Next Article in Journal / Special Issue
Safety and Tolerability of an Antimalarial Herbal Remedy in Healthy Volunteers: An Open-Label, Single-Arm, Dose-Escalation Study on Maytenus senegalensis in Tanzania
Previous Article in Journal
Assessment of Post-Dengue Rheumatic Symptoms Using the WOMAC and DAS-28 Questionnaires in a Honduran Population after a Four-Month Follow-Up
Previous Article in Special Issue
Exploring Potential Antimalarial Candidate from Medicinal Plants of Kheaw Hom Remedy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

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

by
Walaiporn Plirat
1,2,
Prapaporn Chaniad
1,2,
Arisara Phuwajaroanpong
1,2,
Abdi Wira Septama
3 and
Chuchard Punsawad
1,2,*
1
Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
2
Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
3
Research Center for Pharmaceutical Ingredient and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Bogor 16915, Indonesia
*
Author to whom correspondence should be addressed.
Trop. Med. Infect. Dis. 2022, 7(12), 395; https://doi.org/10.3390/tropicalmed7120395
Submission received: 23 October 2022 / Revised: 16 November 2022 / Accepted: 21 November 2022 / Published: 23 November 2022
(This article belongs to the Special Issue Advances in Malaria Treatment and Prevention)

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.

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   cells number   of   total   red   blood   cells
The percentage of parasitemia suppression was calculated using the following formula:
% suppression = [ A B ] 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).

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.

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.

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).

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.

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).

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).

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.

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.

Funding

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

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.

