Skip to main content
Tropical Medicine and Infectious Disease logoLink to Tropical Medicine and Infectious Disease
. 2022 Nov 23;7(12):395. doi: 10.3390/tropicalmed7120395

Phytochemical, Antimalarial, and Acute Oral Toxicity Properties of Selected Crude Extracts of Prabchompoothaweep Remedy in Plasmodium berghei-Infected Mice

Walaiporn Plirat 1,2, Prapaporn Chaniad 1,2, Arisara Phuwajaroanpong 1,2, Abdi Wira Septama 3, Chuchard Punsawad 1,2,*
Editors: Borimas Hanboonkunupakarn, Kesinee Chotivanich, Lorenz Von Seidlein, Liwang Cui
PMCID: PMC9785619  PMID: 36548650

Abstract

Malaria remains a life-threatening health problem and encounters with the increasing of antimalarial drug resistance. Medicinal plants play a critical role in synthesizing novel and potent antimalarial agents. This study aimed to investigate the phytochemical constituents, antiplasmodial activity, and evaluate the toxicity of crude ethanolic extracts of Myristica fragrans, Atractylodes lancea, and Prabchompoothaweep remedy in a mouse model. The phytochemical constituents were characterized by liquid chromatography-mass spectrometry (LC-MS). Antimalarial efficacy against Plasmodium berghei was assessed using 4-day suppressive tests at doses of 200, 400, and 600 mg/kg body weight. Acute toxicity was assessed at a dose of 2000 mg/kg body weight of crude extracts. The 4-day suppression test showed that all crude extracts significantly suppressed parasitemia (p < 0.05) compared to the control group. Higher parasitemia suppression was observed both in Prabchompoothaweep remedy at a dose of 600 mg/kg (60.1%), and A. lancea at a dose of 400 mg/kg (60.1%). The acute oral toxicity test indicated that the LD50 values of all extracts were greater than 2000 mg/kg and that these extracts were not toxic in the mouse model. LC-MS analysis revealed several compounds in M. fragrans, A. lancea, and Prabchompoothaweep remedy. For quantitative analysis, 1,2,6,8-tetrahydroxy-3-methylanthraquinone 2-O-b-D-glucoside, chlorogenic acid, and 3-O-(beta-D-glucopyranosyl-(1->6)-beta-D-glucopyranosyl) ethyl 3-hydroxyoctanoate were found in A. lancea, while (7′x,8′x)-4,7′-epoxy-3,8′-bilign-7-ene-3,5′-dimethoxy-4′,9,9′-triol, edulisin III, and tetra-hydrosappanone A trimethyl ether are found in M. fragrans. 6′-O-Formylmarmin was present in the Prabchompoothaweep remedy, followed by pterostilbene glycinate and amlaic acid. This study showed that the ethanolic extracts of A. lancea and Prabchompoothaweep remedy possess antimalarial activity against Plasmodium berghei. None of the extracts had toxic effects on liver and kidney function. Therefore, the ethanolic extract of A. lancea rhizome and Prabchompoothaweep remedy could be used as an alternative source of new antimalarial agents. Further studies are needed to determine the active compounds in both extracts.

Keywords: antimalarial activity, toxicity, Prabchompoothaweep remedy, Myritica fragrans, Atractylodes lancea, malaria

1. Introduction

Malaria is one of the most serious and life-threatening infectious diseases caused by protozoan parasites of the Plasmodium genus. It is responsible for the high rates of mortality and morbidity in the tropical and subtropical regions of the world, where the climate is suitable for parasite development [1]. According to the World Health Organization report in 2021, there were approximately 241 million cases of malaria which caused 0.6 million deaths worldwide [2]. The mortality rate from malaria has been reduced in recent years due to extensive malaria control through the use of insecticide-impregnated bed nets and treatment with artemisinin derivatives; however, the state of artemisinin resistance, the standard drug for treating malaria, is of great concern [3]. Artemisinin-based combination therapies (ACTs) are the first-line drugs for malaria in a large majority of endemic countries, and intravenous artesunate is usually used for the treatment of severe malaria [4]. Although ACTs act as a fast-acting artemisinin derivative and a slow-acting combined drug, the efficacy of ACTs is limited by long-lasting parasite clearance, contributing to ACT failure [5,6]. Furthermore, Plasmodium falciparum infection has become resistant to almost all available antimalarial drugs, which is estimated to be 10% in Southeast Asia and 93% in Thailand [7]. To manage this pathology, a new antimalarial compound that is safer, more effective than older drugs, and has a novel mode of action is urgently required.

For centuries, plants and herbs have been an important source of drugs being developed to provide a potential treatment for many diseases. Plants contain a large number of bioactive molecules and are a valuable source of pharmacotherapeutics [8,9]. Furthermore, antimalarial drugs, especially quinine and artemisinin, are derived from traditional medicines and plant extracts [10]. Therefore, natural plants are a good source of inspiration in searching for a new antimalarial agent.

Prabchompoothaweep is a traditional Thai medicine that is part of the National List of Essential Medicines (NLEM), which includes 23 herbs [11]. The bioactivity of the ethanolic extract of Prabchompoothaweep remedy, including antiallergic activity, anti-inflammation, and antioxidant activities, has been reported. According to the NLEM, the Prabchompoothaweep remedy is usually suggested to be useful for the treatment of many types of fever, including malaria-like symptoms such as intermittent fever and common cold [12]. In addition, Prabchompoothaweep remedy and two-component plants of Prabchompoothaweep remedy, Myristica fragrans, and Atractylodes lancea, have been reported to show in vitro antimalarial activity. From our previous studies, the in vitro antimalarial activity of the ethanolic extracts of the Prabchompoothaweep remedy, M. fragrans, and A. lancea, displayed antimalarial activity (IC50 = 14.13 µg/mL, 5.96 µg/mL, and 7.73 µg/mL, respectively) (unpublished data). M. fragrans is an aromatic evergreen tropical tree belonging to the Myristicaceae family [13]. M. fragrans has been used to treat several diseases. In particular, the mace part, which is an aril of M. fragrans, has been used for asthma, fever, and gastrointestinal treatment in Ayurvedic medicine [14]. Furthermore, M. fragrans has been suggested to have various medicinal properties, such as antimicrobial, chemoprotective, antioxidant, anti-inflammatory effects, anti-atherosclerosis, and behavioral effects [15]. A. lancea belongs to the Asteraceae (Compositae) family [16]. A. lancea has been used to treat rheumatic diseases, digestive disorders, night blindness, and influenza [17]. The pharmacological properties of rhizomes, including anti-cancer, anti-inflammatory, and antimicrobial activities and activities on the central nervous, cardiovascular, and gastrointestinal systems, have been investigated [18]. Prabchompoothaweep remedy and two-component plants have shown good in vitro antimalarial activity. Therefore, the Prabchompoothweep remedy and its two components are good candidates for further investigation of the in vivo antimalarial activity.

Based on ethnobotanical evidence and our in vitro study of antimalarial activity and toxicity, ethanolic mace extracts of M. fragrans, ethanolic rhizome extract of A. lancea, and ethanolic crude extract of Prabchompoothaweep remedy were found to have good activity against parasite infection without cytotoxicity to Vero cells. Therefore, this study aimed to investigate the potential antimalarial activity and toxicological assessment of two crude extracts from the Prabchompoothaweep remedy in a mouse model. Furthermore, the phytochemical content of selected crude extracts of the Prabchompoothaweep remedy was explored to understand the origin of the bioactivity.

2. Materials and Methods

2.1. Plant Collection

The dried arils (mace) of M. fragrans, dried rhizome of A. lancea, and Prabchompoothaweep remedy were purchased from a traditional Thai drug store in the Nakhon Si Thammarat region of Thailand. The authorization for plant materials complied with the relevant guidelines and regulations of the Plant Varieties Protection, Department of Agriculture, Ministry of Agriculture and Cooperatives, Thailand. The botanical identification of the plant samples was confirmed by a botanist at the School of Pharmacy, Walailak University. Specimens with voucher numbers for M. fragrans (SMD177004003-2) and A. lancea (SMD072010001) were deposited in the School of Medicine, Walailak University.

2.2. Preparation of Plant Extracts

First, the plant samples were powdered using a herb grinder (Jincheng, Model; SF, China). M. fragrans aril powder (60 g), A. lancea rhizome powder (60 g), and Prabchompoothaweep remedy powder (60 g) were soaked in 600 mL of 95% ethanol for 72 h at room temperature (1:10 (w/v) ratio). The mixed solutions were filtered using gauze and Whatman filter No. 1. The unfiltered residues were remacerated in 95% ethanol for 72 h. This procedure was repeated two times. The filtered solutions were combined and concentrated using a rotary evaporator (Buchi® rotary evaporator, Model R-210, Shanghai, China). The residues were then dried in a water bath at 60 °C. Finally, the dried crude extracts of M. fragrans aril, A. lancea rhizome, and Prabchompoothaweep remedy were stored in a refrigerator at 4 °C until use. For animal experiments, each crude extract was dissolved in 7% Tween 80 and 3% ethanol in distilled water to obtain the working concentration.

2.3. Phytochemical Screening

The ethanolic extract was qualitatively investigated to reveal the presence of phytochemical constituents, including flavonoids, terpenoids, alkaloids, tannins, anthraquinones, cardiac glycosides, saponins, and coumarins. These were identified by characteristic color changes using standard procedures [19,20,21].

2.4. Liquid Chromatography-Quadrupole Time-of-Flight Mass Spectrometry (LC-QTOF MS) Analysis

The metabolite profiles of M. fragrans extract, A. lancea extract, and Prabchompoothaweep remedy extract were determined using by an ultra-high performance liquid chromatography (UHPLC) instrument equipped with an electrospray ionization source (ESI). The UHPLC system consisted of a Zorbax Eclipse Plus C18 Rapid Resolution HD column (150 mm length × 2.1 mm inner-diameter, particle size 1.8 µm) with an LC-QTOF MS instrument (1290 Infinity II LC-6545 Quadrupole-TOF, Agilent Technologies, Santa Clara, CA, USA). The mobile phase comprised solvent A (0.1% formic acid in water) and solvent B (acetonitrile). The volume of injection was 2.0 µL, and the column temperature was set at 25 °C. Qualitative analysis of LC-MS/MS was performed in negative ion mode with a scanning range from m/z 100 to 1200 using a Dual AJS ESI ion source. The phytochemical compounds in the extract samples were identified by comparing the retention time, mass data, and fragmentation patterns with known compounds in the library search of the Mass Hunter METLIN database (Agilent Technologies). The compound selection was selected and identified from the peak with 90% similarity in the database.

