S.; Chakraborty, S.; Das, P.; Karan, S.; Naskar, D.; Debnath, S.; Chatterjee, T.K. PITC–2 loaded solid lipid nanoparticle: Design, preparation, characterization, and therapeutic comparison with free phytochemical PITC–2 isolated from tissue cultured plant Pluchea indica. Int. J. Pharm. Biol. Sci. 2019, 9, 23–36. [Google Scholar]
  96. Gomes, A.; Saha, A.; Chatterjee, I.; Chakravarty, A.K. Viper and cobra venom neutralization by beta–sitosterol and stigmasterol isolated from the root extract of Pluchea indica L. (Asteraceae). Phytomedicine 2007, 14, 637–643. [Google Scholar] [CrossRef] [PubMed]
  97. Alam, M.I.; Auddy, B.; Gomes, A. Viper venom neutralization by Indian medicinal plant (Hemidesmus indicus and Pluchea indica) root extracts. Phytother. Res. 1996, 10, 58–61. [Google Scholar] [CrossRef]
  98. Sutherland, S.K. Antivenom use in Australia: Premedication, adverse reactions and the use of venom detection kits. Med. J. Aust. 1992, 157, 734–739. [Google Scholar] [CrossRef] [PubMed]
  99. Stahel, E.; Wellauer, R.; Freyvogel, T.A. Vergiftungem durch einheimische (Vipera vipera berrus and Vipera aspis) Eine retrospective studies on 113 patient. Schweiz. Med. Wochenschr. 1985, 115, 890–896. [Google Scholar] [PubMed]
  100. Kadali, V.N.; Kameswara, R.K.; Sandeep, B.V. Medicinal plants with anti-Snake Venom property–A review. Pharm. Innov. J. 2015, 4, 11–15. [Google Scholar]
  101. Nopparat, J.; Nualla–Ong, A.; Phongdara, A. Treatment with Pluchea indica (L.) leaf ethanol extract alleviates liver injury in multiple low-dose streptozotocin-induced diabetic BALB/c mice. Exp. Therap. Med. 2020, 20, 1385–1396. [Google Scholar] [CrossRef]
  102. Mahapatra, P.K.; Chaudhuri, A.K.N. Neuropharmacological Studies on Pluchea indica. Planta Med. 1986, 6, 546–547. [Google Scholar] [CrossRef] [PubMed]
  103. Campbell, C.D.; Lee, J.Z. Fertility control in historical China revisited: New Methods for an Old Debate. Hist. Fam. 2010, 15, 370–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Payne, C.; Goldberg, E. Male contraception: Past, present and future. Curr. Mol. Pharmacol. 2014, 7, 175–181. [Google Scholar] [CrossRef] [PubMed]
  105. Dias, T.; Alves, M.; Oliveira, P.F.; Silva, B.M. Natural Products as Modulators of Spermatogenesis: The Search for a Male Contraceptive. Curr. Mol. Pharmacol. 2014, 7, 154–166. [Google Scholar] [CrossRef] [PubMed]
  106. Amalina, N.; Suyatmi, S.; Suparyanti, E.L. Effect of Beluntas (Pluchea indica) leaf extract on mice spermatogenesis. Biofarmasi 2010, 8, 47–51. [Google Scholar] [CrossRef] [Green Version]
  107. O’Donnell, L. Mechanisms of spermiogenesis and spermiation and how they are disturbed. Spermatogenesis 2015, 4, e979623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Susetyarini, E.; Latifa, R.; Miharja, F.J. DNA profile of white male rats spermatozoa after treatment with tannins Beluntas (Pluchea indica). Int. J. Eng. Technol. 2019, 8, 302–305. [Google Scholar]
  109. Sivastana, S.; Desai, P.; Coutinno, E.; Govil, G. Mechanism of action of L–arginine on the vitality of spermatozoa is primarily through increase biosynthesis of nitric oxide. Biol. Reprod. 2006, 74, 954–958. [Google Scholar]
  110. Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef]
  111. Guo, S.; DiPietro, L.A. Factors Affecting Wound Healing. J. Dent. Res. 2010, 89, 219–229. [Google Scholar] [CrossRef]
  112. Sugiaman, V.K.; Nisyah, N.Q.; Anisa, N.; Pranata, N. Pluchea indica extract as a potential source of nutrition for accelerate wound healing. Sys. Rev. Pharm. 2021, 12, 570–573. [Google Scholar]
  113. Buranasukhon, W.; Athikomkulchai, S.; Tadtong, S.; Chittasupho, C. Wound healing activity of Pluchea indica leaf extract in oral mucosal cell line and oral spray formulation containing nanoparticles of the extract. Pharm. Biol. 2017, 55, 1767–1774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Reinke, J.; Sorg, H. Wound Repair and Regeneration. Eur. Surg. Res. 2012, 49, 35–43. [Google Scholar] [CrossRef]
  115. Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell. Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Ichim, T.E.; O’Heeron, P.; Kesari, S. Fibroblasts as a practical alternative to mesenchymal stem cells. J. Transl. Med. 2018, 16, 212. [Google Scholar] [CrossRef] [Green Version]
  117. Stipcevic, T.; Piljac, J.; Berghe, D.V. Effect of Different Flavonoids on Collagen Synthesis in Human Fibroblasts. Mater. Veg. 2006, 61, 29–34. [Google Scholar] [CrossRef]
  118. Caliskan, U.K.; Aka, C.; Oz, M.G. Plants used in Anatolian traditional medicine for the treatment of hemorrhoid. Rec. Nat. Prod. 2017, 11, 235–250. [Google Scholar]
  119. Senvorasinh, K.; Phunikhom, K.; Sattayasai, J. Anti-Hemorrhoidal activity of Pluchea indica leaves aqueous extract in croton oil–induced hemorrhoids in experimental animals. Srinagarind Med. J. 2019, 34, 590–594. [Google Scholar]
  120. Nurrohman, E.; Pantiwati, Y.; Susetyarini, E.; Umami, E.K. Extract of Beluntas (Pluchea indica) as an Antibacterial Towards Streptococcus mutans ATCC 25175 Causes of Dental Carries. BIO-EDU J. Pendidik. Biol. 2021, 6, 9–17. [Google Scholar] [CrossRef]