Acknowledgments

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

Conflicts of Interest

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

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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. World Health Organization. World Malaria Report. 2021. Available online: https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2021 (accessed on 20 May 2022).
  3. World Health Organization. Artemisinin Resistance and Artemisinin-Based Combination Therapy Efficacy: Status Report. 2018. Available online: https://apps.who.int/iris/handle/10665/274362 (accessed on 20 May 2022).
  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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nsanzabana, C. Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug! Trop. Med. Infect. Dis. 2019, 4, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Fairhurst, R.M.; Dondorp, A.M. Artemisinin-resistant Plasmodium falciparum malaria. Microbiol. Spectr. 2016, 4, 409–429. [Google Scholar] [CrossRef] [Green Version]
  7. World Health Oragnization. World Malaria Report. 2019. Available online: https://www.who.int/publications/i/item/9789241565721 (accessed on 20 May 2022).
  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. [Google Scholar] [CrossRef] [Green Version]
  9. Mojab, F. Antimalarial natural products: A review. Avicenna J. Phytomed. 2012, 2, 52–62. [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. [Google Scholar] [CrossRef]
  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. [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. [Google Scholar] [CrossRef]
  14. Arumugam, G.; Purushotham, B.; Swamy, M.K. Myristica fragrans Houtt.: Botanical, pharmacological, and toxicological aspects. In Natural Bio-Active Compounds; Springer: New York, NY, USA, 2019; 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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  21. Shaikh, J.R.; Patil, M. Qualitative tests for preliminary phytochemical screening: An overview. Int. J. Chem. Stud. 2020, 8, 603–608. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [PubMed]
  27. World Health Organization. WHO Traditional Medicine Strategy: 2014–2023. 2013. Available online: https://www.who.int/publications/i/item/9789241506096 (accessed on 3 June 2022).
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  42. Masihi, K.N. Fighting infection using immunomodulatory agents. Expert Opin. Biol. Ther. 2001, 1, 641–653. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [Green Version]
  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. [Google Scholar] [CrossRef] [Green Version]
  59. Kaur, K.; Jain, M.; Kaur, T.; Jain, R. Antimalarials from nature. Bioorg. Med. Chem. 2009, 17, 3229–3256. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
Figure 1. LC-MS chromatogram of ethanolic M. fragrans mace extract.
Figure 1. LC-MS chromatogram of ethanolic M. fragrans mace extract.
Tropicalmed 07 00395 g001
Figure 2. LC-MS chromatogram of ethanolic A. lancea rhizome extract.
Figure 2. LC-MS chromatogram of ethanolic A. lancea rhizome extract.
Tropicalmed 07 00395 g002