2.5. Animals and Rodent Parasites

Healthy male Institute of Cancer Research (ICR) mice aged 6–8 weeks, weighing 20–30 g, were purchased from Nomura Siam International Co., Ltd., Bangkok, Thailand. The animals were housed and acclimatized for 7 days under standard and constant laboratory conditions (22 ± 3 °C, 50–60% humidity and 12 h light/dark cycles) with free access to food and clean water. The animal care staff controlled the hygiene by cleaning and removing waste from the cages daily. The mice were handled according to the international guidelines for the animals used in the experiments. The wild-type rodent Plasmodium berghei ANKA strain was obtained from Biodefense and Emerging Infections Research Resources Repository (BEI Resources), National Institute of Allergy and Infectious Diseases (NIAID), and National Institute of Health (NIH), which was received from Thomas F. McCutchan. Mouse donors were injected with P. berghei-infected red blood cells via an intraperitoneal route. When the mouse donors had parasitemia levels of 20–30%, blood was drawn from the heart by cardiac puncture and kept in a heparinized tube for injection into experimental mice.

2.6. Animal Grouping and Dosing

For Peter’s 4-day suppressive test, infected male ICR mice were randomly divided into 12 groups of five mice per group. Group 1 (infected control mice) was administered a mixture of 7% Tween 80 and 3% ethanol in distilled water. Groups 2 and 3 (positive control) received 6 mg/kg body weight of artesunate (Art) and 25 mg/kg body weight of chloroquine (CQ), respectively. Groups 4, 5, and 6 were administered 200, 400, and 600 mg/kg body weight of M. fragrans crude extract, respectively. Groups 7, 8, and 9 were administered 200, 400, and 600 mg/kg body weight A. lancea crude extract, respectively. Groups 10, 11, and 12 were administered 200, 400, and 600 mg/kg body weight of Prabchompoothaweep remedy crude extract, respectively. Dosage selection was chosen based on the results of oral acute toxicity and preliminary results were obtained for the extracts. For oral acute toxicity testing, mice were randomly assigned to five groups of five mice each. Group 1 (untreated control group) received no treatment; Group 2 (negative control group) was treated with a mixture of 7% Tween 80 and 3% ethanol in distilled water; Group 3 was treated with a dose of 2000 mg/kg body weight of M. fragrans crude extract; Group 4 was treated with a dose of 2000 mg/kg body weight of A. lancea crude extract; and Group 5 was treated with a dose of 2000 mg/kg body weight of Prabchompoothaweep remedy crude extract. Acute toxicity in mice was induced by oral administration.

2.7. Four-Day Suppressive Test (Peter’s Test)

The protocol for a 4-day suppressive test was evaluated according to previous studies [22]. First, all mice were injected with 0.2 mL of 1 × 107 infected blood cells (intraperitoneally); 3 h after infection, the mice in each group were treated with the crude extract as described above and continued to be treated for 3 consecutive days (24, 48, and 72 h after infection). Treatment was administered via oral gavage to mimic the traditional route of administration. On day 5 post-infection, blood was collected from the vascular tail vein to prepare a thin blood smear film. Thin blood smears were stained with 10% Giemsa solution (Biotech Reagent Company Limited, Bangkok, Thailand) to evaluate parasitemia. Parasitemia was observed under a light microscope (Olympus, model: CX-31, Tokyo, Japan) with a 100X objective lens. The percentage of parasitemia was determined from five different fields with an estimated 300 red blood cells per field, and the percentage of parasitemia was calculated using the following formula:

%parasitemia=number of parasitized red blood cellsnumber of total red blood cells

The percentage of parasitemia suppression was calculated using the following formula:

%suppression=[AB]A × 100

where A is the mean percentage of parasitemia in the infected control group and B is the mean percentage of parasitemia in each treatment group.

2.8. Pack Cell Volume (PCV)

The effectiveness of the crude extracts in preventing hemolysis due to increasing parasite levels was measured using PCV. The tail vein of mice was cut to collect the blood, and the blood was kept in heparinized micro-hematocrit capillary tubes by filling them up to 3/4. One side of the capillary was plugged with clay. The capillary tubes were then centrifuged at 9520× g for 5 min with the sealed ends outwards. The PCV of each mouse was determined on day 0 before infection with P. berghei and day 4 after treatment.

2.9. Acute Toxicity Measurement

The oral acute toxicity of ethanolic crude extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy was investigated in male ICR mice according to the standard guidelines of the Organization for Economic Co-operation and Development (OECD) [23]. Twenty-five mice were randomly separated into five groups of five, which were explained in the animal grouping and dosing section. On day 1, the mice were not allowed to obtain food and water for 3 h before treatment. Subsequently, the mice in the treatment group were orally administered a single dose of 2000 mg/kg body weight of M. fragrans, A. lancea, or Prabchompoothaweep remedy extract. A mixture of 7% Tween 80 and 3% ethanol in distilled water served as the negative control, and untreated mice served as the control. Three hours after treatment, the mice were noted to have physical and behavioral changes such as muscle tone, mood, sleep, excretion, appetite, and hair erection. The animals were observed daily for 14 days. Food and water intake were recorded daily. The body weight of the mice was measured on days 0 and 14 using a sensitive digital weighing balance (Mettler Toledo, model: ML3002E, Bekasi City, Indonesia). On day 14, the mice were anesthetized with 50 mg/kg body weight sodium pentobarbital (Ceva Sante Animale, Maassluis, The Netherlands) by intraperitoneal injection. After anesthetization, mouse blood was collected for biochemical analysis. Liver and kidney tissues were harvested for histopathological examination using hematoxylin and eosin (H&E) staining.

2.10. Biochemical Analysis

Blood samples from the acute toxicity test group were collected from the heart using a cardiac puncture technique. Blood was centrifuged at 3000× g for 5 min to separate the plasma, which was collected to evaluate liver and kidney function. Liver and kidney functions were tested for biochemical parameters, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphate (ALP), blood urea nitrogen (BUN), and creatinine levels, using an AU 480 chemistry analyzer (Beckman Coulter, Brea, CA, USA).

2.11. Histopathological Examination

Histopathological investigation was performed using a standard laboratory procedure, as previously reported [24,25,26]. The tissues were fixed in 10% (v/v) formalin at room temperature, dehydrated with a series of alcohol concentrations, cleared with xylene, and embedded in paraffin. After tissue processing, the liver and kidney tissues were cut to 5 µm thickness using a microtome, stained with hematoxylin and eosin solution, and evaluated under a light microscope by two independent observers blinded to the condition groups.

2.12. Statistical Analysis

Statistical analysis was performed with SPSS statistical software version 23 (IBM, Armonk, NY, USA). Quantitative data were presented as means ± standard errors of the means (means ± SEMs). The Kolmogorov–Smirnov test was used to assess the normal distribution of each parameter. Differences in the mean parameters between the groups, such as the percentage of parasitemia, percentage of suppression, food and water consumption, body weight, and liver and lung biochemical parameters, were analyzed with a one-way analysis of variance followed by a post-hoc Tukey’s multiple comparison test. A p-value of less than 0.05 was considered statistically significant for all tests.

3. Results

3.1. Percentage Yield and Phytochemical Screening of Ethanolic Crude Extracts

The percentage yield of the ethanolic mace extract of M. fragrans, rhizome extract of A. lancea, and Prabchompoothaweep remedy was 20.71%, 22.11%, and 5.43%, respectively. The phytochemical constituents included flavonoids, terpenoids, alkaloids, tannins, and coumarins (Table 1).

Table 1.

Phytochemical screening of ethanolic extract of M. fragrans mace, A. lancea rhizome, and Prabchompoothaweep remedy.

Phytochemical Constituents M. fragrans A. lancea Prabchompoothaweep Remedy
Flavonoid + - -
Terpenoids + + +
Alkaloids + + +
Tannins - - +
Anthraquinones - - -
Cardiac glycosides - - -
Saponins - - -
Coumarins + - +

(+), detected; (-), not detected phytochemical constituents.

3.2. LC-QTOF-MS Analysis

Qualitative analysis of the compounds in extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy was performed using LC-QTOF-MS in negative mode. Metabolite profiling of the crude extract compounds was performed using a database of well-known compounds in the Library METLIN database. The complete list of compounds detected using LC-QTOF-MS is given in Table 2, Table 3 and Table 4 and supported by Figure 1, Figure 2 and Figure 3.

Table 2.

Compounds identified in the ethanolic M. fragrans extract by LC-QTOF-MS.

No. M/Z RT
(min)
Compounds Molecular
Formula
Molecular Weight
1 133.014 2.087 Malic acid C4H6O5 134.021
2 149.009 1.886 Tartaric acid C4H6O6 150.016
3 285.040 27.493 Luteolin C15H10O6 286.047
4 357.134 27.969 (7′x,8′x)-4,7′-Epoxy-3,8′-bilign-7-ene-3,5′-dimethoxy-4′,9,9′-triol C20H22O6 358.141
5 201.149 31.852 3-Hydroxynonyl acetate C11H22O3 202.156
6 265.056 2.1360 Monoglyceride citrate C9H14O9 266.063
7 373.165 38.781 Sonchifolin C21H26O6 374.172
8 345.134 31.690 Gibberellin A92 C19H22O6 346.141
9 161.045 4.429 3-Hydroxy-3-methyl-glutaric acid C6H10O5 162.052
10 179.071 15.867 Propyl 2-furanacrylate C10H12O3 180.078
11 207.066 18.673 Sinapyl aldehyde C11H12O4 208.073
12 219.050 5.294 1-Hydroxypentane-1,2,5-tricarboxylate C8H12O7 220.058
13 329.103 35.937 Isoamericanol A C18H18O6 330.110
14 299.092 33.581 2,4-Dihydroxy-6,4′-dimethoxychalcone C17H16O5 300.099
15 167.034 9.353 Dihydroxyphenylacetic acid C8H8O4 168.042
16 183.102 34.258 Ascariadole epoxide C10H16O3 184.109
17 375.144 31.978 alpha-Peroxyachifolide C20H24O7 376.152
18 191.019 2.124 Citric acid C6H8O7 192.026
19 287.055 21.191 3′,4′,5,7-Tetrahydroxyisoflavanone C15H12O6 288.063
20 149.060 26.290 2-(2-Furanyl)-3-methyl-2-butenal C9H10O2 150.067
21 329.139 36.576 Tetrahydrosappanone A trimethyl ether C19H22O5 330.146
22 371.186 34.358 Tanabalin C22H28O5 372.193
23 315.123 39.745 5′-Hydroxy-3′,4′,7-trimethoxyflavan C18H20O5 316.130
24 265.144 38.918 Isoleptospermone C15H22O4 266.151
25 237.113 20.077 Benzyl b-L-arabinopyranoside C13H18O4 238.120
26 301.035 27.794 Hieracin C15H10O7 302.042
27 285.040 32.491 Kaempferol C15H10O6 286.047
28 389.160 39.444 Rosmic acid C21H26O7 390.167
29 271.060 29.798 Methylnorlichexanthone C15H12O5 272.068
30 267.071 1.936 2(α-D-Mannosyl)-D-glycerate C9H16O9 268.078
31 177.040 3.139 L-Sorbosone C6H10O6 178.047
32 303.050 16.481 (±)-Taxifolin C15H12O7 304.058
33 177.019 9.265 Esculetin C9H6O4 178.026
34 359.149 35.899 6′-O-Formylmarmin C20H24O6 360.156
35 387.144 31.364 Edulisin III C21H24O7 388.151
36 331.118 23.709 5′,8-Dihydroxy-3′,4′,7-trimethoxyflavan C18H20O6 332.125
37 359.076 34.408 Jaceidin C18H16O8 360.083
38 271.060 31.401 (±)-Naringenin C15H12O5 272.067
39 201.112 6.847 2,6-Dimethyl-1,8-octanedioic acid C10H18O4 202.120
40 329.232 33.243 9S,10S,11R-trihydroxy-12Z-octadecenoic acid C18H34O5 330.240
41 311.128 39.657 Gancaonin V C19H20O4 312.135
42 117.018 2.688 Succinic acid C4H6O4 118.026
43 163.039 16.068 m-Coumaric acid C9H8O3 164.047
44 197.045 9.904 2-Hydroxy-3,4-dimethoxybenzoic acid C9H10O5 198.052
45 133.050 2.713 2,3-Dihydroxy-2-methylbutanoic acid C5H10O4 134.057
46 281.138 10.129 Bisbynin C15H22O5 282.146
47 221.081 13.762 2,3-Dihydro-3-hydroxy-6-methoxy-2,2-dimethyl-4H-1-benzopyran-4-one C12H14O4 222.088
48 239.070 37.691 2,4-Dihydroxychalcone C15H12O3 240.078
49 317.066 21.617 Dihydroisorhamnetin C16H14O7 318.073
50 443.191 5.794 Cynaroside A C21H32O10 444.198
51 371.134 20.577 Citrusin E C17H24O9 372.141
52 447.092 18.473 Kaempferol-7-O-glucoside C21H20O11 448.099
53 205.086 32.679 2,3-Dihydro-6-methoxy-2,2-dimethyl-4H-1-benzopyran-4-one C12H14O3 206.094
54 343.154 31.101 Safficinolide C20H24O5 344.161
55 353.102 32.303 1-(3,4-Dihydroxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione C20H18O6 354.109
56 313.107 33.706 7-Hydroxyenterolactone C18H18O5 314.114
57 445.170 11.357 Crosatoside B C20H30O11 446.177
58 331.115 24.787 Mytilin A C13H20N2O8 332.122
59 263.128 24.737 (+)-Abscisic acid C15H20O4 264.135
60 426.227 35.285 Dihydroxyacidissiminol C25H33NO5 427.234
61 343.118 22.682 Diosbulbin B C19H20O6 344.125
62 187.096 19.475 Methyl N-(a-methylbutyryl) glycine C9H16O4 188.104