  121. Fatimatuzzahr, N.; Rahayu, F.; Ningsih, N.S.; Darsono, A.; Salasia, S.I.O. Anticariogenic effect of Beluntas (Pluchea indica) Leaces extract as growth inhibitor of Streptococcus mutans which caused dental caries. J. Sain Vet. 2016, 34, 182–193. [Google Scholar]
  122. Fatimatuzzahr, N.; Rahayu, F.; Ningsih, N.S.; Darsono, A.; Salasia, S.I.O. Rontgen Result of Caries Molar in White Rat (Rattus novergicus) with Treatment of Herbal Toothpaste (Pluchea Indica Leaves Extract). In Proceedings of the 4th Asian Academic Society International Conference (AASIC), Globalizing Asia: Integrating Science, Technology and Humanities for Future Growth and Development, Nakhon Pathom, Thailand, 12–13 May 2016; pp. 183–187. [Google Scholar]
  123. Demolsky, W.L.; Sugiaman, V.K.; Pranata, N. Antifungal activity of Beluntas “Indian Camphorweed” (Pluchea indica) ethanol extract on Candida albicans in vitro using different solvent concentrations. Eur. J. Dent. 2021. [Google Scholar] [CrossRef] [PubMed]
  124. Sulistyani, N.; Alvionida, F.; Sugihartini, N. Composition of carbopol 940 and HPMC affects antibacterial activity of beluntas (Pluchea indica (L.)) leaves extract gel. Pharmaciana 2021, 11, 427–438. [Google Scholar] [CrossRef]
  125. Komala, O.; Wiendarlina, I.Y.; Rizqiyana, N. Antibacterial activity roll on deodorant with Pluchea indica (L.) leaf extract against Staphylococcus epidermidis (Evans 1916) in vitro. IOP Conf. Ser. Earth Environ. Sci. 2019, 293, 012031. [Google Scholar] [CrossRef]
  126. Sylvana, D.; Amir, M.; Purnamasari, C.B.; Iskandar, A.; Asfirizal, V. Antibacterial activity of ethanol extract of Beluntas leaves on Streptococcus mutans, Porphyromonas gingivalis, and Enterococcus faecalis. Padjadjaran J. Dent. 2021, 33, 191–198. [Google Scholar] [CrossRef]
  127. Pargaputri, A.F.; Munadziroh, E.; Indrawati, R. The Effect of Pluchea indica L. Leaves Extract Againts Biofilm of Enterococcus faecalis and Fusobacterium nucleatum In Vitro. Denta 2017, 11, 51. [Google Scholar] [CrossRef]
  128. Simanjuntak, N.; Yuniarni, U.; Prayugo, D. Antibacterial activity of Pluchea indica and Piper beetle ethanol extract on Staphylococcus epidermidis and Pseudomonas aeruginosa. Pharmacol. Clin. Pharm. Res. 2016, 1, 62–68. [Google Scholar]
  129. Sittiwet, C. In vitro Antimicrobial Activity of Pluchea indica Aqueous Extract: The Potential for Urinary Tract Infection Treatment. J. Pharmacol. Toxicol. 2009, 4, 87–90. [Google Scholar] [CrossRef] [Green Version]
  130. Pramanik, K.C.; Chatterjee, T.K. In vitro and in vivo antibacterial activities of root extract of tissue cultured Pluchea indica (L.) Less. Orient. Pharm. Exp. Med. 2008, 8, 295–301. [Google Scholar] [CrossRef] [Green Version]
  131. Srimoon, R.; Ngiewthaisong, S. Antioxidant and antibacterial activities of Indian marsh fleabane (Pluchea indica (L.). KKU Res J. 2015, 20, 144–154. [Google Scholar]
  132. Farhamzah, H.A.; Mursal, I.L. Formulation and antibacterial activity test of foot spray with Beluntas leaf ethanol extract (Pluchea indica L.). IOP Conf. Ser. Mater. Sci. Eng. 2021, 1071, 12013. [Google Scholar] [CrossRef]
  133. Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free Radicals: Properties, Sources, Targets, and Their Implication in Various Diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Halliwell, B.; Gutteridge, J.M.C. Free Radicals in Biology and Medicine, 5th ed.; Oxford University Press: New York, NY, USA, 2015. [Google Scholar]
  135. Srisook, K.; Buapool, D.; Boonbai, R.; Simmasut, P.; Charoensuk, Y.; Srisook, E. Antioxidant and anti-inflammatory activities of hot water extract from Pluchea indica L. herbal tea. J. Med. Plant Res. 2012, 6, 4077–4408. [Google Scholar]
  136. Widyawati, P.S.; Budianta, T.D.W.; Werdani, Y.D.W.; Halim, M.O. Antioxidant Activity of Pluchea Leaves–Black Tea Drink (Pluchea indica L.–Camelia sinensis). Agritech 2018, 38, 200–207. [Google Scholar] [CrossRef] [Green Version]
  137. Widyawati, P.S.; Budianta, T.D.W.; Kusuma, F.A.; Wijaya, E.L.; Yaunatan, D.I.; Wongso, R.S. Potency of Beluntas (Pluchea indica L.) Leaves to Extract as Antioxidant and Anti-Warmed–Over Flavor (WOF) of Duck Meat. In Proceedings of the International Congress Challenges of Biotechnological Research in Food and Health, Surakarta, Indonesia, 15 November 2014; pp. 81–89. [Google Scholar]
  138. Sirichaiwetchakoon, K.; Lowe, G.M.; Eumkeb, G. The Free Radical Scavenging and Anti-Isolated Human LDL Oxidation Activities of Pluchea indica (L.) Tea Compared to Green Tea (Camellia sinensis). BioMed Res. Int. 2020, 2020, 4183643. [Google Scholar] [CrossRef]
  139. Sirichaiwetchakoon, K.; Lowe, G.M.; Kupittayanant, S.; Churproong, S.; Eumkeb, G. Pluchea indica (L.) tea ameliorates hyperglycemia, dyslipidemia, and obesity in high fat diet–fed mice. Evid. Based Complemen. Altern. Med. 2020, 2020, 8746137. [Google Scholar] [CrossRef] [PubMed]
  140. Werdani, Y.D.W.; Widyawati, P.S. Antidiabetic Effect on Tea of Pluchea indica Less as Functional Beverage in Diabetic Patients. In Proceedings of the 1st International Conference Postgraduate School Universitas Airlangga: “Implementation of Climate Change Agreement to Meet Sustainable Development Goals” (ICPSUAS 2017), Jawa Timur, Indonesia, 1–2 August 2017; Atlantis Press: Paris, France, 2018; Volume 98, pp. 164–167. [Google Scholar]
Figure 1. Structures of compounds 19.