Figure 3. LC-MS chromatogram of ethanolic extract of Prabchompoothaweep remedy.
Figure 3. LC-MS chromatogram of ethanolic extract of Prabchompoothaweep remedy.
Tropicalmed 07 00395 g003
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.
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.
Tropicalmed 07 00395 g004
Table 1. Phytochemical screening of ethanolic extract of M. fragrans mace, A. lancea rhizome, and Prabchompoothaweep remedy.
Table 1. Phytochemical screening of ethanolic extract of M. fragrans mace, A. lancea rhizome, and Prabchompoothaweep remedy.
Phytochemical ConstituentsM. fragransA. lanceaPrabchompoothaweep Remedy
Flavonoid+--
Terpenoids+++
Alkaloids+++
Tannins--+
Anthraquinones---
Cardiac glycosides---
Saponins---
Coumarins+-+
(+), detected; (-), not detected phytochemical constituents.
Table 2. Compounds identified in the ethanolic M. fragrans extract by LC-QTOF-MS.
Table 2. Compounds identified in the ethanolic M. fragrans extract by LC-QTOF-MS.
No.M/ZRT
(min)
CompoundsMolecular
Formula
Molecular Weight
1133.0142.087Malic acidC4H6O5134.021
2149.0091.886Tartaric acidC4H6O6150.016
3285.04027.493LuteolinC15H10O6286.047
4357.13427.969(7′x,8′x)-4,7′-Epoxy-3,8′-bilign-7-ene-3,5′-dimethoxy-4′,9,9′-triolC20H22O6358.141
5201.14931.8523-Hydroxynonyl acetateC11H22O3202.156
6265.0562.1360Monoglyceride citrateC9H14O9266.063
7373.16538.781SonchifolinC21H26O6374.172
8345.13431.690Gibberellin A92C19H22O6346.141
9161.0454.4293-Hydroxy-3-methyl-glutaric acidC6H10O5162.052
10179.07115.867Propyl 2-furanacrylateC10H12O3180.078
11207.06618.673Sinapyl aldehydeC11H12O4208.073
12219.0505.2941-Hydroxypentane-1,2,5-tricarboxylateC8H12O7220.058
13329.10335.937Isoamericanol AC18H18O6330.110
14299.09233.5812,4-Dihydroxy-6,4′-dimethoxychalconeC17H16O5300.099
15167.0349.353Dihydroxyphenylacetic acidC8H8O4168.042
16183.10234.258Ascariadole epoxideC10H16O3184.109
17375.14431.978alpha-PeroxyachifolideC20H24O7376.152
18191.0192.124Citric acidC6H8O7192.026
19287.05521.1913′,4′,5,7-TetrahydroxyisoflavanoneC15H12O6288.063
20149.06026.2902-(2-Furanyl)-3-methyl-2-butenalC9H10O2150.067
21329.13936.576Tetrahydrosappanone A trimethyl etherC19H22O5330.146
22371.18634.358TanabalinC22H28O5372.193
23315.12339.7455′-Hydroxy-3′,4′,7-trimethoxyflavanC18H20O5316.130
24265.14438.918IsoleptospermoneC15H22O4266.151
25237.11320.077Benzyl b-L-arabinopyranosideC13H18O4238.120
26301.03527.794HieracinC15H10O7302.042
27285.04032.491KaempferolC15H10O6286.047
28389.16039.444Rosmic acidC21H26O7390.167
29271.06029.798MethylnorlichexanthoneC15H12O5272.068
30267.0711.9362(α-D-Mannosyl)-D-glycerateC9H16O9268.078
31177.0403.139L-SorbosoneC6H10O6178.047
32303.05016.481(±)-TaxifolinC15H12O7304.058
33177.0199.265EsculetinC9H6O4178.026
34359.14935.8996′-O-FormylmarminC20H24O6360.156
35387.14431.364Edulisin IIIC21H24O7388.151
36331.11823.7095′,8-Dihydroxy-3′,4′,7-trimethoxyflavanC18H20O6332.125
37359.07634.408JaceidinC18H16O8360.083
38271.06031.401(±)-NaringeninC15H12O5272.067
39201.1126.8472,6-Dimethyl-1,8-octanedioic acidC10H18O4202.120
40329.23233.2439S,10S,11R-trihydroxy-12Z-octadecenoic acidC18H34O5330.240
41311.12839.657Gancaonin VC19H20O4312.135