Table 3.

Compounds identified in the ethanolic A. lancea extract by LC-QTOF-MS.

No. M/Z RT
(min)
Compounds Molecular
Formula
Molecular Weight
1 191.056 1.970 Quinic acid C7H12O6 192.063
2 179.035 9.637 Caffeic acid C9H8O4 180.042
3 177.019 9.274 Esculetin C9H6O4 178.026
4 243.062 1.995 Pseudouridine C9H12N2O6 244.069
5 191.034 14.798 Scopoletin C10H8O4 192.042
6 209.118 33.628 3-Ethenyl-2,5-dimethyl-4-oxohex-5-en-2-yl acetate C12H18O3 210.125
7 161.024 14.673 3-Hydroxycoumarin C9H6O3 162.031
8 353.087 7.419 Chlorogenic acid C16H18O9 354.094
9 281.139 32.475 Bisbynin C15H22O5 282.146
10 207.029 11.341 Fraxetin C10H8O5 208.036
11 207.066 28.153 5-(3′,5′-Dihydroxyphenyl)-gamma-valerolactone C11H12O4 208.073
12 265.144 33.653 Isoleptospermone C15H22O4 266.151
13 193.050 15.124 Scytalone C10H10O4 194.057
14 311.128 28.454 Gancaonin V C19H20O4 312.135
15 153.019 8.372 Gentisic acid C7H6O4 154.026
16 147.029 2.721 D-threo-3-methylmalate C5H8O5 148.036
17 341.108 1.907 Sucrose C12H22O11 342.115
18 353.087 8.008 5Z-Caffeoylquinic acid C16H18O9 354.094
19 341.087 7.156 Glucocaffeic acid C15H18O9 342.094
20 447.092 12.368 1,2,6,8-Tetrahydroxy-3-methylanthraquinone 2-O-b-D-glucoside C21H20O11 448.100
21 427.196 16.703 Taraxacolide 1-O-b-D-glucopyranoside C21H32O9 428.204
22 225.112 25.472 3,7-Dimethyl-2E,6E-decadien-1,10-dioic acid C12H18O4 226.120
23 221.045 15.487 Isofraxidin C11H10O5 222.052
24 128.035 2.208 Pyroglutamic acid C5H7NO3 129.042
25 381.175 10.163 1,2,10-Trihydroxydihydro-trans-linalyl oxide 7-O-beta-D-glucopyranoside C16H30O10 382.183
26 337.092 10.338 Hydrojuglone glucoside C16H18O8 338.099
27 441.175 15.600 Lusitanicoside C21H30O10 442.183
28 485.199 14.673 Glucosylgalactosyl hydroxylysine C18H34N2O13 486.207
29 401.144 8.847 Benzyl O-(arabinofuranosyl-(1->6)-glucoside) C18H26O10 402.151
30 385.164 26.675 Gingerenone B C22H26O6 386.172
31 335.076 13.169 4-O-Caffeoylshikimic acid C16H16O8 336.083
32 305.138 32.788 Achillicin C17H22O5 306.146
33 425.144 16.414 6-(2-Carboxyethyl)-7-hydroxy-2,2-dimethyl-4-chromanone glucoside C20H26O10 426.151
34 353.144 26.675 Isopropyl apiosylglucoside C14H26O10 354.151
35 503.175 13.169 (S)-Multifidol 2-(apiosyl-(1->6)-glucoside) C22H32O13 504.183
36 329.232 33.164 9S,10S,11R-trihydroxy-12Z-octadecenoic acid C18H34O5 330.239
37 461.238 13.270 xi-Linalool 3-(rhamnosyl-(1->6)-glucoside) C22H38O10 462.245
38 511.238 10.789 3-O-(beta-D-glucopyranosyl-(1->6)-beta-D-glucopyranosyl) ethyl 3-hydroxyoctanoate C22H40O13 512.245
39 393.133 39.996 Rotenone C23H22O6 394.141
40 447.092 15.976 Kaempferol-7-O-glucoside C21H20O11 448.099
41 461.165 9.837 Verbasoside C20H30O12 462.172
42 479.248 10.263 3-O-(alpha-L-rhamnopyranosyl-(1-2)-alpha-L-rhamnopyranosyl)-3-hydroxydecanoic acid C22H40O11 480.255
43 529.264 18.356 Cinncassiol D2 glucoside C26H42O11 530.271
44 515.118 20.235 3″,4″-Diacetylafzelin C25H24O12 516.125
45 128.035 2.496 (r)-(+)-2-Pyrrolidone-5-carboxylic acid C5H7NO3 129.042
46 441.212 16.828 CAY10509 C23H35FO5S 442.219
47 299.141 32.688 Bifenazate C17H20N2O3 300.148

Table 4.

Compounds identified in the ethanolic extract of Prabchompoothaweep remedy by LC-QTOF-MS.