Figure 1. Structures of compounds 19.
Molecules 27 02383 g001
Figure 2. Structures of compounds 1025.
Figure 2. Structures of compounds 1025.
Molecules 27 02383 g002
Figure 3. Structures of compounds 2633.
Figure 3. Structures of compounds 2633.
Molecules 27 02383 g003
Figure 4. Structures of compounds 3439.
Figure 4. Structures of compounds 3439.
Molecules 27 02383 g004
Figure 5. Structures of compounds 4043.
Figure 5. Structures of compounds 4043.
Molecules 27 02383 g005
Figure 6. Structures of compounds 4457.
Figure 6. Structures of compounds 4457.
Molecules 27 02383 g006
Figure 7. Structures of compounds 5869.
Figure 7. Structures of compounds 5869.
Molecules 27 02383 g007
Figure 8. Structures of compounds 7083.
Figure 8. Structures of compounds 7083.
Molecules 27 02383 g008
Figure 9. Structures of compounds 8491.
Figure 9. Structures of compounds 8491.
Molecules 27 02383 g009
Figure 10. Structures of compounds 9296.
Figure 10. Structures of compounds 9296.
Molecules 27 02383 g010
Figure 11. Structures of compounds 97101.
Figure 11. Structures of compounds 97101.
Molecules 27 02383 g011
Figure 12. Structures of compounds 102112.
Figure 12. Structures of compounds 102112.
Molecules 27 02383 g012
Figure 13. Structures of compounds 113122.
Figure 13. Structures of compounds 113122.
Molecules 27 02383 g013
Figure 14. Number of metabolites from various classes reported from P. indica. MT: Monoterpenes; ST: Sesquiterpenes; TT: Triterpenes; CQs: Caffeoylquinic acid derivatives; Flav: Flavonoids; Phen: Phenolics; Lig: Lignans; Thioph: Thiophenes.
Figure 14. Number of metabolites from various classes reported from P. indica. MT: Monoterpenes; ST: Sesquiterpenes; TT: Triterpenes; CQs: Caffeoylquinic acid derivatives; Flav: Flavonoids; Phen: Phenolics; Lig: Lignans; Thioph: Thiophenes.
Molecules 27 02383 g014
Figure 15. Number of reported metabolites from P. indica from various countries.
Figure 15. Number of reported metabolites from P. indica from various countries.
Molecules 27 02383 g015
Figure 16. Number of metabolites isolated from different parts of P. indica.
Figure 16. Number of metabolites isolated from different parts of P. indica.
Molecules 27 02383 g016
Table 1. List of reported metabolites from Pluchea indica.
Table 1. List of reported metabolites from Pluchea indica.
Compound NamePlant PartExtract/FractionMol. Wt.Mol. FormulaCity, CountryRef.