42117.0182.688Succinic acidC4H6O4118.026
43163.03916.068m-Coumaric acidC9H8O3164.047
44197.0459.9042-Hydroxy-3,4-dimethoxybenzoic acidC9H10O5198.052
45133.0502.7132,3-Dihydroxy-2-methylbutanoic acidC5H10O4134.057
46281.13810.129BisbyninC15H22O5282.146
47221.08113.7622,3-Dihydro-3-hydroxy-6-methoxy-2,2-dimethyl-4H-1-benzopyran-4-oneC12H14O4222.088
48239.07037.6912,4-DihydroxychalconeC15H12O3240.078
49317.06621.617DihydroisorhamnetinC16H14O7318.073
50443.1915.794Cynaroside AC21H32O10444.198
51371.13420.577Citrusin EC17H24O9372.141
52447.09218.473Kaempferol-7-O-glucosideC21H20O11448.099
53205.08632.6792,3-Dihydro-6-methoxy-2,2-dimethyl-4H-1-benzopyran-4-oneC12H14O3206.094
54343.15431.101SafficinolideC20H24O5344.161
55353.10232.3031-(3,4-Dihydroxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dioneC20H18O6354.109
56313.10733.7067-HydroxyenterolactoneC18H18O5314.114
57445.17011.357Crosatoside BC20H30O11446.177
58331.11524.787Mytilin AC13H20N2O8332.122
59263.12824.737(+)-Abscisic acidC15H20O4264.135
60426.22735.285DihydroxyacidissiminolC25H33NO5427.234
61343.11822.682Diosbulbin BC19H20O6344.125
62187.09619.475Methyl N-(a-methylbutyryl) glycineC9H16O4188.104
Table 3. Compounds identified in the ethanolic A. lancea extract by LC-QTOF-MS.
Table 3. Compounds identified in the ethanolic A. lancea extract by LC-QTOF-MS.
No.M/ZRT
(min)
CompoundsMolecular
Formula
Molecular Weight
1191.0561.970Quinic acidC7H12O6192.063
2179.0359.637Caffeic acidC9H8O4180.042
3177.0199.274EsculetinC9H6O4178.026
4243.0621.995PseudouridineC9H12N2O6244.069
5191.03414.798ScopoletinC10H8O4192.042
6209.11833.6283-Ethenyl-2,5-dimethyl-4-oxohex-5-en-2-yl acetateC12H18O3210.125
7161.02414.6733-HydroxycoumarinC9H6O3162.031
8353.0877.419Chlorogenic acidC16H18O9354.094
9281.13932.475BisbyninC15H22O5282.146
10207.02911.341FraxetinC10H8O5208.036
11207.06628.1535-(3′,5′-Dihydroxyphenyl)-gamma-valerolactoneC11H12O4208.073
12265.14433.653IsoleptospermoneC15H22O4266.151
13193.05015.124ScytaloneC10H10O4194.057
14311.12828.454Gancaonin VC19H20O4312.135
15153.0198.372Gentisic acidC7H6O4154.026
16147.0292.721D-threo-3-methylmalateC5H8O5148.036
17341.1081.907SucroseC12H22O11342.115
18353.0878.0085Z-Caffeoylquinic acidC16H18O9354.094
19341.0877.156Glucocaffeic acidC15H18O9342.094
20447.09212.3681,2,6,8-Tetrahydroxy-3-methylanthraquinone 2-O-b-D-glucosideC21H20O11448.100
21427.19616.703Taraxacolide 1-O-b-D-glucopyranosideC21H32O9428.204
22225.11225.4723,7-Dimethyl-2E,6E-decadien-1,10-dioic acidC12H18O4226.120
23221.04515.487IsofraxidinC11H10O5222.052
24128.0352.208Pyroglutamic acidC5H7NO3129.042
25381.17510.1631,2,10-Trihydroxydihydro-trans-linalyl oxide 7-O-beta-D-glucopyranosideC16H30O10382.183
26337.09210.338Hydrojuglone glucosideC16H18O8338.099
27441.17515.600LusitanicosideC21H30O10442.183
28485.19914.673Glucosylgalactosyl hydroxylysineC18H34N2O13486.207
29401.1448.847Benzyl O-(arabinofuranosyl-(1->6)-glucoside)C18H26O10402.151
30385.16426.675Gingerenone BC22H26O6386.172
31335.07613.1694-O-Caffeoylshikimic acidC16H16O8336.083
32305.13832.788AchillicinC17H22O5306.146
33425.14416.4146-(2-Carboxyethyl)-7-hydroxy-2,2-dimethyl-4-chromanone glucosideC20H26O10426.151