No. M/Z RT
(min)
Compounds Molecular
Formula
Molecular Weight
1 173.045 1.988 Shikimic acid C7H10O5 174.052
2 197.045 9.842 2-Hydroxy-3,4-dimethoxybenzoic acid C9H10O5 198.052
3 169.014 3.816 Gallic acid C7H6O5 170.021
4 177.019 9.216 Esculetin C9H6O4 178.026
5 137.024 7.725 3,4-Dihydroxybenzaldehyde C7H6O3 138.031
6 169.014 6.497 2,4,6-Trihydroxybenzoic acid C7H6O5 170.021
7 243.051 3.491 1-O-Galloylglycerol C10H12O7 244.058
8 166.050 7.976 2-Amino-3-methoxy-benzoic acid C8H9NO3 167.058
9 447.129 24.625 Piperenol C C22H24O10 448.136
10 153.019 5.320 3,4-Dihydroxybenzoic acid C7H6O4 154.026
11 211.061 19.401 Eudesmic acid C10H12O5 212.068
12 187.097 19.489 Methyl N-(a-methylbutyryl) glycine C9H16O4 188.104
13 197.045 13.212 3,4-O-Dimethylgallic acid C9H10O5 198.052
14 313.056 3.290 Salicyl phenolic glucuronide C13H14O9 314.063
15 161.081 7.249 Potassium 2-(1’-ethoxy) ethoxypropanoate C7H14O4 162.089
16 179.035 9.629 Caffeic acid C9H8O4 180.042
17 151.040 9.078 4-Acetoxyphenol C8H8O3 152.047
18 290.088 2.012 Sarmentosin epoxide C11H17NO8 291.095
19 191.034 22.608 5,7-Dihydroxy-4-methylcoumarin C10H8O4 192.042
20 353.087 7.713 5Z-Caffeoylquinic acid C16H18O9 354.094
21 237.113 20.065 Benzyl b-L-arabinopyranoside C13H18O4 238.120
22 222.040 14.340 (R)-2,3-Dihydro-3,5-dihydroxy-2-oxo-3-indoleacetic acid C10H9NO5 223.047
23 218.103 3.516 Pantothenic acid C9H17NO5 219.110
24 195.102 16.169 Isobutyl 2-furanpropionate C11H16O3 196.109
25 421.186 32.869 Picrasin F C22H30O8 422.193
26 355.030 2.137 (+)-Chebulic acid C14H12O11 356.037
27 153.019 8.352 Gentisic acid C7H6O4 154.026
28 325.056 3.992 Fertaric acid C14H14O9 326.063
29 243.123 23.360 Polyethylene, oxidized C12H20O5 244.130
30 233.045 6.735 7-Hydroxy-2-methyl-4-oxo-4H-1-benzopyran-5-acetic acid C12H10O5 234.052
31 299.055 32.242 Diosmetin C16H12O6 300.063
32 310.140 11.734 Leonurine C14H21N3O5 311.147
33 300.998 15.430 Ellagic acid C14H6O8 302.005
34 328.118 20.604 N-trans-Feruloyloctopamine C18H19NO5 329.125
35 225.112 11.947 3,7-Dimethyl-2E,6E-decadien-1,10-dioic acid C12H18O4 226.120
36 163.039 13.538 m-Coumaric acid C9H8O3 164.047
37 321.024 7.900 Digallate C14H10O9 322.032
38 359.149 34.898 6′-O-Formylmarmin C20H24O6 360.156
39 651.083 9.316 Amlaic acid C27H24O19 652.090
40 285.040 32.518 Kaempferol C15H10O6 286.047
41 463.087 15.881 Quercetin 3-galactoside C21H20O12 464.094
42 347.076 7.024 alpha-(1,2-Dihydroxyethyl)-1,2,3,4-tetrahydro-7-hydroxy-9-methoxy-3,4-dioxocyclopenta(c) benzopyran-6-acetaldehyde C17H16O8 348.083
43 431.170 33.708 Melledonal A C23H28O8 432.177
44 261.040 17.396 2-Acetyl-5,8-dihydroxy-3-methoxy-1,4-naphthoquinone C13H10O6 262.047
45 315.050 33.082 1,3,5,8-Tetrahydroxy-6-methoxy-2-methylanthraquinone C16H12O7 316.057
46 326.087 7.449 Blepharin C14H17NO8 327.094
47 461.108 25.966 Rhamnetin 3-rhamnoside C22H22O11 462.115
48 161.060 18.223 Allyl benzoate C10H10O2 162.067
49 128.035 2.426 Pyroglutamic acid C5H7NO3 129.042
50 271.060 31.302 (±)-Naringenin C15H12O5 272.067
51 264.066 34.710 Piperolactam A C16H11NO3 265.073
52 272.129 29.724 (2E)-Piperamide-C5:1 C16H19NO3 273.136
53 191.055 1.887 Quinic acid C7H12O6 192.063
54 134.024 10.356 2-Benzoxazolol C7H5NO2 135.032
55 361.165 22.821 Gibberellin A98 C20H26O6 362.172
56 201.112 26.041 2,6-Dimethyl-1,8-octanedioic acid C10H18O4 202.120
57 476.040 13.688 Isoterchebin C41H30O27 954.096
58 312.123 24.863 Pterostilbene glycinate C18H19NO4 313.131
59 285.040 27.444 Luteolin C15H10O6 286.047
60 635.088 11.308 3-O-Galloylhamamelitannin C27H24O18 636.095
61 351.053 26.191 4′-O-Methyl-(-)-epicatechin-7-O-sulfate C16H16O7S 352.061
62 269.045 31.453 Apigenin C15H10O5 270.052
63 343.045 36.564 Aflatoxin GM1 C17H12O8 344.052
64 307.081 18.474 4R,5R,6S-Trihydroxy-2-hydroxymethyl-2-cyclohexen-1-one 6-(2-hydroxy-6-methylbenzoate) C15H16O7 308.089
65 447.092 18.487 Kaempferol-7-O-glucoside C21H20O11 448.099
66 461.072 19.726 3-Methylellagic acid 8-rhamnoside C21H18O12 462.079
67 201.018 26.341 6-Hydroxyangelicin C11H6O4 202.026
68 623.197 15.943 Isoacteoside C29H36O15 624.204
69 211.060 5.119 3-Hydroxy-4-methoxyphenyllactic acid C10H12O5 212.067
70 301.034 27.820 Hieracin C15H10O7 302.042
71 477.139 24.462 Eugenol O-[3,4,5-Trihydroxybenzoyl-(->6)-b-D-glucopyranoside] C23H26O11 478.146
72 342.134 25.665 N-trans-Feruloyl-4-O-methyldopamine C19H21NO5 343.141
73 547.144 21.255 Puerarin xyloside C26H28O13 548.152
74 251.128 15.667 QH (2) C14H20O4 252.135
75 329.029 28.897 2,8-Di-O-methylellagic acid C16H10O8 330.036
76 256.133 34.309 Coumaperine C16H19NO2 257.140
77 491.118 28.772 3′,7-Dimethoxy-4′,5,8-trihydroxyflavone 8-glucoside C23H24O12 492.125
78 281.138 10.080 Bisbynin C15H22O5 282.146
79 403.175 32.255 Myristicanol B C22H28O7 404.183
80 403.123 5.921 Oleoside 11-methyl ester C17H24O11 404.131
81 465.102 17.998 (-)-Epicatechin 7-O-glucuronide C21H22O12 466.110
82 379.175 20.403 6b-Angeloyl-3b,8b,9b-trihydroxy-7(11)-eremophilen-12,8-olide C20H28O7 380.182
83 593.150 17.221 Saponarin C27H30O15 594.157
84 379.012 28.672 Tectorigenin 7-sulfate C16H12O9S 380.019
85 241.071 6.046 Elenaic acid C11H14O6 242.078
86 955.104 16.570 Chebulinic acid C41H32O27 956.111
87 477.102 19.000 Myricetin 3,4′-dimethyl ether 3′-xyloside C22H22O12 478.110
88 497.223 19.614 2-O-(beta-D-galactopyranosyl-(1->6)-beta-D-galactopyranosyl) 2S-hydroxynonanoic acid C21H38O13 498.230
89 593.129 27.795 6″-O-p-Coumaroyltrifolin C30H26O13 594.136
90 515.118 20.177 3″,4″-Diacetylafzelin C25H24O12 516.125
91 161.045 4.443 3-Hydroxy-3-methyl-glutaric acid C6H10O5 162.052
92 447.092 12.385 1,2,6,8-Tetrahydroxy-3-methylanthraquinone 2-O-b-D-glucoside C21H20O11 448.099
93 387.107 16.945 7-Hydroxy-3′,4′,5,6,8-pentamethoxyflavone C20H20O8
388.115
94 769.254 18.349 Leonoside A C35H46O19 770.261
95 637.176 13.876 Quercetin 3,3′-dimethyl ether 7-rutinoside C29H34O16 638.183
96 431.097 19.150 Apigenin 7-O-glucoside C21H20O10 432.104
97 581.222 12.498 (+)-Lyoniresinol 9-glucoside C28H38O13 582.230
98 461.238 13.976 xi-Linalool 3-(rhamnosyl-(1->6)-glucoside) C22H38O10 462.245
99 695.399 31.177 Glucosyl passiflorate C37H60O12 696.407
100 461.165 4.543 Verbasoside C20H30O12 462.172
101 755.238 15.292 Hesperetin 7-(2,6-dirhamnosylglucoside) C34H44O19 756.246
102 435.128 19.075 Phenethyl 6-galloylglucoside C21H24O10 436.135
103 429.152 23.685 2,3-dinor Fluprostenol C21H25F3O6 430.159
104 651.228 24.262 (-)-Matairesinol 4′-(apiosyl-(1->2)-glucoside) C31H40O15 652.235
105 153.055 4.944 2-Furanylmethyl propanoate C8H10O3 154.062
106 665.207 23.460 Tetramethylquercetin 3-rutinoside C31H38O16 666.214
107 637.212 19.376 4′-Hydroxy-5,7,2′-trimethoxyflavanone 4′-rhamnosyl-(1->6)-glucoside C30H38O15 638.219
108 582.259 30.238 N1, N5, N10-Tricoumaroyl spermidine C34H37N3O6
583.266
109 433.149 20.854 Vestitone 7-glucoside C22H26O9 434.156
110 453.248 33.457 Rhodojaponin IV C24H38O8 454.255
111 137.024 8.088 m-Salicylic acid C7H6O3 138.031
112 477.066 10.080 Quercetin 3′-O-glucuronide C21H18O13 478.073
113 787.098 14.741 1,2’,3,5-Tetra-O-galloylhamamelofuranose C34H28O22 788.105
114 577.154 17.647 Scutellarein 7,4′-dirhamnoside C27H30O14 578.162
115 939.108 17.171 1,2,3,4,6-Pentakis-O-galloyl-beta-D-glucose C41H32O26 940.115
116 331.081 16.795 2′,3,5-Trihydroxy-5′,7-dimethoxyflavanone C17H16O7 332.088
117 347.037 3.941 2-(α-D-Mannosyl)-3-phosphoglycerate C9H17O12P 348.045
118 315.159 34.910 Isopulegone caffeate C19H24O4 316.166

Figure 1.

Figure 1

LC-MS chromatogram of ethanolic M. fragrans mace extract.

Figure 2.

Figure 2

LC-MS chromatogram of ethanolic A. lancea rhizome extract.

Figure 3.

Figure 3

LC-MS chromatogram of ethanolic extract of Prabchompoothaweep remedy.

3.3. Four-Day Suppressive Test

The antimalarial activity of M. fragrans extract, A. lancea extract, and Prabchompoothaweep remedy extract against P. berghei ANKA was measured using a 4-day suppressive test. Animals in each condition were treated with daily doses of crude extracts at 200, 400, and 600 mg/kg body weight by an oral route. The results showed that mice treated with extracts of M. fragrans and Prabchompoothaweep remedy showed significant suppression of parasitemia in a dose-dependent response (M. fragrans: 38.32, 44.17, and 46.86, respectively; and Prabchompoothaweep remedy: 39.18, 48.35, and 60.11, respectively) compared to the negative control group (p < 0.05). The A. lancea group also showed suppressed parasites compared to the negative control group (p < 0.05), especially at a dose of 400 mg/kg body weight. Parasite levels decreased after treatment with the ethanolic extract of M. fragrans, A. lancea, and Prabchompoothaweep remedy. However, all treatment groups in the crude extract did not completely suppress parasitemia, whereas the parasites were suppressed by more than 95% in the positive control groups (6 mg/kg body weight artesunate and 25 mg/kg body weight chloroquine). Parasite levels and parasite suppression are shown in Table 5.

Table 5.

Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on parasite level and parasite suppression in the 4-day suppressive test.

Group Dose (mg/kg) % Parasitemia % Suppression
7% Tween 80 - 40.45 ± 2.15 b,c,d,e,f,g,h,i,j,k,l -
Artesunate 6 2.18 ± 0.50 a,d,e,f,g,h,i,j,k,l 95.32 ± 0.57 d,e,f,g,h,i,j,k,l
Chloroquine 25 0.27 ± 0.15 a,d,e,f,g,h,i,j,k,l 99.34 ± 0.37 d,e,f,g,h,i,j,k,l
M. fragrans 200 24.94 ± 2.50 a,b,c,h,l 38.32 ± 6.18 b,c,h,i,k,l
400 22.36 ± 1.26 a,b,c,h,l 44.17 ± 3.12 b,c,h,l
600 21.48 ± 0.73 a,b,c,h,l 46.86 ± 1.80 b,c,h,l
A. lancea 200 21.53 ± 2.47 a,b,c,h,l 46.75 ± 6.11 b,c,h,j,k,l
400 16.13 ± 0.41 a,b,c,d,e,f,g,i,j,k 60.09 ± 1.03 b,c,d,e,f,g,i,j,k
600 20.91 ± 1.15 a,b,c,h,l 48.29 ± 2.86 b,c,d,h,l
Prabchompoothaweep remedy 200 24.60 ± 1.03 a,b,c,h,l 39.18 ± 2.56 b,c,h,l
400 20.88 ± 3.08 a,b,c,h,l 48.35 ± 7.62 b,c,d,h,l
600 16.13 ± 0.58 a,b,c,d,e,f,g,i,j,k 60.11 ± 1.44 b,c,d,e,f,g,i,j,k

Data are presented as mean ± SEM (n = 5 per group), p < 0.05. a Compared to the negative control, b compared to artesunate, c compared to chloroquine, d compared to 200 mg/kg of M. fragrans, e compared to 400 mg/kg of M. fragrans, f compared to 600 mg/kg of M. fragrans, g compared to 200 mg/kg of A. lancea, h compared to 400 mg/kg of A. lancea, i compared to 600 mg/kg of A. lancea, j compared to 200 mg/kg of Prabchompoothaweep remedy, k compared to 400 mg/kg of Prabchompoothaweep remedy, and l compared to 600 mg/kg of Prabchompoothaweep remedy.