Monoterpenes
(+)-Linalool (1)Aerial partsPolar fraction of MeOH extract154C10H18ODrigh Road, Karachi, Pakistan[47]
Linaloyl glucopyranoside (2)Aerial partsPolar fraction of MeOH extract316C16H28O6Drigh Road, Karachi, Pakistan[47]
Linaloyl apiosyl glucopyranoside (3)Aerial partsPolar fraction of MeOH extract448C21H36O10Drigh Road, Karachi, Pakistan[47]
(+)-9-Hydroxylinalool (4)Aerial partsPolar fraction of MeOH extract170C10H18O2Drigh Road, Karachi, Pakistan[47]
9-Hydroxylinaloyl glucopyranoside (5)Aerial partsPolar fraction of MeOH extract332C16H28O7Drigh Road, Karachi, Pakistan[47]
Plucheoside B (6)Aerial partsPolar fraction of MeOH extract388C19H32O8Drigh Road, Karachi, Pakistan[47]
Thymol (7)Aerial partsPolar fraction of MeOH extract150C10H14ODrigh Road, Karachi, Pakistan[48]
Plucheoside C (8)Aerial partsPolar fraction of MeOH extract444C21H32O10Drigh Road, Karachi, Pakistan[48]
(E)-4-(3-Hydroxybut-1-en-1-yl)-3,5,5-trimethylcyclohex-3-ene-1,2-diol (9)Aerial partsPolar fraction of MeOH extract226C13H22O3Drigh Road, Karachi, Pakistan[47]
Sesquiterpenes
3-(2′,3′-Diacetoxy-2′-methyl butyryl)-cuauhtemone (10)LeavesCHCl3 fraction of EtOH extract452C24H36O8Nakornpathom, Thailand[49]
Cuauhtemone (11)LeavesCHCl3 fraction of EtOH extract252C15H24O3Nakornpathom, Thailand[49]
Plucheoside A (12)Aerial partsPolar fraction of MeOH extract412C21H32O8Drigh Road, Karachi, Pakistan[47]
Plucheoside E (13)Aerial partsPolar fraction of MeOH extract400C21H36O7Drigh Road, Karachi, Pakistan[48]
Herbolide A (14)Aerial partsPolar fraction of MeOH extract292C17H24O4Drigh Road, Karachi, Pakistan[47]
Pterocarptriol (15)Aerial partsPolar fraction of MeOH extract256C15H28O3Drigh Road, Karachi, Pakistan[48]
Plucheol A (16)Aerial partsPolar fraction of MeOH extract254C15H26O3Drigh Road, Karachi, Pakistan[48]
Plucheol B (17)Aerial partsPolar fraction of MeOH extract254C15H26O3Drigh Road, Karachi, Pakistan[48]
(+)-Cyperone (18)RootsCH2Cl2 fraction of MeOH extract218C15H22OKaohsiung, Taiwan[50]
Costunolide (19)RootsCH2Cl2 fraction of MeOH extract232C15H20O2Kaohsiung, Taiwan[50]
Caryolane-1,9β-diol (20)Aerial partsn-Hexane fraction of EtOH extract238C15H26O2Hepu, Guangxi, China[51]
(8R,9R)-Isocaryolane-8,9-diol (21)Aerial partsn-Hexane fraction of EtOH extract238C15H26O2Hepu, Guangxi, China[51]
Clovane-2α,9β-diol (22)Aerial partsn-Hexane fraction of EtOH extract238C15H26O2Hepu, Guangxi, China[51]
Valenc-1(10)-ene-8,11-diol (23)Aerial partsn-Hexane fraction of EtOH extract238C15H26O2Hepu, Guangxi, China[51]
Valenc-1(10)-ene-8-hydroxy-11-O-glucopyranoside (24)Aerial partsEtOAc fraction of MeOH extract400C21H37O7China[52]
(10S,11S)-Himachala-3-(12)-4-diene (25)LeavesEssential oil204C15H24Sidoarjo and Surabaya, Indonesia[43]
Triterpenes
Taraxasterol (26)Leavesn-Hexane fraction of MeOH extract426C30H50OGia Lam, Hanoi, Vietnam[53]
Taraxasterol acetate (27)Leavesn-Hexane fraction of MeOH extract468C32H52O2Gia Lam, Hanoi, Vietnam[53]
Multiflorenol (28)Aerial partsn-Hexane fraction of MeOH extract426C30H50OChina[52]
Sterols
Stigmasterol (29)Aerial partsn-Hexane fraction of EtOH extract412C29H48OHepu, Guangxi, China[51]
Twigsn-Hexane fraction of MeOH extract--Vietnam[54]
Leavesn-Hexane fraction of MeOH extract412C29H48OGia Lam, Hanoi, Vietnam[53]
Stigmasterol 3-O-β-D-glucopyranoside (30)Aerial partsPolar fraction of MeOH extract574C35H58O6Drigh Road, Karachi, Pakistan[47]
TwigsEtOAc fraction of MeOH extract--Vietnam[54]
LeavesEtOAc fraction of MeOH extract--Gia Lam, Hanoi, Vietnam[53]
β-Sitosterol (31)Twigsn-Hexane fraction of MeOH extract414C29H50OVietnam[54]
β-Sitosterol 3-O-β-D-glucopyranoside (32)LeavesEtOAc fraction of MeOH extract576C35H60O6Gia Lam, Hanoi, Vietnam[53]
Campesteryl ferulate (33)LeavesEtOH extract576C38H56O4Hat Yai, Songkhla, Thailand[45]
Caffeoylquinic acid derivatives
3-O-Caffeoylquinic acid (34)Aerial partsAcidified MeOH354C16H18O9Seberang Perai, Malaysia[55]
Leaves50% EtOH extract--Different provinces in Thailand[56]
4-O-Caffeoylquinic acid (35)Leaves50% EtOH extract354C16H18O9Different provinces in Thailand[56]
5-O-Caffeoylquinic acid (36)Aerial partsAcidified MeOH354C16H18O9Seberang Perai, Malaysia[55]
Leaves50% EtOH extract--Different provinces in Thailand[56]
3,4-Di-O-Caffeoylquinic acid (37)Aerial partsAcidified MeOH--Seberang Perai, Malaysia[55]
Leaves50% EtOH extract530C26H26O12Different provinces in Thailand[56]
Leaves50% EtOH extract--Different provinces in Thailand[46]
3,5-Di-O-Caffeoylquinic acid (38)Aerial partsAcidified MeOH516C25H24O12Seberang Perai, Malaysia[55]
LeavesEtOAc fraction of MeOH extract--Yogyakarta, Indonesia[57]
Leaves50% EtOH extract--Different provinces in Thailand[56]
Leaves50% EtOH extract--Different provinces in Thailand[46]
4,5-Di-O-Caffeoylquinic acid (39)Aerial partsAcidified MeOH516C25H24O12Seberang Perai, Malaysia[55]
4,5-Di-O-caffeoylquinic acid methyl ester (40)LeavesEtOAc fraction of MeOH extract530C26H26O12Yogyakarta, Indonesia[57]
Leaves50% EtOH extract--Different provinces in Thailand[56]
3,4,5-Tri-O-Caffeoylquinic acid (41)LeavesEtOAc fraction of MeOH extract678C34H30O15Khon Kaen, Thailand[58]
LeavesEtOAc fraction of MeOH extract--Yogyakarta, Indonesia[57]
3,4,5-Tri-O-Caffeoylquinic acid methyl ester (42)LeavesEtOAc fraction of MeOH extract692C35H32O15Yogyakarta, Indonesia[57]
1,3,4,5-Tetra-O-Caffeoylquinic acid (43)LeavesEtOAc fraction of MeOH extract840C43H36O18Khon Kaen, Thailand[58]
LeavesEtOAc fraction of MeOH extract--Yogyakarta, Indonesia[57]
Flavonoids
Quercetin (44)LeavesEtOAc fraction of MeOH extract302C15H10O7Khon Kaen, Thailand[58]
Aerial partsAcidified 50% MeOH--Bogor, west Java, Indonesia[29]
Aerial partsEtOAc fraction of MeOH extract--Chantaburi, Thailand[59]
Quercetin-3-O-β-D-glucopyranoside (45)Aerial partsEtOAc fraction of MeOH extract464C21H20O12China[52]
Aerial partsAcidified MeOH--Seberang Perai, Malaysia[55]
Quercetin-3-O-β-D-galactopyranoside (46)Aerial partsAcidified MeOH464C21H20O12Seberang Perai, Malaysia[55]
3′,4′,5,7-Tetrahydroxy-flavone-3-O-β-D-mannopyranoside (47)Aerial partsEtOAc fraction of MeOH extract464C21H20O12China[52]
Quercetin-3-O-sulphate (48)Aerial partsAcidified MeOH382C15H10O10SSeberang Perai, Malaysia[55]
Myricetin (49)Aerial partsAcidified 50% MeOH318C15H10O8Bogor, west Java, Indonesia[29]
Kaempferol (50)Aerial partsAcidified 50% MeOH286C15H10O6Bogor, west Java, Indonesia[29]
6-Hydroxykaempferol 7-glucoside (51) 464C20H20O12Hat Yai, Songkhla, Thailand[45]
4′,5,7-Trihydroxyflavone-3-O-β-D-glucoside (52)Aerial partsEtOAc fraction of MeOH extract448C21H20O11China[52]
Apigenin (53)Aerial partsAcidified 50% MeOH270C15H10O5Bogor, west Java, Indonesia[29]
Aerial partsEtOAc fraction of MeOH extract--Chantaburi, Thailand[59]
Apigenin 7-(2″,3″diacetylglucoside (54) [45]
Chrysoeriol (55)Aerial partsEtOAc fraction of MeOH extract300C16H12O6Chantaburi, Thailand[59]
Luteolin (56)Aerial partsAcidified 50% MeOH286C15H10O6Bogor, west Java, Indonesia[29]
Aerial partsEtOAc fraction of MeOH extract--Chantaburi, Thailand[59]
8-Hydroxyluteolin 8-glucoside (57)LeavesEtOH extract464C20H20O12Hat Yai, Songkhla, Thailand[45]
Phenolic acids, aldehydes, esters, and ketones
P-Hydroxybenzoic acid (58)Aerial partsCHCl3 fraction of EtOH extract138C7H6O3Hepu, Guangxi, China[51]
3,4,5-Trimethoxybenzoic acid (59)RootsCH2Cl2 fraction of MeOH extract212C10H12O5Kaohsiung, Taiwan[50]
Caffeic acid (60)Aerial partsEtOAc fraction of MeOH extract180C9H8O4China[52]
P-Hydroxybenzaldehyde (61)RootsCH2Cl2 fraction of MeOH extract122C7H6O2Kaohsiung, Taiwan[50]
3,4-Dihydroxy benzaldehyde (62)Aerial partsCHCl3 fraction of EtOH extract138C7H6O3Hepu, Guangxi, China[51]
Vanillin (63)Aerial partsCHCl3 fraction of EtOH extract152C8H8O3Hepu, Guangxi, China[51]
3,4-Dihydroxy-5-methoxybenzaldehyde (64)Aerial partsEtOAc fraction of MeOH extract168C8H8O4China[52]
Aerial partsCHCl3 fraction of EtOH extract--Hepu, Guangxi, China[51]
Syringicaldehyde (65)Aerial partsCHCl3 fraction of EtOH extract182C9H10O4Hepu, Guangxi, China[51]
Trans-Coniferyl aldehyde (66)Aerial partsCHCl3 fraction of EtOH extract178C10H10O3Hepu, Guangxi, China[51]
Dibutylphthalate (67)Aerial partsCHCl3 fraction of EtOH extract278C16H22O4Hepu, Guangxi, China[51]
Ethyl caffeate (68)Aerial partsCHCl3 fraction of EtOH extract208C11H12O4Hepu, Guangxi, China[51]
2,3-Dihydroxy-1-(4-hydroxy-3-methoxyphenyl)-propan-1-one (69)Aerial partsCHCl3 fraction of EtOH extract212C10H12O5Hepu, Guangxi, China[51]
Phenolics and phenolic glucosides
1,2-Bis-(4-Hydroxy-3-methoxyphenyl)-propane-1,3-diol (erythro) (70)Aerial partsPolar fraction of MeOH extract320C17H20O6Drigh Road, Karachi, Pakistan[47]
1,2-Bis-(4-Hydroxy-3-methoxyphenyl)-propane-1,3-diol (threo) (71)Aerial partsPolar fraction of MeOH extract320C17H20O6Drigh Road, Karachi, Pakistan[47]
1-(4-Hydroxy-3-methoxyphenyl)-2-{4-[(1E)-3-hydroxyprop-1-en-1-yl]-2-methoxyphenoxy}-propane-1,3-diol (erythro) (72)Aerial partsPolar fraction of MeOH extract360C20H24O6Drigh Road, Karachi, Pakistan[47]
1-(4-Hydroxy-3-methoxyphenyl)-2-{4-[(1E)-3-hydroxyprop-1-en-1-yl]-2-methoxyphenoxy}-propane-1,3-diol (threo) (73)Aerial partsPolar fraction of MeOH extract360C20H24O6Drigh Road, Karachi, Pakistan[47]
Esculetin (74)Aerial partsCHCl3 fraction of EtOH extract178C9H6O4Hepu, Guangxi, China[51]
3,4,5-Trimethoxyphenyl-β-D-glucopyranoside (75)RootsCH2Cl2 fraction of MeOH extract346C15H22O9Kaohsiung, Taiwan[50]
Benzyl glucopyranoside (76)Aerial partsPolar fraction of MeOH extract270C13H18O6Drigh Road, Karachi, Pakistan[47]
Phenylethyl glucopyranoside (77)Aerial partsPolar fraction of MeOH extract284C14H20O6Drigh Road, Karachi, Pakistan[47]
Methyl salicylate glucoside (78)Aerial partsPolar fraction of MeOH extract314C14H18O8Drigh Road, Karachi, Pakistan[47]
4-Allyl-2-methoxy-6-hydroxyphenylglucoside (79)Aerial partsEtOAc fraction of MeOH extract342C16H22O8China[52]
4-Allyl-2,6-dimethoxy phenyl glucopyranoside (80)Aerial partsPolar fraction of MeOH extract356C17H24O8Drigh Road, Karachi, Pakistan[47]
Eugenyl glucoside (Citrucin C) (81)Aerial partsPolar fraction of MeOH extract326C16H22O7Drigh Road, Karachi, Pakistan[47]
Tangshenoside Ⅱ (82)Aerial partsEtOAc fraction of MeOH extract372C17H24O9China[52]
Thalictoside (83)RootsCH2Cl2 fraction of MeOH extract329C14H19NO8Kaohsiung, Taiwan[50]
Lignans and their derivatives
(+)-Syringaresinol (84)RootsCH2Cl2 fraction of MeOH extract418C22H26O8Kaohsiung, Taiwan[50]
(+)-Diasyringaresinol (85)RootsCH2Cl2 fraction of MeOH extract418C22H26O8Kaohsiung, Taiwan[50]
Aerial partsCHCl3fraction of EtOH extract--Hepu, Guangxi, China[51]
(+)-Epi-Syringaresinol (86)RootsCH2Cl2 fraction of MeOH extract418C22H26O8Kaohsiung, Taiwan[50]
Syringaresinol monoglucopyranoside (87)Aerial partsPolar fraction of MeOH extract580C28H36O13Drigh Road, Karachi, Pakistan[47]
Pinoresinol (88)Aerial partsEtOAc fraction of MeOH extract358C20H22O6China[52]
(+)-Epi-Pinoresinol (89)Aerial partsEtOAc fraction of MeOH extract358C20H22O6China[52]
Pinoresinol monoglucopyranoside (90)Aerial partsPolar fraction of MeOH extract520C26H32O11Drigh Road, Karachi, Pakistan[47]
Liriodendrin (91)RootsCH2Cl2 fraction of MeOH extract742C34H46O18Kaohsiung, Taiwan[50]
Plucheoside D1 (92)Aerial partsPolar fraction of MeOH extract518C26H30O11Drigh Road, Karachi, Pakistan[48]
Plucheoside D2 (93)Aerial partsPolar fraction of MeOH extract548C27H32O12Drigh Road, Karachi, Pakistan[48]
Plucheoside D3 (94)Aerial partsPolar fraction of MeOH extract534C27H34O11Drigh Road, Karachi, Pakistan[48]
Hedyotisol A (95)Aerial partsPolar fraction of MeOH extract810C42H50O16Drigh Road, Karachi, Pakistan[47]
Hedyotisol B (96)Aerial partsPolar fraction of MeOH extract810C42H50O16Drigh Road, Karachi, Pakistan[47]
Trans-Trismethoxy resveratrol-d4 (97)LeavesEtOH extract274C17H14D4O3Hat Yai, Songkhla, Thailand[45]
Threo-2,3-Bis(4-hydroxy-3-methoxyphenyl)-3-ethoxypropan-1-ol (98)Aerial partsEtOAc fraction of EtOH extract348C19H24O6Hepu, Guangxi, China[51]
Erythro-2,3-Bis(4-hydroxy-3-methoxyphenyl)-3-ethoxypropan-1-ol (99)Aerial partsEtOAc fraction of EtOH extract348C19H24O6Hepu, Guangxi, China[51]
(+)-Isolariciresinol (100)Aerial partsEtOAc fraction of EtOH extract360C20H24O6Hepu, Guangxi, China[51]
(+)-9′-Isovaleryllariciresinol (101)Aerial partsEtOAc fraction of EtOH extract444C25H32O7Hepu, Guangxi, China[51]
Thiophenes and their derivatives
2-(Pro-1-ynyl)-5-(5,6-dihydroxypenta-1,3-diynyl) thiophene (PYDDT) = 2-(Prop-1-ynyl)-5(5,6-dihydroxyhexa-1, 3-diynyl)-thiophene = PITC-2 = R/J/3 (102)RootsEtOAc fraction of MeOH extract230C13H10O2SWest Bengal, India[60]
Aerial partsn-Hexane fraction of MeOH extract--Chantaburi, Thailand[59]
Leavesn-Hexane fraction of MeOH extract--Gia Lam, Hanoi, Vietnam[53]
RootsEtOAc fraction of MeOH extract--West Bengal, India[61]
Twigsn-Hexane fraction of MeOH extract--Vietnam[54]
2-(4-Hydroxy-3-methoxybut-1-yn-1-yl)-5-(penta-1,3-diyn-1-yl)thiophene (103)Aerial partsEtOAc fraction of MeOH extract244C14H12O2SGuangzhou, China[62]
2-(3,4-Dihydroxybut-1-yn-1-yl)-5-(penta-1,3-diyn1-yl)thiophene (104)Aerial partsEtOAc fraction of MeOH extract230C13H10O2SGuangzhou, China[62]
2-(3-Acetoxy-4-hydroxybut-1-yn-1-yl)-5-(penta-1,3-diyn-1-y1) thiophene (105)Aerial partsEtOAc fraction of MeOH extract272C15H12O3SGuangzhou, China[62]
2-(Prop-1-yn-1-yl)-5-(6-acetoxy-5-hydroxyhexa-1,3-diyn-1-yl) thiophene = 2-(Prop-1-inyl)-5-(6-acetoxy-5-hydroxyhexa-1,3-diinyl) thiophene (106)Aerial partsEtOAc fraction of MeOH extract272C15H12O3SGuangzhou, China[62]
Aerial partsn-Hexane fraction of MeOH extract--Chantaburi, Thailand[59]
2-(Penta-1,3-diyn-1-yl)-5-(4-acetoxy-3-hydroxybuta-1-yn-1-yl) thiophene (107)Aerial partsn-Hexane fraction of MeOH extract272C15H12O3SChantaburi, Thailand[59]
3″R-Pluthiophenol (108)Aerial partsn-Hexane fraction of EtOH extract230C13H10O2SHepu, Guangxi, China[51]
3″R-Pluthiophenol-4″-acetate (109)Aerial partsn-Hexane fraction of EtOH extract272C15H12O3SHepu, Guangxi, China[51]
3″-Ethoxy-3″S-pluthiophenol (110)Aerial partsn-Hexane fraction of EtOH extract258C15H14O2SHepu, Guangxi, China[51]
3″-Ethoxy-3″S-pluthiophenol-4″-acetate (111)Aerial partsn-Hexane fraction of EtOH extract300C17H16O3SHepu, Guangxi, China[51]
2-(4-O-β-Glucopyranosyl-3-hydroxybut-1-yn-1-yl)-5-(penta-1,3-diyn-1-yl)thiophene (112)Aerial partsEtOAc fraction of MeOH extract392C19H20O7SGuangzhou, China[62]
Other metabolites
1-Dotriacontanol (113)Leavesn-Hexane fraction of MeOH extract466C32H66OGia Lam, Hanoi, Vietnam[53]
9,12,13-Trihydroxyoctadeca-10(E),15(Z)-dienoic acid (114)Aerial partsn-Hexane fraction of EtOH extract328C18H32O5Hepu, Guangxi, China[51]
Pinellic acid (115)Aerial partsn-Hexane fraction of EtOH extract330C18H34O5Hepu, Guangxi, China[51]
1-Eicosanoyl glycerol (116)Twigsn-Hexane fraction of MeOH extract386C23H46O4Vietnam[54]
Methyl 9-hydroxynonanoate (117)Aerial partsn-Hexane fraction of EtOH extract188C10H20O3Hepu, Guangxi, China[51]
Triethyl citrate (118)Aerial partsn-Hexane fraction of EtOH extract276C12H20O7Hepu, Guangxi, China[51]
(Z)-3-Hexenyl glucopyranoside (119)Aerial partsPolar fraction of MeOH extract262C12H22O6Drigh Road, Karachi, Pakistan[47]
Fraxinellone (120)Aerial partsn-Hexane fraction of EtOH extract232C14H16O3Hepu, Guangxi, China[51]
Adenosine (121)Aerial partsEtOAc fraction of EtOH extract267C10H13N5O4Hepu, Guangxi, China[51]
Uridine 5′-monophosphate (122)Aerial partsEtOAc fraction of MeOH extract324C9H13N2O9PChina[52]
Table 3. Biological activity of reported metabolites from Pluchea indica.
Table 3. Biological activity of reported metabolites from Pluchea indica.
Compound NameBiological ActivityAssay, Organism, or Cell LineBiological ResultsRef.
CompoundPositive Control
Caryolane-1,9β-diol (20)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells104.8 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
(8R,9R)-Isocaryolane-8,9-diol (21)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells95.1 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Clovane-2α,9β-diol (22)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells101.6 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Valenc-1(10)-ene-8,11-diol (23)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells103.8 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Stigmasterol (29)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells92.5 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
3,5-Di-O-caffeoylquinic acid (38)α-Glucosidase inhibitionColorimetric/Rat intestinal maltase1166 µM (IC50)Acarbose 0.5 µM (IC50)[57]
4,5-Di-O-caffeoylquinic acid methyl ester (40)α-Glucosidase inhibitionColorimetric/Rat intestinal maltase208.0 µM (IC50)Acarbose 0.5 µM (IC50)[57]
3,4,5-Tri-O-caffeoylquinic acid (41)α-Glucosidase inhibitionColorimetric/Rat intestinal maltase13.0 µM (IC50)Acarbose 0.5 µM (IC50)[57]
Collagenase inhibitionFluorometric/Collagenase type IV1.5 µM (IC50)Phosphramidon 7.4 µM (IC50)[58]
MMP-2 inhibitionFluorometric/MMP-2 proenzyme2.5 µM (IC50)Chlorhexidine 7.3 µM (IC50)[58]
MMP-9 inhibitionFluorometric/MMP-9 monomer6.4 µM (IC50)Chlorhexidine 25.2 µM (IC50)[58]
3,4,5-tri-O-caffeoylquinic acid methyl ester (42)α-Glucosidase inhibitionColorimetric/Rat intestinal maltase2.0 µM (IC50)Acarbose 0.5 µM (IC50)[57]
1,3,4,5-Tetra-O-Caffeoylquinic acid (43)α-Glucosidase inhibitionColorimetric/Rat intestinal maltase11.0 µM (IC50)Acarbose 0.5 µM (IC50)[57]
Collagenase inhibitionFluorometric/Collagenase type IV6.3 µM (IC50)Phosphramidon 7.4 µM (IC50)[58]
MMP-2 inhibitionFluorometric/MMP-2 proenzyme18.4 µM (IC50)Chlorhexidine 7.3 µM (IC50)[58]
MMP-9 inhibitionFluorometric/MMP-9 monomer16.8 µM (IC50)Chlorhexidine 25.2 µM (IC50)[58]
Quercetin (44)Collagenase inhibitionFluorometric/Collagenase type IV16.9 µM (IC50)Phosphramidon 7.4 µM (IC50)[58]
CYP2A6 inhibitionEnzymatic reconstitution2.66 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 0.80 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
Apigenin (53)CYP2A6 inhibitionEnzymatic reconstitution0.9 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 0.05 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
Luteolin (56)CYP2A6 inhibitionEnzymatic reconstitution1.38 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 0.18 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
Chrysoeriol (55)CYP2A6 inhibitionEnzymatic reconstitution1.14 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 0.82 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
3,4-Dihydroxy benzaldehyde (62)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells92.9 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Vanillin (63)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells99.6 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
3,4-Dihydroxy-5-methoxybenzaldehyde (64)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells103.9 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Syringicaldehyde (65)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells92.6 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Trans-Coniferyl aldehyde (66)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells94.2 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Dibutylphthalate (67)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells101.1 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Ethyl caffeate (68)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells77.9 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
2,3-Dihydroxy-1-(4-hydroxy-3-methoxyphenyl)-propan-1-one (69)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells100.9 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Esculetin (74)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells88.5 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
(+)-Diasyringaresinol (85)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells101.7 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Threo-2,3-Bis(4-hydroxy-3-methoxyphenyl)-3-ethoxypropan-1-ol (98)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells101.7 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Erythro-2,3-Bis(4-hydroxy-3-methoxyphenyl)-3-ethoxypropan-1-ol (99)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells99.7 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
(+)-Isolariciresinol (100)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells101.9 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
(+)-9′-Isovaleryllariciresinol (101)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells77.6 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
2-(Prop-1-inyl)-5-(5,6-dihydroxyhexa-1,3-diinyl) thiophene (102)CYP2A6 inhibitionEnzymatic reconstitution3.90 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 2.40 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
Anti-amoebicCell count/Entamoeba histolytica (HM1)50 µg/mL (MIC)Metronidazole 5 µg/mL (MIC)[60]
2-(Prop-1-inyl)-5-(6-acetoxy-5-hydroxyhexa-1, 3-diinyl) thiophene (106)CYP2A6 inhibitionEnzymatic reconstitution4.44 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 2.94 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
2-(Penta-1,3-diyn-1-yl)-5-(4-acetoxy-3-hydroxybuta-1-yn-1-yl) thiophene (107)CYP2A6 inhibitionEnzymatic reconstitution6.43 µM (IC50)Methoxsalen 0.19 µM (IC50)[59]
CYP2A13 inhibition 6.18 µM (IC50)Methoxsalen 0.43 µM (IC50)[59]
3″R-Pluthiophenol (108)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells84.5 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
3″R-Pluthiophenol-4″-acetate (109)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells83.4 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
3″-Ethoxy-3″S-pluthiophenol (110)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells86.9 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
3″-Ethoxy-3″S-pluthiophenol-4″-acetate (111)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells90.1 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
9,12,13-Trihydroxyoctadeca-10(E),15(Z)-dienoic acid (114)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells90.3 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Pinellic acid (115)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells89.5 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Methyl 9-hydroxynonanoate (117)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells93.6 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Triethyl citrate (118)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells91.1 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Fraxinellone (120)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells52.1 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Adenosine (121)Anti-inflammatory/inhibition of NO productionLPS-stimulated production in RAW 264.7 macrophages cells88.7 (NRC % inhibition)Dexamethasone 62.2 (NRC % inhibition)[51]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ibrahim, S.R.M.; Bagalagel, A.A.; Diri, R.M.; Noor, A.O.; Bakhsh, H.T.; Mohamed, G.A. Phytoconstituents and Pharmacological Activities of Indian Camphorweed (Pluchea indica): A Multi-Potential Medicinal Plant of Nutritional and Ethnomedicinal Importance. Molecules 2022, 27, 2383. https://doi.org/10.3390/molecules27082383

AMA Style

Ibrahim SRM, Bagalagel AA, Diri RM, Noor AO, Bakhsh HT, Mohamed GA. Phytoconstituents and Pharmacological Activities of Indian Camphorweed (Pluchea indica): A Multi-Potential Medicinal Plant of Nutritional and Ethnomedicinal Importance. Molecules. 2022; 27(8):2383. https://doi.org/10.3390/molecules27082383

Chicago/Turabian Style

Ibrahim, Sabrin R. M., Alaa A. Bagalagel, Reem M. Diri, Ahmad O. Noor, Hussain T. Bakhsh, and Gamal A. Mohamed. 2022. "Phytoconstituents and Pharmacological Activities of Indian Camphorweed (Pluchea indica): A Multi-Potential Medicinal Plant of Nutritional and Ethnomedicinal Importance" Molecules 27, no. 8: 2383. https://doi.org/10.3390/molecules27082383

Article Metrics

Back to TopTop