34353.14426.675Isopropyl apiosylglucosideC14H26O10354.151
35503.17513.169(S)-Multifidol 2-(apiosyl-(1->6)-glucoside)C22H32O13504.183
36329.23233.1649S,10S,11R-trihydroxy-12Z-octadecenoic acidC18H34O5330.239
37461.23813.270xi-Linalool 3-(rhamnosyl-(1->6)-glucoside)C22H38O10462.245
38511.23810.7893-O-(beta-D-glucopyranosyl-(1->6)-beta-D-glucopyranosyl) ethyl 3-hydroxyoctanoateC22H40O13512.245
39393.13339.996RotenoneC23H22O6394.141
40447.09215.976Kaempferol-7-O-glucosideC21H20O11448.099
41461.1659.837VerbasosideC20H30O12462.172
42479.24810.2633-O-(alpha-L-rhamnopyranosyl-(1-2)-alpha-L-rhamnopyranosyl)-3-hydroxydecanoic acidC22H40O11480.255
43529.26418.356Cinncassiol D2 glucosideC26H42O11530.271
44515.11820.2353″,4″-DiacetylafzelinC25H24O12516.125
45128.0352.496(r)-(+)-2-Pyrrolidone-5-carboxylic acidC5H7NO3129.042
46441.21216.828CAY10509C23H35FO5S442.219
47299.14132.688BifenazateC17H20N2O3300.148
Table 4. Compounds identified in the ethanolic extract of Prabchompoothaweep remedy by LC-QTOF-MS.
Table 4. Compounds identified in the ethanolic extract of Prabchompoothaweep remedy by LC-QTOF-MS.
No.M/ZRT
(min)
CompoundsMolecular
Formula
Molecular Weight
1173.0451.988Shikimic acidC7H10O5174.052
2197.0459.8422-Hydroxy-3,4-dimethoxybenzoic acidC9H10O5198.052
3169.0143.816Gallic acidC7H6O5170.021
4177.0199.216EsculetinC9H6O4178.026
5137.0247.7253,4-DihydroxybenzaldehydeC7H6O3138.031
6169.0146.4972,4,6-Trihydroxybenzoic acidC7H6O5170.021
7243.0513.4911-O-GalloylglycerolC10H12O7244.058
8166.0507.9762-Amino-3-methoxy-benzoic acidC8H9NO3167.058
9447.12924.625Piperenol CC22H24O10448.136
10153.0195.3203,4-Dihydroxybenzoic acidC7H6O4154.026
11211.06119.401Eudesmic acidC10H12O5212.068
12187.09719.489Methyl N-(a-methylbutyryl) glycineC9H16O4188.104
13197.04513.2123,4-O-Dimethylgallic acidC9H10O5198.052
14313.0563.290Salicyl phenolic glucuronideC13H14O9314.063
15161.0817.249Potassium 2-(1’-ethoxy) ethoxypropanoateC7H14O4162.089
16179.0359.629Caffeic acidC9H8O4180.042
17151.0409.0784-AcetoxyphenolC8H8O3152.047
18290.0882.012Sarmentosin epoxideC11H17NO8291.095
19191.03422.6085,7-Dihydroxy-4-methylcoumarinC10H8O4192.042
20353.0877.7135Z-Caffeoylquinic acidC16H18O9354.094
21237.11320.065Benzyl b-L-arabinopyranosideC13H18O4238.120
22222.04014.340(R)-2,3-Dihydro-3,5-dihydroxy-2-oxo-3-indoleacetic acidC10H9NO5223.047
23218.1033.516Pantothenic acidC9H17NO5219.110
24195.10216.169Isobutyl 2-furanpropionateC11H16O3196.109
25421.18632.869Picrasin FC22H30O8422.193
26355.0302.137(+)-Chebulic acidC14H12O11356.037
27153.0198.352Gentisic acidC7H6O4154.026
28325.0563.992Fertaric acidC14H14O9326.063
29243.12323.360Polyethylene, oxidizedC12H20O5244.130
30233.0456.7357-Hydroxy-2-methyl-4-oxo-4H-1-benzopyran-5-acetic acidC12H10O5234.052
31299.05532.242DiosmetinC16H12O6300.063
32310.14011.734LeonurineC14H21N3O5311.147
33300.99815.430Ellagic acidC14H6O8302.005
34328.11820.604N-trans-FeruloyloctopamineC18H19NO5329.125
35225.11211.9473,7-Dimethyl-2E,6E-decadien-1,10-dioic acidC12H18O4226.120
36163.03913.538m-Coumaric acidC9H8O3164.047
37321.0247.900DigallateC14H10O9322.032
38359.14934.8986′-O-FormylmarminC20H24O6360.156
39651.0839.316Amlaic acidC27H24O19652.090
40285.04032.518KaempferolC15H10O6286.047
41463.08715.881Quercetin 3-galactosideC21H20O12464.094
42347.0767.024alpha-(1,2-Dihydroxyethyl)-1,2,3,4-tetrahydro-7-hydroxy-9-methoxy-3,4-dioxocyclopenta(c) benzopyran-6-acetaldehydeC17H16O8348.083
43431.17033.708Melledonal AC23H28O8432.177
44261.04017.3962-Acetyl-5,8-dihydroxy-3-methoxy-1,4-naphthoquinoneC13H10O6262.047
45315.05033.0821,3,5,8-Tetrahydroxy-6-methoxy-2-methylanthraquinoneC16H12O7316.057
46326.0877.449BlepharinC14H17NO8327.094
47461.10825.966Rhamnetin 3-rhamnosideC22H22O11462.115
48161.06018.223Allyl benzoateC10H10O2162.067
49128.0352.426Pyroglutamic acidC5H7NO3129.042
50271.06031.302(±)-NaringeninC15H12O5272.067
51264.06634.710Piperolactam AC16H11NO3265.073
52272.12929.724(2E)-Piperamide-C5:1C16H19NO3273.136
53191.0551.887Quinic acidC7H12O6192.063
54134.02410.3562-BenzoxazololC7H5NO2135.032
55361.16522.821Gibberellin A98C20H26O6362.172
56201.11226.0412,6-Dimethyl-1,8-octanedioic acidC10H18O4202.120
57476.04013.688IsoterchebinC41H30O27954.096
58312.12324.863Pterostilbene glycinateC18H19NO4313.131
59285.04027.444LuteolinC15H10O6286.047
60635.08811.3083-O-GalloylhamamelitanninC27H24O18636.095
61351.05326.1914′-O-Methyl-(-)-epicatechin-7-O-sulfateC16H16O7S352.061
62269.04531.453ApigeninC15H10O5270.052
63343.04536.564Aflatoxin GM1C17H12O8344.052
64307.08118.4744R,5R,6S-Trihydroxy-2-hydroxymethyl-2-cyclohexen-1-one 6-(2-hydroxy-6-methylbenzoate)C15H16O7308.089
65447.09218.487Kaempferol-7-O-glucosideC21H20O11448.099
66461.07219.7263-Methylellagic acid 8-rhamnosideC21H18O12462.079
67201.01826.3416-HydroxyangelicinC11H6O4202.026
68623.19715.943IsoacteosideC29H36O15624.204
69211.0605.1193-Hydroxy-4-methoxyphenyllactic acidC10H12O5212.067
70301.03427.820HieracinC15H10O7302.042
71477.13924.462Eugenol O-[3,4,5-Trihydroxybenzoyl-(->6)-b-D-glucopyranoside]C23H26O11478.146
72342.13425.665N-trans-Feruloyl-4-O-methyldopamineC19H21NO5343.141
73547.14421.255Puerarin xylosideC26H28O13548.152
74251.12815.667QH (2)C14H20O4252.135
75329.02928.8972,8-Di-O-methylellagic acidC16H10O8330.036
76256.13334.309CoumaperineC16H19NO2257.140
77491.11828.7723′,7-Dimethoxy-4′,5,8-trihydroxyflavone 8-glucosideC23H24O12492.125
78281.13810.080BisbyninC15H22O5282.146
79403.17532.255Myristicanol BC22H28O7404.183
80403.1235.921Oleoside 11-methyl esterC17H24O11404.131
81465.10217.998(-)-Epicatechin 7-O-glucuronideC21H22O12466.110
82379.17520.4036b-Angeloyl-3b,8b,9b-trihydroxy-7(11)-eremophilen-12,8-olideC20H28O7380.182
83593.15017.221SaponarinC27H30O15594.157
84379.01228.672Tectorigenin 7-sulfateC16H12O9S380.019
85241.0716.046Elenaic acidC11H14O6242.078
86955.10416.570Chebulinic acidC41H32O27956.111
87477.10219.000Myricetin 3,4′-dimethyl ether 3′-xylosideC22H22O12478.110
88497.22319.6142-O-(beta-D-galactopyranosyl-(1->6)-beta-D-galactopyranosyl) 2S-hydroxynonanoic acidC21H38O13498.230
89593.12927.7956″-O-p-CoumaroyltrifolinC30H26O13594.136
90515.11820.1773″,4″-DiacetylafzelinC25H24O12516.125
91161.0454.4433-Hydroxy-3-methyl-glutaric acidC6H10O5162.052
92447.09212.3851,2,6,8-Tetrahydroxy-3-methylanthraquinone 2-O-b-D-glucosideC21H20O11448.099
93387.10716.9457-Hydroxy-3′,4′,5,6,8-pentamethoxyflavoneC20H20O8
388.115
94769.25418.349Leonoside AC35H46O19770.261
95637.17613.876Quercetin 3,3′-dimethyl ether 7-rutinosideC29H34O16638.183
96431.09719.150Apigenin 7-O-glucosideC21H20O10432.104
97581.22212.498(+)-Lyoniresinol 9-glucosideC28H38O13582.230
98461.23813.976xi-Linalool 3-(rhamnosyl-(1->6)-glucoside)C22H38O10462.245
99695.39931.177Glucosyl passiflorateC37H60O12696.407
100461.1654.543VerbasosideC20H30O12462.172
101755.23815.292Hesperetin 7-(2,6-dirhamnosylglucoside)C34H44O19756.246
102435.12819.075Phenethyl 6-galloylglucosideC21H24O10436.135
103429.15223.6852,3-dinor FluprostenolC21H25F3O6430.159
104651.22824.262(-)-Matairesinol 4′-(apiosyl-(1->2)-glucoside)C31H40O15652.235
105153.0554.9442-Furanylmethyl propanoateC8H10O3154.062
106665.20723.460Tetramethylquercetin 3-rutinosideC31H38O16666.214
107637.21219.3764′-Hydroxy-5,7,2′-trimethoxyflavanone 4′-rhamnosyl-(1->6)-glucosideC30H38O15638.219
108582.25930.238N1, N5, N10-Tricoumaroyl spermidineC34H37N3O6
583.266
109433.14920.854Vestitone 7-glucosideC22H26O9434.156
110453.24833.457Rhodojaponin IVC24H38O8454.255
111137.0248.088m-Salicylic acidC7H6O3138.031
112477.06610.080Quercetin 3′-O-glucuronideC21H18O13478.073
113787.09814.7411,2’,3,5-Tetra-O-galloylhamamelofuranoseC34H28O22788.105
114577.15417.647Scutellarein 7,4′-dirhamnosideC27H30O14578.162
115939.10817.1711,2,3,4,6-Pentakis-O-galloyl-beta-D-glucoseC41H32O26940.115
116331.08116.7952′,3,5-Trihydroxy-5′,7-dimethoxyflavanoneC17H16O7332.088
117347.0373.9412-(α-D-Mannosyl)-3-phosphoglycerateC9H17O12P348.045
118315.15934.910Isopulegone caffeateC19H24O4316.166
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.
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.
GroupDose (mg/kg)% Parasitemia% Suppression
7% Tween 80-40.45 ± 2.15 b,c,d,e,f,g,h,i,j,k,l-
Artesunate62.18 ± 0.50 a,d,e,f,g,h,i,j,k,l95.32 ± 0.57 d,e,f,g,h,i,j,k,l
Chloroquine250.27 ± 0.15 a,d,e,f,g,h,i,j,k,l99.34 ± 0.37 d,e,f,g,h,i,j,k,l
M. fragrans20024.94 ± 2.50 a,b,c,h,l38.32 ± 6.18 b,c,h,i,k,l
40022.36 ± 1.26 a,b,c,h,l44.17 ± 3.12 b,c,h,l
60021.48 ± 0.73 a,b,c,h,l46.86 ± 1.80 b,c,h,l
A. lancea20021.53 ± 2.47 a,b,c,h,l46.75 ± 6.11 b,c,h,j,k,l
40016.13 ± 0.41 a,b,c,d,e,f,g,i,j,k60.09 ± 1.03 b,c,d,e,f,g,i,j,k
60020.91 ± 1.15 a,b,c,h,l48.29 ± 2.86 b,c,d,h,l
Prabchompoothaweep remedy20024.60 ± 1.03 a,b,c,h,l39.18 ± 2.56 b,c,h,l
40020.88 ± 3.08 a,b,c,h,l48.35 ± 7.62 b,c,d,h,l
60016.13 ± 0.58 a,b,c,d,e,f,g,i,j,k60.11 ± 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.
Table 6. Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on pack cell volume in the 4-day suppressive test.
Table 6. Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on pack cell volume in the 4-day suppressive test.
GroupDose
(mg/kg)
Day 0Day 4% Change
7% Tween 80-49.60 ± 1.0145.00 ± 1.89−10.35 ± 3.67% b,c
Artesunate652.20 ± 1.3254.80 ± 1.164.74 ± 1.43% a,d,e,f,g,h,i,j,k,l
Chloroquine2551.80 ± 0.4353.40 ± 1.472.87 ± 2.36% a,d,j
M. fragrans20054.00 ± 0.8949.80 ± 2.03−8.58 ± 3.89% b,c
40051.20 ± 1.1648.40 ± 0.80−5.78 ± 1.52% b
60051.00 ± 1.0948.80 ± 1.93−4.64 ± 4.05% b
A. lancea20052.00 ± 2.7349.40 ± 2.17−5.58 ± 8.67% b
40052.40 ± 1.0150.00 ± 1.67−4.86 ± 2.15% b
60051.20 ± 1.1648.80 ± 1.46−5.01 ± 3.73% b
Prabchompoothaweep remedy20052.60 ± 1.3548.20 ± 2.13−9.25 ± 3.46% b,c
40051.80 ± 1.8349.40 ± 1.35−4.87 ± 3.02% b
60050.80 ± 2.6348.60 ± 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.
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.
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 1Week 2
Normal mice25.0 ± 3.521.6 ± 2.7
7% Tween 8022.1 ± 1.720.8 ± 2.5
M. fragrans 2000 mg/kg20.8 ± 2.020.7 ± 0.8
A. lancea 2000 mg/kg22.8 ± 2.522.4 ± 2.3
Prabchompoothaweep
remedy 2000 mg/kg
23.6 ± 3.922.0 ± 1.9
Water Consumption (mL)Week 1Week 2
Normal mice122.2 ± 4.7125.8 ± 7.9
7% Tween 80122.4 ± 8.3126.7 ± 8.3
M. fragrans 2000 mg/kg122.4 ± 3.5127.7 ± 3.5
A. lancea 2000 mg/kg126.0 ± 4.8130.4 ± 5.0
Prabchompoothaweep
remedy 2000 mg/kg
125.0 ± 2.6130.5 ± 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.
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.
GroupMean Body Weight
Day 0Day 14% Change
Normal mice33.4 ± 1.539.2 ± 2.214.6 ± 1.7%
7% Tween 8032.5 ± 1.436.5 ± 1.511.1 ± 1.3%
M. fragrans 2000 mg/kg32.8 ± 1.238.0 ± 2.413.4 ± 3.3%
A. lancea 2000 mg/kg33.0 ± 1.638.0 ± 2.713.0 ± 2.6%
Prabchompoothaweep
remedy 2000 mg/kg
32.1 ± 1.136.8 ± 1.412.6 ± 1.5%
Data are presented as mean ± SEM (n = 5 per group).
Table 9. Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on kidney and liver functions in the acute toxicity test.
Table 9. Effect of ethanolic extracts of M. fragrans, A. lancea, and Prabchompoothaweep remedy on kidney and liver functions in the acute toxicity test.
ParametersNormal Mice7% Tween 80 M. fragransA. lanceaPrabchompoothaweep Remedy
Liver Function Test
AST (U/L)83.80 ± 7.1383.00 ± 9.1887.75 ± 12.5794.60 ± 8.7792.00 ± 5.17
ALT (U/L)36.80 ± 8.0838.80 ± 3.7031.75 ± 6.9634.60 ± 5.5734.60 ± 7.05
ALP (U/L)92.10 ± 11.3591.04 ± 7.8690.50 ± 8.9688.40 ± 7.0389.20 ± 12.79
Kidney Function Test
BUN (mg/dL)26.42 ± 3.8631.04 ± 3.9625.45 ± 2.3025.56 ± 3.8925.26 ± 2.12
Creatinine (mg/dL)0.66 ± 0.040.69 ± 0.070.66 ± 0.030.65 ± 0.040.61 ± 0.06
Data are presented as mean ± SEM (n = 5 per group).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Plirat, W.; Chaniad, P.; Phuwajaroanpong, A.; Septama, A.W.; Punsawad, C. Phytochemical, Antimalarial, and Acute Oral Toxicity Properties of Selected Crude Extracts of Prabchompoothaweep Remedy in Plasmodium berghei-Infected Mice. Trop. Med. Infect. Dis. 2022, 7, 395. https://doi.org/10.3390/tropicalmed7120395

AMA Style

Plirat W, Chaniad P, Phuwajaroanpong A, Septama AW, Punsawad C. Phytochemical, Antimalarial, and Acute Oral Toxicity Properties of Selected Crude Extracts of Prabchompoothaweep Remedy in Plasmodium berghei-Infected Mice. Tropical Medicine and Infectious Disease. 2022; 7(12):395. https://doi.org/10.3390/tropicalmed7120395

Chicago/Turabian Style

Plirat, Walaiporn, Prapaporn Chaniad, Arisara Phuwajaroanpong, Abdi Wira Septama, and Chuchard Punsawad. 2022. "Phytochemical, Antimalarial, and Acute Oral Toxicity Properties of Selected Crude Extracts of Prabchompoothaweep Remedy in Plasmodium berghei-Infected Mice" Tropical Medicine and Infectious Disease 7, no. 12: 395. https://doi.org/10.3390/tropicalmed7120395

Article Metrics

Back to TopTop