3.4. PCV

The effects of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on PCVs are presented in Table 6. In the positive treatment control group (artesunate and chloroquine), there was a significant decrease in PCV compared with the negative control group (p > 0.05). The PCV loss was protected by 200, 400, and 600 mg/kg doses of crude extracts compared to the negative control group. However, the protection of crude extracts did not significantly reduce PCV loss at any dose of crude extract compared to the negative control group (p < 0.05).

Table 6.

Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on pack cell volume in the 4-day suppressive test.

Group Dose
(mg/kg)
Day 0 Day 4 % Change
7% Tween 80 - 49.60 ± 1.01 45.00 ± 1.89 −10.35 ± 3.67% b,c
Artesunate 6 52.20 ± 1.32 54.80 ± 1.16 4.74 ± 1.43% a,d,e,f,g,h,i,j,k,l
Chloroquine 25 51.80 ± 0.43 53.40 ± 1.47 2.87 ± 2.36% a,d,j
M. fragrans 200 54.00 ± 0.89 49.80 ± 2.03 −8.58 ± 3.89% b,c
400 51.20 ± 1.16 48.40 ± 0.80 −5.78 ± 1.52% b
600 51.00 ± 1.09 48.80 ± 1.93 −4.64 ± 4.05% b
A. lancea 200 52.00 ± 2.73 49.40 ± 2.17 −5.58 ± 8.67% b
400 52.40 ± 1.01 50.00 ± 1.67 −4.86 ± 2.15% b
600 51.20 ± 1.16 48.80 ± 1.46 −5.01 ± 3.73% b
Prabchompoothaweep remedy 200 52.60 ± 1.35 48.20 ± 2.13 −9.25 ± 3.46% b,c
400 51.80 ± 1.83 49.40 ± 1.35 −4.87 ± 3.02% b
600 50.80 ± 2.63 48.60 ± 1.62 −4.47 ± 2.65% b

Data are presented as mean ± SEM (n = 5 per group), p < 0.05. a Compared to the negative control, b compared to artesunate, c compared to chloroquine, d compared to 200 mg/kg of M. fragrans, e compared to 400 mg/kg of M. fragrans, f compared to 600 mg/kg of M. fragrans, g compared to 200 mg/kg of A. lancea, h compared to 400 mg/kg of A. lancea, i compared to 600 mg/kg of A. lancea, j compared to 200 mg/kg of Prabchompoothaweep remedy, k compared to 400 mg/kg Prabchompoothaweep remedy, and l compared to 600 mg/kg of Prabchompoothaweep remedy.

3.5. Acute Oral Toxicity Test

3.5.1. Physical Activity and Behavior, Food and Water Uptake, and Body Weight

On the first day of the experiment, mice were administered a single dose of 2000 mg/kg M. fragrans ethanolic extract, A. lancea ethanolic extract, or Prabchompoothaweep remedy ethanolic extract. Physical activity and behavioral changes were observed for 14 consecutive days after treatment. The results showed no signs or symptoms of toxicity, such as rigidity, mood changes, ataxia, abnormal sleep, diarrhea, vomiting, consumption changes, and hair erection, during the experiment period. The mice in the acute toxicity test did not show mortality within the first 24 h or 14 days of treatment. Therefore, lethal doses of M. fragrans extracts, A. lancea extracts, or Prabchompoothaweep remedy extracts are greater than 2000 mg/kg body weight. According to water and food consumption in acute toxicity tests after treatment with ethanolic extracts, the mean water and food consumption of mice in the treatment groups treated with a single dose of 2000 mg/kg body weight of M. fragrans extract, A. lancea extract, Prabchompoothaweep remedy extract, and those in the 7% Tween 80 group (negative control group) did not show significant differences compared to those of mice in the control group (untreated group) (p > 0.05) (Table 7). Furthermore, the body weight changes in mice treated with 2000 mg/kg crude extracts and 7% Tween 80 were not significantly different from those in the control group (p > 0.05) (Table 8) at week 2 after receiving crude extracts.

Table 7.

Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on food and water uptake in the acute toxicity test at week 1 and week 2 after treatment.

Food Consumption (g) Week 1 Week 2
Normal mice 25.0 ± 3.5 21.6 ± 2.7
7% Tween 80 22.1 ± 1.7 20.8 ± 2.5
M. fragrans 2000 mg/kg 20.8 ± 2.0 20.7 ± 0.8
A. lancea 2000 mg/kg 22.8 ± 2.5 22.4 ± 2.3
Prabchompoothaweep
remedy 2000 mg/kg
23.6 ± 3.9 22.0 ± 1.9
Water Consumption (mL) Week 1 Week 2
Normal mice 122.2 ± 4.7 125.8 ± 7.9
7% Tween 80 122.4 ± 8.3 126.7 ± 8.3
M. fragrans 2000 mg/kg 122.4 ± 3.5 127.7 ± 3.5
A. lancea 2000 mg/kg 126.0 ± 4.8 130.4 ± 5.0
Prabchompoothaweep
remedy 2000 mg/kg
125.0 ± 2.6 130.5 ± 4.8

Data are presented as mean ± SEM (n = 5 per group).

Table 8.

Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on body weight changes in the acute toxicity test on day 0 and day 14 after treatment.

Group Mean Body Weight
Day 0 Day 14 % Change
Normal mice 33.4 ± 1.5 39.2 ± 2.2 14.6 ± 1.7%
7% Tween 80 32.5 ± 1.4 36.5 ± 1.5 11.1 ± 1.3%
M. fragrans 2000 mg/kg 32.8 ± 1.2 38.0 ± 2.4 13.4 ± 3.3%
A. lancea 2000 mg/kg 33.0 ± 1.6 38.0 ± 2.7 13.0 ± 2.6%
Prabchompoothaweep
remedy 2000 mg/kg
32.1 ± 1.1 36.8 ± 1.4 12.6 ± 1.5%

Data are presented as mean ± SEM (n = 5 per group).

3.5.2. Biochemical Assessment of Liver and Kidney Functions

The levels of liver function, such as AST, ALT, and ALP, in mice that received a single 2000 mg/kg dose of M. fragrans extract, A. lancea extract, Prabchompoothaweep remedy extract, and those of the 7% Tween 80 group (negative control group) did not show statistically significant differences compared to the control group (untreated group) (p > 0.05) at the end of this study. Furthermore, the level of biochemical parameters of kidney functions, such as creatinine and BUN, in mice treated with 2000 mg/kg body weight of crude extracts and 7% Tween 80 showed no significant difference from those in mice in the control group (p > 0.05) (Table 9).

Table 9.

Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on kidney and liver functions in the acute toxicity test.

Parameters Normal Mice 7% Tween 80 M. fragrans A. lancea Prabchompoothaweep Remedy
Liver Function Test
AST (U/L) 83.80 ± 7.13 83.00 ± 9.18 87.75 ± 12.57 94.60 ± 8.77 92.00 ± 5.17
ALT (U/L) 36.80 ± 8.08 38.80 ± 3.70 31.75 ± 6.96 34.60 ± 5.57 34.60 ± 7.05
ALP (U/L) 92.10 ± 11.35 91.04 ± 7.86 90.50 ± 8.96 88.40 ± 7.03 89.20 ± 12.79
Kidney Function Test
BUN (mg/dL) 26.42 ± 3.86 31.04 ± 3.96 25.45 ± 2.30 25.56 ± 3.89 25.26 ± 2.12
Creatinine (mg/dL) 0.66 ± 0.04 0.69 ± 0.07 0.66 ± 0.03 0.65 ± 0.04 0.61 ± 0.06

Data are presented as mean ± SEM (n = 5 per group).

3.5.3. Histological Examination of Liver and Kidney Tissues

Histopathological examination of the liver and kidney samples is shown in Figure 4. The liver tissue morphology of the mice that received a single 2000 mg/kg dose of M. fragrans extract, A. lancea extract, and Prabchompoothaweep remedy extract manifested normal hepatocytes containing a red–pink cytoplasm and normal structures in the hepatic sinusoids and central vein. The sinusoidal vasodilation or inflammatory infiltration was not observed in the H&E staining of the liver tissue. Furthermore, the kidney morphology of the mice treated with a single dose of the crude extract revealed a normal structure of the glomerulus, Bowman’s capsule, and kidney epithelial cells compared to those of the control group (Figure 4f) and the 7% Tween 80 group (Figure 4g).

Figure 4.

Figure 4

Histopathological examination of liver and kidney tissues from ICR mice that administrated with ethanolic extract from M. fragrans, A. lancea, and Prabchompoothaweep remedy in acute toxicity test: (a) histology of the liver tissue of control group, (b) histology of the liver tissue of 7% Tween 80 group, (c) histology of the liver tissue of M. fragrans treated mice, (d) histology of the liver tissue of A. lancea treated mice, (e) histology of the liver tissue of Prabchompoothaweep remedy treated mice, (f) histology of the kidney tissue of control group, (g) histology of the kidney tissue of 7% Tween 80 group, (h) histology of the kidney tissue of M. fragrans treated mice, (i) histology of the kidney tissue of A. lancea treated mice, (j) histology of the kidney tissue of Prabchompoothaweep remedy treated mice. All images were acquired at 20× magnification. Bar = 200 µm. CV, central vein; H, hepatocyte; T, tubule; G, glomerulus.

4. Discussion

Antimalarial treatment remains a public health concern in several countries. The use of traditional medicine that is safe, effective, and cost-efficient is a way to ensure that all patients have access to treatment [8]. From 2014 to 2023, the World Health Organization’s traditional medicine strategy has become popular worldwide and constantly increased each year [27]. Furthermore, natural plants are important sources of bioactive compounds, and many studies have focused on finding new substances to solve the antimalarial drug problem [24]. Therefore, this study focused on natural plants to stimulate the development of a new, effective antimalarial agent. In our previous report, in vitro studies showed that the ethanolic extracts of the mace of M. fragrans, rhizome of A. lancea, and Prabchompoothaweep remedy had anti-plasmodium activity against the P. falciparum K1 strain, with IC50 values of 5.96, 7.37, and 14.13 µg/mL, respectively (unpublished data). All IC50 values of the crude extracts were categorized as a good or promising activity for antimalarial effects [28]. A selectivity index (SI), which is calculated from the ratio between the toxic concentration to human cells (CC50) and the effective concentration to prevent parasite growth (IC50), which is lower than two, indicates the general toxicity of the compound [29]. These results showed that the ethanolic extract of the mace of M. fragrans, rhizome of A. lancea, and Prabchompoothaweep remedy exhibited SI values higher than two. Because the ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy showed strong in vitro therapeutic effects with promising antimalarial activity and low toxicity to human cells, these two plants and one remedy were considered for in vivo antimalarial evaluation in this study. An in vivo model is commonly used to investigate the effects of a prodrug, the elimination of parasites by the immune system and the safety of the drug before processing into the clinical phase [30]. Mouse models have been used to identify a large number of conventional antimalarial agents, including chloroquine, halofantrine, mefloquine, and artemisinin derivatives [31]. In this study, ICR mice were inoculated with the wild-type P. berghei ANKA strain, a common model for the induction of malaria in mice and evaluation of antimalarial effects. The P. berghei ANKA strain is a suitable parasite that has higher accessibility and can sequester within the blood microcirculation. In this study, we used the 4-day suppressive test because it is a commonly used method for testing the antimalarial effects of candidate compounds in early infection. Moreover, this model shows the most reliable parameters, such as percentage of suppression of blood parasitemia [32].

In the present study, the 4-day suppressive test showed inhibition of parasitemia, which showed a high percentage in mice receiving 600 mg/kg M. fragrans (46.86%), 600 mg/kg Prabchompoothaweep remedy (60.11%), while A. lancea showed a high percentage of suppression in mice receiving 400 mg/kg (60.09%). A. lancea showed a high percentage of suppression in mice at a dose of 400 mg/kg because of its immunomodulatory property. Normally, cytokines play a major role in modulating the symptoms of malaria, parasitemia load, and the severity of malaria disease [33]. Moreover, the pro-inflammatory cytokines such as TNF-α, and IL-6 have been associated with severe malaria and death [34]. A previous study found that the low concentration of atractylodin, which is a bioactive compound of A. Lancea, significantly inhibited the expression of both TNF and IL-6, while the high concentration of atractylodin significantly suppressed only IL-6 expression [35]. Consistent with our results, the crude extract was identified as a considered active when parasitemia suppression was more than 30% [31]. Therefore, it can be implied that these crude extracts are active in schizonticide activity against P. berghei ANKA-infected mice. The antimalarial effects of the crude extract are associated with bioactive compounds such as polyphenols, flavonoids, alkaloids, terpenoids, and saponins [36]. Therefore, the antimalarial effect of ethanolic crude extracts could be due to a single or combined mechanism of action of these active compounds [37].

The results of phytochemical screening revealed that the ethanolic extract of M. fragrans, A. lancea, and Prabchompoothaweep remedy is rich in several plant secondary metabolites. M. fragrans extract contained flavonoids, terpenoids, alkaloids, and coumarins, while the extract of A. lancea contained terpenoids, alkaloids, and coumarins, and Prabchompoothaweep remedy contained terpenoid, alkaloids, tannins, and coumarins, all of which are associated with antimalarial activity. These results were consistent with a previous study on secondary plant metabolites. They have shown antimalarial activities posed by the classes of alkaloids, terpenes, flavonoids, xanthones, anthraquinones, phenolic compounds, sesquiterpenes, and other compounds [38,39]. The phytochemical constituents of the ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy may have a single or synergistic effect to provide antimalarial properties through various mechanisms. In this context, flavonoids have been shown to prevent the transportation of L-glutamine and myoinositol into infected red blood cells, which play a role in parasite growth [40], while terpenoids (e.g., artemisinin) may exert their effect by the endoperoxidation that forms potentially toxic heme-adducts. Alkaloids (e.g., quinine) act as antimalarial agents by inhibiting protein synthesis and preventing heme (toxic) from being converted into hemozoin pigments (non-toxic) in parasite food vacuole [41]. Consequently, tannins also exhibit antimalarial effects by scavenging free radicals. Furthermore, coumarin compounds might contribute to antiplasmodial activity by controlling oxidative enzymes, such as superoxide dismutase, and inhibiting DNA synthesis. The antioxidant effects can disrupt heme polymerization, which oxidizes heme before heme polymerization, and unpolymerized heme is toxic to intraerythrocytic parasites [10]. Furthermore, phytochemical constituents, such as steroids, flavonoids, and other components, might act as antimalarial agents not only by directly attacking parasites but also by indirectly modulating the immune system of the host [42]. Therefore, the antiplasmodial activity observed in plants could have been derived from a single or synergistic effect of these metabolites.

Qualitative analysis of M. fragrans mace extracts presented many compounds. M. fragrans is an important source of secondary compounds consisting of coumarin (edulisin III), and flavonoids (kaempferol, (7′x,8′x)-4,7′-epoxy-3,8′-bilign-7-ene-3,5′-dimethoxy-4′,9,9′-triol), including citric acid and propyl 2-furanacrylate (fatty acid esters). Analysis of A. lancea rhizome extracts revealed the presence of polyphenols (chlorogenic acid), hydroxyanthraquinones (1,2,6,8-Tetrahydroxy-3-methylanthraquinone 2-O-b-D-glucoside), sesquiterpene lactone (taraxacolide 1-O-b-D-glucopyranoside), and salicylic acid. Furthermore, the Prabchompoothaweep remedy extracts allowed us to putatively identify 10 major peaks. The results showed the presence of flavonoid (luteolin), coumarins (6′-O-formylmarmin), and phenolic compounds (caffeic acid, eudesmic acid, gallic acid, and ellagic acid) constituents in the remedy. Some constituents analyzed by LC-MS are biologically active compounds. Luteolin has been shown to possess anti-inflammatory, antiallergy, anticancer, and antioxidant activity [43]. In addition, vanillic acid has been shown to exhibit anti-inflammatory and antioxidant effects both in vitro and in vivo in a carrageenan-induced inflammation model, and it has also shown anticancer, antifungal, antibacterial, and anti-viral effects [44,45]. Kaempferol has several pharmacological effects, including antioxidant and antibacterial activities [46]. Caffeic acid has potential as an antioxidant, anti-inflammatory, and antineoplastic agent [47]. Gallic acid exhibits antibacterial, anticancer, and antiplasmodial activities [48]. Among the identified compounds, ellagic acid has been reported to have anti-plasmodium properties. A previous study by Verotta et al. found that ellagic acid isolated from Tristaniopsis callobuxus (Myrtaceae) showed significant antiplasmodial activity against the resistant strain of Plasmodium, with an IC50 between 0.331 and 0.480 µM [49]. A study by Banzouzi et al. suggested that ellagic acid has also shown anti-plasmodium in mice infected with Plasmodium vinckei pettri using the Peter’s test [50]. Furthermore, Soh et al. found that ellagic acid inhibits parasitemia in a dose-dependent manner, with 50% suppression in mice receiving 1 mg/kg and 100% suppression in mice receiving 50 and 100 mg/kg via the intraperitoneal route [51]. In addition to antimalarial activity, ellagic acid also shows antioxidant properties and anti-inflammation that could prolong the survival rate after the administration of T. albida in experimental cerebral malaria (ECM) [52]. Flavonoid compounds also have antimalarial effects by stimulating the immune system, inhibiting the synthesis of fatty acids in parasites and preventing protein synthesis [53]. Therefore, the selected crude extract of the Prabchompoothaweep remedy might be responsible, at least partially, for antimalarial property, which is produced by a single phytoconstituent or the synergistic effect of these compounds, as mentioned above. However, further studies are needed to isolate, identify, and characterize active compounds, as well as to understand the mechanism of inhibition.

A decrease in PCV is one of the characteristics of malaria infection in mice. PCV was determined to investigate the effectiveness of the ethanolic crude extract in inhibiting erythrocyte damage caused by an increase in parasitemia [54]. To prevent PCV reduction, plants with antimalarial activity are expected to maintain PCV during mouse infection. Surprisingly, in a 4-day suppressive test, the ethanolic extract of M. fragrans, A. lancea, and Prabchompoothaweep remedy at all doses prevented PCV loss compared to the negative control group. It is possible that phenols and other metabolites in plants have antioxidant effects and membrane protection. Phenolic compounds have excellent antioxidant effects due to their hydroxyl groups, which can donate electrons to reactive oxygen species (ROS) [55]. The protective effect of the crude extracts was consistent with the results of studies by Wannang et al. [56], Saba et al. [57], and Misganaw et al. [58]. Moreover, the prevention of PCV reduction may be due to the absence of saponins in this crude extract. Normally, saponins act as phytodetergents, leading to cholesterol release from the cell membrane and promoting the permeability of the red blood cell membrane with strong hemolytic activity [59]. The effect of the plant extract on PCV loss may be due to the elimination of parasites from infected erythrocytes before hemolysis. Furthermore, the activation of the immune system and release of free radicals and ROS caused by malaria infection contribute to the degradation of hemoglobin and development of anemia [60]. In addition, the antioxidant activity of crude extracts, especially polyphenolic compounds, may protect red blood cells (RBCs) from ROS and promote the survival rate of both normal and infected RBCs during malaria infection.

The toxicity of the plant extracts was assessed using an oral acute toxicity test. Under these conditions, the mice received a single dose of 2000 mg/kg of the ethanolic extract of M. fragrans, A. lancea, and Prabchompoothaweep remedy, where a single high dose is suggested for acute toxicity testing [23]. The results of acute toxicity of all crude extracts revealed that there was no mortality and no signs of toxicity over 14 days. Therefore, the approximate median lethal dose (LD50) of the crude extracts was greater than 2000 mg/kg. According to the OECD’s Globally Harmonized System of Classification, crude extracts presented a low acute toxicity hazard with a category 5 classification. The results observed in the acute toxicity study with M. fragrans and A. lancea remedies are consistent with those of a previous study, indicating the safety profiles of this crude extract in a broad range of dose levels (1000−5000 mg/kg body weight) [17,61]. Food and water uptake were recorded to monitor toxicity because these parameters can be used to identify the harmful effects of crude extracts [62]. These results indicated that food and water consumption were not significantly different between the treatment and control groups. Body weight loss is a sensitive toxicity index after exposure to toxic compounds. In the acute toxicity test study, all treatments with M. fragrans, A. lancea, and Prabchompoothaweep remedy did not show significant differences (p < 0.05) in body weight loss compared with the control group on day 14. This observation suggests that the crude extracts did not disturb metabolism in these animals. In addition, the functions of the liver and kidneys were examined using biochemical analyses. Liver abnormalities were indicated by AST, ALT, and ALP levels. Damage to liver cells depends on the levels of AST, ALT, and ALP [63,64]. In all liver marker enzyme activities assessed, AST, ALT, and ALP levels were not significantly different between the treatment groups and the untreated control group. The levels of BUN and creatinine were analyzed for kidney function [65]. These results show that the levels of BUN and creatinine in mice receiving ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy have normal functions in kidney organs that were not different from those in the untreated control group. Additionally, histopathological evaluation of kidney and liver tissue after treatment with the ethanolic extract of M. fragrans, A. lancea, and Prabchompoothaweep remedy did not show any abnormalities. Therefore, the results suggest that oral administration of crude extracts is neither harmful nor unsafe.

5. Conclusions

This study is the first to report the antimalarial activity of ethanolic extracts of M. fragrans mace and A. lancea rhizomes in a mouse model. The ethanolic crude extracts contained several phytoconstituents with important medicinal properties and antimalarial activity. The extracts significantly suppressed parasitemia. Moreover, the crude extracts also showed no adverse health effects on behavioral changes or liver or kidney function in the acute toxicity test. The overall results of this study illustrated that the use of rhizome extracts of A. lancea at 400 mg/kg body weight and extract of Prabchompoothaweep remedy at 600 mg/kg body weight could be developed as a new antimalarial drug treatment. More studies are required to isolate and identify the active compounds and to understand their mechanism of action.

Acknowledgments

This work was supported by Walailak University Ph.D. Scholarships for High-Potential Candidates to Enroll in Doctoral Programs (Contract No. HP005/2021).

Author Contributions

Conceptualization, W.P., P.C. and C.P.; methodology, W.P., A.P., P.C. and C.P.; formal analysis, W.P., P.C. and C.P.; investigation, W.P., A.P., P.C. and C.P.; resources, P.C. and C.P.; data curation, W.P., P.C., A.W.S. and C.P.; writing—original draft preparation, W.P.; writing—review and editing, P.C., A.W.S. and C.P.; visualization, W.P., P.C., A.P. and C.P.; supervision, P.C. and C.P.; project administration, C.P.; funding acquisition, W.P. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

The study protocol was reviewed and approved by the Human Ethics Committee of Walailak University before recruitment (approval number: WUEC-22152-01) and followed the tenets of the Declaration of Helsinki. Written informed consent was obtained from all participants before data and blood sample collection. Animal care requirements were considered during the experiments as required by the National Guidelines for Handling Laboratory Animals. The 4-day suppressive test and oral acute toxicity tests were approved and authorized with permit numbers (WU-ACUC-64027) from the Walailak University Ethical Review Committee before carrying out the experiments.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data associated with this study have been included in this published article. Additional files are available from the corresponding authors upon request.

Conflicts of Interest

The authors declare no competing interests regarding the publication of this paper.

Funding Statement

This research was financially supported by Walailak University Graduate Research Fund (contract no. 2022/07).

Footnotes

Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Moxon C.A., Gibbins M.P., McGuinness D., Milner D.A., Marti M. New Insights into Malaria Pathogenesis. Annu. Rev. Pathol. 2020;15:315–343. doi: 10.1146/annurev-pathmechdis-012419-032640. [DOI] [PubMed] [Google Scholar]
  • 2.World Health Organization World Malaria Report. 2021. [(accessed on 20 May 2022)]. Available online: https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2021.
  • 3.World Health Organization Artemisinin Resistance and Artemisinin-Based Combination Therapy Efficacy: Status Report. 2018. [(accessed on 20 May 2022)]. Available online: https://apps.who.int/iris/handle/10665/274362.
  • 4.Ashley E.A., Dhorda M., Fairhurst R.M., Amaratunga C., Lim P., Suon S., Sreng S., Anderson J.M., Mao S., Sam B., et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N. Engl. J. Med. 2014;371:411–423. doi: 10.1056/NEJMoa1314981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Nsanzabana C. Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug! Trop. Med. Infect. Dis. 2019;4:26. doi: 10.3390/tropicalmed4010026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fairhurst R.M., Dondorp A.M. Artemisinin-resistant Plasmodium falciparum malaria. Microbiol. Spectr. 2016;4:409–429. doi: 10.1128/microbiolspec.EI10-0013-2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.World Health Oragnization World Malaria Report. 2019. [(accessed on 20 May 2022)]. Available online: https://www.who.int/publications/i/item/9789241565721.
  • 8.Haidara M., Haddad M., Denou A., Marti G., Bourgeade-Delmas S., Sanogo R., Bourdy G., Aubouy A. In vivo validation of anti-malarial activity of crude extracts of Terminalia macroptera, a Malian medicinal plant. Malar. J. 2018;17:68. doi: 10.1186/s12936-018-2223-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mojab F. Antimalarial natural products: A review. Avicenna J. Phytomed. 2012;2:52–62. [PMC free article] [PubMed] [Google Scholar]
  • 10.Alehegn A.A., Yesuf J.S., Birru E.M. Antimalarial activity of crude extract and solvent fractions of the leaves of Bersama abyssinica fresen. (Melianthaceae) against Plasmodium berghei infection in Swiss albino mice. Evid.-Based Complement. Altern. Med. 2020;2020:9467359. doi: 10.1155/2020/9467359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Leangpanich S., Itharat A., Chanvimalueng W., Mukkasombat N. A preliminary study on efficacy of Prapchompoothaweep remedy for treatment of allergic rhinitis patients and their quality of life after the treatment. TMJ. 2019;19:537–546. [Google Scholar]
  • 12.Jai-aue A., Makchuchit S., Juckmeta T., Itharat A. Anti-allergic, anti-inflammatory and antioxidant activities of the different extracts of Thai traditional remedy called prabchompoothaweep for allergic rhinitis treatment. J. Med. Assoc. Thail. 2014;97:140–1488. [PubMed] [Google Scholar]
  • 13.Ha M.T., Vu N.K., Tran T.H., Kim J.A., Woo M.H., Min B.S. Phytochemical and pharmacological properties of Myristica fragrans Houtt.: An updated review. Arch. Pharm. Res. 2020;43:1067–1092. doi: 10.1007/s12272-020-01285-4. [DOI] [PubMed] [Google Scholar]
  • 14.Arumugam G., Purushotham B., Swamy M.K. Natural Bio-Active Compounds. Springer; New York, NY, USA: 2019. Myristica fragrans Houtt.: Botanical, pharmacological, and toxicological aspects; pp. 81–106. [Google Scholar]
  • 15.Latha P., Sindhu P., Suja S., Geetha B., Pushpangadan P., Rajasekharan S. Pharmacology and chemistry of Myristica fragrans Houtt.—A review. JOSAC. 2005;14:94–101. [Google Scholar]
  • 16.Zhang W.-J., Zhao Z.-Y., Chang L.-K., Cao Y., Wang S., Kang C.-Z., Wang H.-Y., Zhou L., Huang L.-Q., Guo L.-P. Atractylodis Rhizoma: A review of its traditional uses, phytochemistry, pharmacology, toxicology and quality control. J. Ethnopharmacol. 2021;266:113415. doi: 10.1016/j.jep.2020.113415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Koonrungsesomboon N., Na-Bangchang K., Karbwang J. Therapeutic potential and pharmacological activities of Atractylodes lancea (Thunb.) DC. Asian Pac. J. Trop. Med. 2014;7:421–428. doi: 10.1016/S1995-7645(14)60069-9. [DOI] [PubMed] [Google Scholar]
  • 18.Na-Bangchang K., Kulma I., Plengsuriyakarn T., Tharavanij T., Kotawng K., Chemung A., Muhamad N., Karbwang J. Phase I clinical trial to evaluate the safety and pharmacokinetics of capsule formulation of the standardized extract of Atractylodes lancea. J. Tradit. Complement. Med. 2021;11:343–355. doi: 10.1016/j.jtcme.2021.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Malar C.G.R., Chellaram C. Phytochemical Screening, Quantification of Total Phenols, Total Flavonoids and Antimicrobial Activity of Stem Extracts of Salacia Oblonga. Indian J. Sci. Technol. 2018;11:1–8. doi: 10.17485/ijst/2018/v11i23/125632. [DOI] [Google Scholar]
  • 20.Shad A.A., Ahmad S., Ullah R., AbdEl-Salam N.M., Fouad H., Rehman N.U., Hussain H., Saeed W. Phytochemical and biological activities of four wild medicinal plants. Sci. World J. 2014;2014:857363. doi: 10.1155/2014/857363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Shaikh J.R., Patil M. Qualitative tests for preliminary phytochemical screening: An overview. Int. J. Chem. Stud. 2020;8:603–608. doi: 10.22271/chemi.2020.v8.i2i.8834. [DOI] [Google Scholar]
  • 22.Peters W., Portus J., Robinson B. The chemotherapy of rodent malaria, XXII: The value of drug-resistant strains of P. berghei in screening for blood schizontocidal activity. Ann. Trop. Med. Parasitol. 1975;69:155–171. doi: 10.1080/00034983.1975.11686997. [DOI] [PubMed] [Google Scholar]
  • 23.Organisation for Economic Co-operation and Development (OECD) Test No. 425: Acute Oral Toxicity: Up-and-Down Procedure. OECD Publishing; Paris, France: 2008. [Google Scholar]
  • 24.Phuwajaroanpong A., Chaniad P., Horata N., Muangchanburee S., Kaewdana K., Punsawad C. In vitro and in vivo antimalarial activities and toxicological assessment of Pogostemon cablin (Blanco) Benth. J. Evid.-Based Integr. Med. 2020;25:2515690X20978387. doi: 10.1177/2515690X20978387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Viriyavejakul P., Khachonsaksumet V., Punsawad C. Liver changes in severe Plasmodium falciparum malaria: Histopathology, apoptosis and nuclear factor kappa B expression. Malar. J. 2014;13:106. doi: 10.1186/1475-2875-13-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wichapoon B., Punsawad C., Viriyavejakul P. Expression of cleaved caspase-3 in renal tubular cells in Plasmodium falciparum malaria patients. Nephrology. 2017;22:79–84. doi: 10.1111/nep.12715. [DOI] [PubMed] [Google Scholar]
  • 27.World Health Organization WHO Traditional Medicine Strategy: 2014–2023. 2013. [(accessed on 3 June 2022)]. Available online: https://www.who.int/publications/i/item/9789241506096.
  • 28.Kaharudin F.A., Zohdi R.M., Mukhtar S.M., Sidek H.M., Bihud N.V., Rasol N.E., Ahmad F.B., Ismail N.H. In vitro antiplasmodial and cytotoxicity activities of crude extracts and major compounds from Goniothalamus lanceolatus. J. Ethnopharmacol. 2020;254:112657. doi: 10.1016/j.jep.2020.112657. [DOI] [PubMed] [Google Scholar]
  • 29.Koch A., Tamez P., Pezzuto J., Soejarto D. Evaluation of plants used for antimalarial treatment by the Maasai of Kenya. J. Ethnopharmacol. 2005;101:95–99. doi: 10.1016/j.jep.2005.03.011. [DOI] [PubMed] [Google Scholar]
  • 30.Haddadi M., Mousavi M.J., Mohseni S., Mardani G. In vitro ADME Screening Instead of in vivo Studies in Preclinical Safety. Biomed. J. Sci. Tech. Res. 2020;24:18371–18376. doi: 10.26717/BJSTR.2020.24.004071. [DOI] [Google Scholar]
  • 31.Fidock D.A., Rosenthal P.J., Croft S.L., Brun R., Nwaka S. Antimalarial drug discovery: Efficacy models for compound screening. Nat. Rev. Drug Discov. 2004;3:509–520. doi: 10.1038/nrd1416. [DOI] [PubMed] [Google Scholar]
  • 32.Mekonnen L.B. In vivo antimalarial activity of the crude root and fruit extracts of Croton macrostachyus (Euphorbiaceae) against Plasmodium berghei in mice. J. Tradit. Complement. Med. 2015;5:168–173. doi: 10.1016/j.jtcme.2014.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Farrington L., Vance H., Rek J., Prahl M., Jagannathan P., Katureebe A., Arinaitwe E., Kamya M.R., Dorsey G., Feeney M.E. Both inflammatory and regulatory cytokine responses to malaria are blunted with increasing age in highly exposed children. Malar. J. 2017;16:499. doi: 10.1186/s12936-017-2148-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Afolayan F.I.D., Adegbolagun O., Mwikwabe N.N., Orwa J., Anumudu C. Cytokine modulation during malaria infections by some medicinal plants. Sci. Afr. 2020;8:e00428. doi: 10.1016/j.sciaf.2020.e00428. [DOI] [Google Scholar]
  • 35.Kulma I., Panrit L., Plengsuriyakarn T., Chaijaroenkul W., Warathumpitak S., Na-Bangchang K. A randomized placebo-controlled phase I clinical trial to evaluate the immunomodulatory activities of Atractylodes lancea (Thunb) DC. in healthy Thai subjects. BMC Complement. Med. Ther. 2021;21:61. doi: 10.1186/s12906-020-03199-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ungogo M.A., Ebiloma G.U., Ichoron N., Igoli J.O., de Koning H.P., Balogun E.O. A review of the antimalarial, antitrypanosomal, and antileishmanial activities of natural compounds Isolated from nigerian flora. Front. Chem. 2020;8:617448. doi: 10.3389/fchem.2020.617448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rasoanaivo P., Wright C.W., Willcox M.L., Gilbert B. Whole plant extracts versus single compounds for the treatment of malaria: Synergy and positive interactions. Malar. J. 2011;10:S4. doi: 10.1186/1475-2875-10-S1-S4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mazid M., Khan T., Mohammad F. Role of secondary metabolites in defense mechanisms of plants. Biol. Med. 2011;3:232–249. [Google Scholar]
  • 39.Batista R., De Jesus Silva A., Jr., De Oliveira A.B. Plant-derived antimalarial agents: New leads and efficient phytomedicines. Part II. Non-alkaloidal natural products. Molecules. 2009;14:3037–3072. doi: 10.3390/molecules14083037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Saxena M., Saxena J., Nema R., Singh D., Gupta A. Phytochemistry of medicinal plants. J. Pharmacogn. Phytochem. 2013;1:168–182. [Google Scholar]
  • 41.Soares J.B.R.C., Menezes D., Vannier-Santos M.A., Ferreira-Pereira A., Almeida G.T., Venancio T.M., Verjovski-Almeida S., Zishiri V.K., Kuter D., Hunter R., et al. Interference with hemozoin formation represents an important mechanism of schistosomicidal action of antimalarial quinoline methanols. PLoS Negl. Trop. Dis. 2009;3:e477. doi: 10.1371/journal.pntd.0000477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Masihi K.N. Fighting infection using immunomodulatory agents. Expert Opin. Biol. Ther. 2001;1:641–653. doi: 10.1517/14712598.1.4.641. [DOI] [PubMed] [Google Scholar]
  • 43.Lin Y., Shi R., Wang X., Shen H.-M. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr. Cancer Drug Targets. 2008;8:634–646. doi: 10.2174/156800908786241050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Arya S.S., Rookes J.E., Cahill D.M., Lenka S.K. Vanillin: A review on the therapeutic prospects of a popular flavouring molecule. Adv. Trad. Med. 2021;21:1–17. doi: 10.1007/s13596-020-00531-w. [DOI] [Google Scholar]
  • 45.Tai A., Sawano T., Yazama F., Ito H. Evaluation of antioxidant activity of vanillin by using multiple antioxidant assays. Biochim. Biophys. Acta Gen. Subj. 2011;1810:170–177. doi: 10.1016/j.bbagen.2010.11.004. [DOI] [PubMed] [Google Scholar]
  • 46.Calderón-Montaño J.M., Burgos-Morón E., Pérez-Guerrero C., López-Lázaro M. A review on the dietary flavonoid kaempferol. Mini-Rev. Med. Chem. 2011;11:298–344. doi: 10.2174/138955711795305335. [DOI] [PubMed] [Google Scholar]
  • 47.Aijaz M., Keserwani N., Yusuf M., Ansari N.H., Ushal R., Kalia P. Chemical, biological, and pharmacological prospects of caffeic acid. Biointerface Res. Appl. Chem. 2022;13:324. doi: 10.33263/BRIAC134.324. [DOI] [Google Scholar]
  • 48.Chaniad P., Phuwajaroanpong A., Plirat W., Techarang T., Chukaew A., Punsawad C. In vivo assessment of the antimalarial activity and acute oral toxicity of an ethanolic seed extract of Spondias pinnata (Lf) Kurz. BMC Complement. Med. Ther. 2022;22:72. doi: 10.1186/s12906-022-03546-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Verotta L., Dell’Agli M., Giolito A., Guerrini M., Cabalion P., Bosisio E. In vitro antiplasmodial activity of extracts of Tristaniopsis species and identification of the active constituents: Ellagic acid and 3, 4, 5-trimethoxyphenyl-(6′-O-galloyl)-O-β-D-glucopyranoside. J. Nat. Prod. 2001;64:603–607. doi: 10.1021/np000306j. [DOI] [PubMed] [Google Scholar]
  • 50.Banzouzi J.-T., Prado-Gotor R., Menan H., Valentin A., Roumestan C., Mallie M., Pelissier Y., Blache Y. In vitro antiplasmodial activity of extracts of Alchornea cordifolia and identification of an active constituent: Ellagic acid. J. Ethnopharmacol. 2002;81:399–401. doi: 10.1016/S0378-8741(02)00121-6. [DOI] [PubMed] [Google Scholar]
  • 51.Soh P.N., Witkowski B., Olagnier D., Nicolau M.-L., Garcia-Alvarez M.-C., Berry A., Benoit-Vical F. In vitro and in vivo properties of ellagic acid in malaria treatment. Antimicrob. Agents Chemother. 2009;53:1100–1106. doi: 10.1128/AAC.01175-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Abiodun O.O., Rodríguez-Nogales A., Algieri F., Gomez-Caravaca A.M., Segura-Carretero A., Utrilla M.P., Rodriguez-Cabezas M.E., Galvez J. Antiinflammatory and immunomodulatory activity of an ethanolic extract from the stem bark of Terminalia catappa L. (Combretaceae): In vitro and in vivo evidences. J. Ethnopharmacol. 2016;192:309–319. doi: 10.1016/j.jep.2016.07.056. [DOI] [PubMed] [Google Scholar]
  • 53.Kırmızıbekmez H., Çalıs I., Perozzo R., Brun R., Dönmez A.A., Linden A., Rüedi P., Tasdemir D. Inhibiting activities of the secondary metabolites of Phlomis brunneogaleata against parasitic protozoa and plasmodial enoyl-ACP reductase, a crucial enzyme in fatty acid biosynthesis. Planta Med. 2004;70:711–717. doi: 10.1055/s-2004-827200. [DOI] [PubMed] [Google Scholar]
  • 54.Okokon J., Ita B., Udokpoh A. The in-vivo antimalarial activities of Uvaria chamae and Hippocratea africana. Ann. Trop. Med. Parasitol. 2006;100:585–590. doi: 10.1179/136485906X118512. [DOI] [PubMed] [Google Scholar]
  • 55.Nicholas S. Antioxidants and Reactive Oxygen Species in Plants. Wiley Online Library; Hoboken, NJ, USA: 2008. [Google Scholar]
  • 56.Wannang N.N., Jimam N.S., Omale S., Dapar M.L., Gyang S.S., Aguiyi J.C. Effects of Cucumis metuliferus (Cucurbitaceae) fruits on enzymes and haematological parameters in albino rats. Afr. J. Biotechnol. 2007;6:2515–2518. doi: 10.5897/AJB2007.000-2400. [DOI] [Google Scholar]
  • 57.Saba A.B., Oridupa O.A., Ofuegbe S.O. Evaluation of haematological and serum electrolyte changes in Wistar rats administered with ethanolic extract of whole fruit of Lagenaria breviflora Robert. J. Med. Plants Res. 2009;3:758–762. [Google Scholar]
  • 58.Misganaw D., Engidawork E., Nedi T. Evaluation of the anti-malarial activity of crude extract and solvent fractions of the leaves of Olea europaea (Oleaceae) in mice. BMC Complement. Altern. Med. 2019;19:171. doi: 10.1186/s12906-019-2567-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kaur K., Jain M., Kaur T., Jain R. Antimalarials from nature. Bioorg. Med. Chem. 2009;17:3229–3256. doi: 10.1016/j.bmc.2009.02.050. [DOI] [PubMed] [Google Scholar]
  • 60.Greve B., Kremsner P., Lell B., Luckner D., Schmid D. Malarial anaemia in African children associated with high oxygen-radical production. Lancet. 2000;355:40–41. doi: 10.1016/s0140-6736(99)04761-3. [DOI] [PubMed] [Google Scholar]
  • 61.Anaduaka E.G., Okagu I.U., Uchendu N.O., Ezeanyika L.U.S., Nwanguma B.C. Hepato-renal toxicity of Myristica fragrans Houtt. (Myristicaceae) seed extracts in rats. J. King Saud Univ. Sci. 2022;34:101694. doi: 10.1016/j.jksus.2021.101694. [DOI] [Google Scholar]
  • 62.Osagie-Eweka S.E., Orhue N.E.J., Omogbai E.K.I., Amaechina F.C. Oral acute and sub-chronic toxicity assessment of aqueous leaf extract of Simarouba glauca DC (Paradise tree) Toxicol. Rep. 2021;8:239–247. doi: 10.1016/j.toxrep.2021.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Anosike C.A., Ugwu U.B., Nwakanma O. Effect of ethanol extract of Pyrenacantha staudtii leaves on carbontetrachloride induced hepatotoxicity in rats. Biokemistri. 2008;20:17–22. doi: 10.4314/biokem.v20i1.56433. [DOI] [Google Scholar]
  • 64.Nigatu T.A., Afework M., Urga K., Ergete W., Makonnen E. Toxicological in vestigation of acute and chronic treatment with Gnidia stenophylla Gilg root extract on some blood parameters and histopathology of spleen, liver and kidney in mice. BMC Res. Notes. 2017;10:625. doi: 10.1186/s13104-017-2964-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Reddy M.K., Gupta S.K., Jacob M.R., Khan S.I., Ferreira D. Antioxidant, antimalarial and antimicrobial activities of tannin-rich fractions, ellagitannins and phenolic acids from Punica granatum L. Planta Med. 2007;53:461–467. doi: 10.1055/s-2007-967167. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data associated with this study have been included in this published article. Additional files are available from the corresponding authors upon request.


Articles from Tropical Medicine and Infectious Disease are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES