Next Article in Journal
The Catechins Profile of Green Tea Extracts Affects the Antioxidant Activity and Degradation of Catechins in DHA-Rich Oil
Next Article in Special Issue
The Total Dietary Antioxidant Capacity, Its Seasonal Variability, and Dietary Sources in Cardiovascular Patients
Previous Article in Journal
Carob (Ceratonia siliqua) as Functional Feed Is Beneficial in Yellow Mealworm (Tenebrio molitor) Rearing: Evidence from Growth, Antioxidant Status and Cellular Responses
Previous Article in Special Issue
Phytochemical Screening, Antioxidant, and Enzyme Inhibitory Properties of Three Prangos Species (P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii) Depicted by Comprehensive LC-MS and Multivariate Data Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Current State of Knowledge in Biological Properties of Cirsimaritin

1
Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Sidi Bouzid B.P. 4162, Morocco
2
Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University, Taza-Gare, Taza B.P. 1223, Morocco
3
Laboratory of Organic Chemistry, Catalysis and Environment, Department of Chemistry, Faculty of Sciences, Ibn Tofail University, B.P. 133, Kenitra 14000, Morocco
4
Department of Clinical Neurophysiology, Hospital of Specialities, Rabat Institute, Ibn Sina University Hospital, Rabat 10056, Morocco
5
Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat 10100, Morocco
6
Department of Scientific Research, V. M. Gorbatov Federal Research Center for Food Systems, 109316 Moscow, Russia
7
Biophotonics Center, Prokhorov General Physics Institute of the Russian Academy of Science, 119991 Moscow, Russia
8
Department of Scientific Research, Russian State Agrarian University—Moscow Timiryazev Agricultural Academy, 49 Timiryazevskaya St., 127550 Moscow, Russia
9
Department of Chemistry, The University of Jordan, Amman 11942, Jordan
10
Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade de Vigo, E-32004 Ourense, Spain
11
Department of Biology, Faculty of Science, Selcuk Universtiy, 42130 Konya, Turkey
12
College of Korean Medicine, Kyung Hee University, Hoigidong, Dongdaemungu, Seoul 02447, Korea
13
Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, and The Shine-Dalgarno Centre for RNA Innovation, The Australian National University, Canberra, ACT 2601, Australia
14
Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
15
Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10106, Morocco
*
Authors to whom correspondence should be addressed.
Antioxidants 2022, 11(9), 1842; https://doi.org/10.3390/antiox11091842
Submission received: 23 August 2022 / Revised: 8 September 2022 / Accepted: 13 September 2022 / Published: 19 September 2022

Abstract

:
The search for natural plant-based products as new pharmacological alternatives to treat various human pathologies has taken on great importance for researchers and research laboratories. In this context, research has intensified to extract and identify natural molecules endowed with biological effects. The objective of this study is to review the source and pharmacological properties of cirsimaritin. The identification and isolation of this flavonoid from various natural sources, including medicinal plants such as Artemisia judaica, Cirsium japonicum, Lithocarpus dealbatus, Microtea debilis, and Ocimum sanctum, has been carried out and verified using different spectral techniques. Biological effect investigations are carried out with a wide variety of experimental models in vitro and in vivo and laboratory techniques. The results of these research works showed the biological properties of cirsimaritin including anticancer, antimicrobial, antidiabetic, antiparasitic, antioxidant, and anti-inflammatory effects. The mechanisms involved in the multiple activities of this molecule are diverse and include sub-cellular, cellular, and molecular levels. Indeed, this bioactive induces anti-inflammatory and antiproliferative effects by inhibiting cell membrane receptors, interference with signaling pathways, and inhibiting transcriptional factors such as Nf-κB involved in cell promotion and proliferation. In the light of these results, cirsimaritin appears as a promising and viable alternative natural bioactive drug to treat many pathological conditions.

1. Introduction

The development of drugs from secondary metabolites of medicinal plants has been widely preoccupied by current scientific research [1,2,3,4,5]. Indeed, some plants synthetize a wide variety of remarkable molecules, such as daucosterol, carvone, pinosylvin, and chrysoeriol, with important health benefits such as anticancer, antidiabetic, antimicrobial, antiparasitic effects [1,6,7,8,9]. These natural bioactive compounds belong to different chemical classes such as phenolic acids, terpenoids, flavonoids, alkaloids, and terpenoids [10,11,12,13,14,15,16]. Moreover, flavonoids are considered as the most abundant and diverse family [17,18] with promising therapeutic benefits since they are non-toxic molecules with many biological activities including anticancer [19], inflammatory [20], antibacterial [21], antiviral [22], neuroprotective effects [23], and other activities. On the other hand, if their proprieties are properly used, these natural products could be efficient, safe, and new therapeutic agents. The exploration of these natural flavonoids can be a promising strategy to identify and develop drugs for pharmaceutical applications. The flavonoid cirsimaritin is found in many plants including, for example, Artemisia judaica, Cirsium japonicum, Lithocarpus dealbatus, Microtea debilis, and Ocimum sanctum [24,25,26,27]. Cirsimaritin has been reported to exert numerous biological effects including antimicrobial, anti-inflammatory and anti-proliferative properties [28,29,30,31].
Indeed, it showed antimicrobial effect against fungi and Gram-positive and Gram-negative bacteria [28,29,30]. This flavonoid has also demonstrated an anti-inflammatory potency, which is mediated by the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the inhibition of c-fos in RAW264 cells [32]. In addition, an antidiabetic benefit was attributed to cirsimaritin [31]. In TNF-α-treated FL83B mouse hepatocytes, its anti-hyperglycemia effects are linked to cirsimaritin’s ability to enhance glucose uptake. In silico, insulin secretion was increased after DPP-4 blockade by cirsimaritin Bower et al. [33].
Additionally, cirsimaritin had antiproliferative activity against human colon carcinoma (HT-29), human gastric adenocarcinoma (AGS), human osteosarcoma (SaOs-2), murine fibrosarcoma (WEHI-164) and human fetal foreskin fibroblast (HFFF-P16), MCF-7, and PC-3 cell lines. Its anticancer mechanisms involve some key cancer targets such as apoptosis, p-Akt, and cAMP/PKA signaling. Other pharmacological effects have also been described, including the inhibitory activity against the influenza A virus via the blockage of the NF-κB/p65 signal pathway [34], antioxidant effect [35,36], antiparasitic activity on Entamoeba histolytica, Leishmania donovani, Plasmodium falciparum, Trypanosoma brucei rhodesiense, and Trypanosoma cruzi [37], antinociceptive, and anxiolytic activities [38], as well as the protective effect of beta-cells against STZ. Even though several studies have highlighted the health benefits and biological properties of cirsimaritin, no reviews have been published to discuss and explore its properties. Therefore, the aim of this review was to represent the pharmacological activities of cirsimaritin by clarifying the molecular mechanisms responsible for these properties in the context to explore new pharmaceutical opportunities for this natural molecule and to provide a significant starting point for future studies.

2. Materials and Methods

The bibliometric research was carried out in a global way, without exclusion criteria and without inclusion criteria, from several databases (since 1963 to 2022), including PubMed, science-direct, Google-Scholar, Scopus, and Web of Sciences. Different keywords including cirsimaritin, biological properties of cirsimaritin, anticancer effects of cirsimaritin, antimicrobial effects of cirsimaritin, pharmacological properties of cirsimaritin were used to find data. The data collected were firstly classified according to different sections (sources and different biological and pharmacological properties). Then, the publications of each section were organized in tables and explored. These data were finally discussed and highlighted. The molecular structure of cirsimaritin was designed using Chem-Draw program.

3. Results and Discussion

3.1. Sources of Cirsimaritin

In 1963, cirsimaritin, known as flavone, was first isolated by Morita and Shimizu [39] using Cirsium martimum, and many others natural sources reported in Table 1; it is a small molecular natural flavonoid (Figure 1).

3.2. Biological and Pharmacological Properties

In vitro and in vivo investigations concerning cirsimaritin have shown multiple biological and pharmacological properties (Figure 2). These induced activities are due to the different mechanisms mediated by this substance. In these following sections, the pharmacological effects as well as the associated mechanisms of this compound will be described, highlighted, and discussed.

3.2.1. Antibacterial and Anti-Fungal Activities

The first research reporting the antimicrobial properties of cirsimaritin dates back to 1983 (Table 2). Miski et al. [78] investigated the antibacterial effect of Salvia palaestina against negative and positive gram bacteria. The results showed that cirsimaritin was the only flavonoid with the highest antibacterial activity against Escherichia coli, Klebsiella pneumonia, Pseudomonas aeruginosa, Proteus vulgaris, Staphylococcus aureus, and Staphylococcus epidermis using the disk diffusion method. In another study reported by Ragasa et al. [28], cirsimaritin was found to be active against the fungi aspergillus niger, Candida albicans, and trichophyton mentagrophytes, and, with activity indices of 0.3, 0.3, and 0.4, respectively. However, cirsimaritin showed weak antimicrobial activity and was even inactive against Bacillus subtilis, Escherichia coli, and Staphylococcus aureus. Similar results were recorded by Rijo et al. [29] on the antimicrobial effect of cirsimaritin using micro-dilution and agar diffusion methods to determine Minimum Inhibitory Concentrations (MICs) and the diameter of inhibition zones, respectively. The authors showed that the most resistant pathogen to cirsimaritin was Escherichia coli, moreover this molecule did not inhibit Candida albicans, Mycobacterium smegmatis, and Pseudomonas aeruginosa, while an anti-Staphylococcus aureus and anti-Enterococcus hirae effect was observed. A study by Marino et al. [30] found that cirsimaritin, one of the major constituents of Asphodeline anatolica acetone extract, exhibits an antibacterial effect against Listeria monocytogenes, Pseudomonas aeruginosa, and Staphylococcus aureus.
Significant antimicrobial effect of Centaurea pseudosinaica extract, with cirsimaritin as one of the main components, was recorded using the micro-dilution technique, against human pathogens such as Aspergilus fumigatus, Candida albicans, Geotrichum candidum, and Syncephalastrum racemosum, and Gram-positive (Bacillis subtilis and Streptococcus pneumoniae) and Gram-negative (Escherichia coli and Pseudomonas aeruginosa) bacteria. The results obtained clearly revealed an excellent efficacy of the alcoholic extract against all micro-organisms, especially against Candida albicans [51]. Interestingly, Ren and co-workers observed the antimicrobial activities of cirsimaritin identified in the bark of Tamarix ramosissima on Bacillus cereus, Escherichia coli, Staphylococcus aureus, Listeria monocytogene, Pseudomonas aeruginosa, Salmonella typhimurium, and Shigella castellani using the disk diffusion method. The inhibitory effect of the bark extract on bacterial pathogens was appreciable. Bacillus cereus, Listeria monocytogenes, and Staphylococcus aureus are more sensitive to the bark extract than Escherichia coli, Pseudomonas aeruginosa, Salmonella typhimurium, and Shigella castellani. Among these bacteria, L. monocytogenes was the most damaging bacteria with the lowest MBC value (10 mg/mL) [83].

3.2.2. Antiviral and Antiparasitic Activities

Cirsimaritin was also studied for its activities against viruses. Indeed, Yan et al. [34] tested cirsimaritin on the influenza virus and its mechanism effect. A concentration-dependent reduction of viral titers, protein synthesis, and influenza A virus (IAV) RNA after cirsimaritin treatment has been demonstrated. From the point of view of the mechanism, the team suggests that the inactivation of the NF-κB/p65 signal pathway might be the origin of cirsimaritin-induced inhibition of IAV. In a computer-aided research, Hussain et al. [89] studied the cirsimaritin effect, isolated from Santolina insularis, on chikungunya virus (CHIKV) replication by targeting the non-structural proteins from CHIKV (nsP4, nsP3, nsP2, and nsP1). As results, cirsimaritin has shown a strong binding affinity on nsP1 compared to others.
Regarding the recent outbreaks of dangerous viruses namely COVID-19 with a rapid spread and which the development of new anti-viral drugs represents a main challenge. In this context, cirsimaritin was tested, in silico, against spike Protein SARS-CoV-2 [90]. According to this research, Cirsimaritin shows moderate binding affinity against the target protein.
Concerning parasitic infections, cirsimaritin exhibited a high inhibition versus Plasmodium falciparum (IC50 = 16.9 µM) [91], and similar activity against Leishmania donovani, Trypanosoma brucei rhodesiense, Trypanosoma cruzi with an IC50 equal of 3.9 µg/mL, 3.3 µg/mL, and 19.7 µg/mL, respectively, according to Tasdemir et al. [37]. Moreover, Quintanilla-Licea et al. [92] reported the antiprotozoal activity of this molecule against Entamoeba histolytica (IC50 = 154.26 µg/mL).

3.2.3. Antioxidant Activity

Cirsimaritin represent an important benefic health molecule used in pharmaceutical industries to produce antioxidants and products against free radicals (Table 3). A study performed by Ibañez et al. [35] using DPPH assay revealed that the extracted compounds from Rosmarinus officinalis leaves, containing the flavonoid cirsimaritin showed high antioxidant activity. Similar results were obtained by a study investigated by Jipa et al. [36], who evaluated the antioxidant activity of Rosmarinus officinalis extract after γ-irradiation. It has been showed in this study that the cirsimaritin present in R. officinalis extract possess a good antioxidant property which enhanced by high-energy irradiation. Likewise, Cavero et al. [71] demonstrated the antioxidant efficiency of Rosmarinus officinalis extract on 1, 1-Diphenyl-2-picrylhydrazyl (DPPH). In this study, cirsimaritin was identified as one of the most important flavonoids in the extract, but the correlation matrix established by the authors, showed that this molecule is poorly correlated with the antioxidant activity (EC50 values). Furthermore, Kelm et al. [27] demonstrate using an antioxidant assay performed by analyzing the oxidation of model liposomes by fluorescence spectroscopy that cirsimaritin extracted from Ocimum sanctum surprisingly displayed poor antioxidant activity. Similarly in another study, Kolak et al. [93], demonstrated that cirsimaritin from Salvia poculata extract did not have antioxidant properties using β-carotene bleaching, ABTS cation radical scavenging activity and superoxide anion radical assays. Ben Sghaier et al. [86] demonstrated that cirsimaritin extracted from Teucrium ramosissimum showed an excellent antioxidant capacity at a Teac value of 2.04 µM using the ABTS. Lee et al. [53] studied the antioxidant effect of cirsimaritin isolated from Korean thistle (Cirsium japonicum) against DPPH. Results suggest that cirsimaritin showed potential reduction of DPPH free radicals with percentages of inhibition between 80% and 100% at a concentration of 100 µg/mL. The antioxidant capacity was observed to be significantly higher in the extract of Artemisia Judaica which is a rich source of cirsimaritin [94]. Another study by Fattahi et al. [57] evaluated the antioxidant potency of cirsimaritin identified among 13 natural populations of Dracocephalum kotschyi using the ferric reduction capacity of plasma (FRAP). The results indicate that the antioxidant ability of the plant extracts was mostly due to the surface flavonoids, notably the cirsimaritin flavonoid for which the concentration varies from 97.38 to 637.66 µg/g DW leading to a high antioxidant activity ranging from 203.39 to 681.27 µmol Fe2+/100 g DW. Burki et al., [95] found that the Monotheca buxifolia bark extract which contain cirsimaritin as an active compound showed significant antioxidant effect against DPPH, superoxide and hydrogen peroxide with respectively an inhibition percentage of 89.55, 82.10 and 80.55% at a concentration of 500 µg/mL. Dawé et al. [56] investigated the antioxidant effect of cirsimaritin isolated from Combretum fragrans extract. The authors showed that cirsimaritin presented potent DPPH radical scavenging activity with a reported IC50 value of 55.9 µM.

3.2.4. Anti-Inflammatory Activity

In a study conducted by Shin et al. [32] on the anti-inflammatory properties and mechanisms of action of cirsimaritin derived from an ethanolic extract of Cirsium japonicum var. maxime maackii, using RAW264.7 cells; the extract and cirsimaritin inhibit nitric oxide (NO) production and inducible expression of NO synthase in RAW264.7 cells. In addition, cirsimaritin can reduce inflammatory response by increasing of MCP-1, CD3+ T, CD68+ (Figure 3).
Cirsimaritin also caused the blockage of the production of different cytokines including IL-6, IL-10, IL-6, IFN-γ, and TNF-α, as well a decrease in NO production in a dose-dependent manner in RAW264.7 via the repression iNOS expression (Figure 3) [32]. It also suppressed the activation of the transcription factors induced by LPS, namely c-fos, STAT3. From these findings, this flavonoid may have an anti-inflammatory effect, which is regulated by the phosphorylation of STAT3 and the inhibition of c-fos in RAW264 cells. This inhibition of gene expression induces a decrease of cyclooxygenase-2 (COX-2) and Myeloperoxidase (MPO) (Figure 3).
Two other research works conducted separately by Al Ati et al. [38] and Cottiglia et al. [68] on cirsimaritin, isolated from traditionally used medicinal plants, showed a very significant anti-inflammatory activity. In the same context, another research study carried out by Kelm et al. [27] evaluated the cyclooxygenase (COX) inhibitory activity of cirsimaritin, at concentrations greater than 1000 µM, purified from the extract of fresh stems and leaves of Ocimum sanctum, while comparing with naproxen, ibuprofen and aspirin at concentrations of 10, 10, and 1000 M, respectively.
During the research performed by Kuo et al. [96], rosemary (Rosmarinus officinalis) extract containing cirsimaritin showed a dose-dependent effect on the expression of inflammatory mediators, in particular on lipid peroxidation. Nevertheless, the simultaneous connection of anti-inflammatory activity between rosemary extract (from SC-CO (2) at 5000 psi and 80 °C) and its pure carnosic acid (AC) via murine macrophage cells RAW 264.7 treated with LPS was determined. In addition, CA and SCCO (2) distinctly inhibited LPS-induced NO production, phosphorylated IkappaB (P-IκB), TNF-α, and NF-κB/p65 the inducible nitric oxide synthase (iNOS) and the expression of COX-2 (Figure 3). From these data, it can be concluded that cirsimaritin exhibits a significant anti-inflammatory effect with better inhibitory activity on NO (IC50 of 22.5 M or 7.47 g/mL) in comparison with the SCCO extract (IC50 of 14.50 g/mL).

3.2.5. Antidiabetic Activity

Numerous studies have revealed the therapeutic potential of cirsimaritin in the treatment of diabetes. Research conducted by Stefkov et al. [85] investigated the biochemical mechanism of the insulinotropic and antihyperglycemic effects of T. polium extracts containing cirsimaritin. Therefore, an interesting insulinotropic effect on INS1E cells at a dose of 500 µg/mL is observed. After 8 h following administration, the doses of 125 mg/kg (the same doses of the extract) administered intra-gastrointestinally in hyperglycemic and normal rats were found to be more effective in lowering blood glucose compared to intraperitoneal injection (35% reduction vs. 24%) with a large effect (50% reduction). After a week and a half of treatment, the comparison of the effect levels following the administration of glibenclamide at 2.5 mg/kg (reduction of 38%), demonstrates the absence of effects on the blood lipid profiles. As for the oral glucose tolerance assay, the extract lowered blood sugar by about 35%. Consequently, it was found that treatment results in a decrease in hepatic glycogen and tends to normalize the effect of gluconeogenesis enzymes. In another study performed with virtual screening, cirsimaritin has good binding affinities for DPP-4 and, once DPP-4 will be blocked, insulin secretion will increase (Figure 4) [33]. Moreover, by facilitating of Na/Ca exchange, cirsimaritin can modulate gene expression of several intracellular proteins involved in glucose metabolism regulation including PKC. It can also increase the expression of key factors involved in apoptosis preventing thus the pancreatic β cells from apoptotic depending on glucotoxicity and/or lipotoxicity (Figure 4).
In TNF-α-treated mouse FL83B hepatocytes, Xu et al. [73] investigated the mechanism by which the bioactive fractions from Ruellia tuberosa enhanced insulin resistance using a glucose uptake assay. Among them, EAF5-5 fraction, which contained syringic acid (27.3 μg/g), p-coumaric acid (95.0 μg/g), and cirsimaritin (805.5 μg/g) markedly improve glucose uptake. Those authors suggested that cirsimaritin may be considered as one major active ingredient from the EAF5-5 combination involved in the enhancement of glucose uptake rate of insulin-resistant FL83B hepatocytes.

3.2.6. Anti-Cancer Activity

Several studies have been carried out on cell cultures showed that cirsimaritin exerts antiproliferative activities [97,98,99,100,101,102,103] on numerous cancer cell lines (Table 4). In this regard, Moghaddam et al. [97] studied, in vitro, the antiproliferative activity of cirsimaritin isolated from Dracocephalum kotschyi against normal and malignant cell lines using the MTT test. As results, this molecule showed a moderate inhibition of HT-29 and AGS cell lines proliferation compared to SaOs-2 and WEHI-164. Likewise, Bai et al. [63] have shown that the said substance also exhibits a moderate anti-proliferative activity on COLO-205 cells with IC50 values equal of 13.1 μM.
Another study released by Sen et al. [50] indicates that cirsimaritin, isolated from the chloroform extract of Centaurea kilaea Boiss, exhibits significant anticancer activity, especially against breast cancer, and the value (0.5–50 μg/mL) was taken versus a normal cell line (L929, mouse fibroblast) and certain human cancer cell lines (cervical carcinoma, MCF-7, prostate carcinoma, PC-3, and breast carcinoma) using the MTT test with IC50 value equal to 4.3 μg/mL.
Using an in vivo cancer model, Awad et al. [101] recorded an antitumor activity of this molecule (separated from Achillea fragrantissima extract), characterized by the decrease in tumor size which can be explained by its antioxidant activities, confirmed by the increase in serum rate of TAC and reduction in serum TNF-α (Figure 5). Furthermore, the results of the histopathological examination demonstrated the induction of apoptosis.
The first report on the antitumor effects and the underlying mechanisms of cirsimaritin versus GBC-SD and GBCSD18H cells (gallbladder carcinoma cell lines), BGC-823 cells (gastric carcinoma cell line), SMMC-7721 cells (hepatoma cell line), SMMC-7721 cells (hepatoma cell line), and BGC-823 cells (gastric carcinoma cell line) was investigated by Quan et al. [99]. They showed that this cirsimaritin of synthetic origin exhibits an important antitumor activity and caused mitochondrial apoptosis in GBC-SD cells via activating caspase-4, -9, and -3 cascades, changing the mitochondrial membrane potential. Furthermore, cirsimaritin lead to the generation of reactive oxygen species in GBC-SD cells which triggers ER stress mitochondrial apoptotic pathways in GBC-SD cells. These molecular events induce an intrinsic apoptosis depending on caspases activation and ROS (Figure 5).
In another study, cirsimaritin as a major component of Cirsium japonicum var. maackii, exhibited an inhibition of the viability of HUVECs with a concentration-dependent manner, which showed it inhibited angiogenesis. This anticancer activity was assigned to decrease secretion of angiogenesis mediator vascular endothelial growth factor (VEGF). This inhibition induces a decrease PK3K, Raf, Src, and Erk which declined the rates of p-Akt and p-ERK in MDA-MB-231 cells, inducing thus a decrease of mTOR and causing an anti-angiogenesis effect [106] (Figure 5).
In recent study, cirsimaritin antiproliferative efficacity has been tested in human cancer cell lines including HaCaT, A431, K562, MDA-MB-231, A549, COLO-205, MCF-7, HaCaT, K562, NCIH- 520, and PC-3, and normal cell lines HEK 293, L132, and WRL-68 and in primary macrophages, by Pathak et al. [98]. It was found that cirsimaritin showed selective antitumor effect against NCIH-520 cell-line with IC50 = 23.29 μM, via increasing the apoptosis (10 and 100 μM). Moreover, cirsimaritin also inhibits the action of CATD and ODC which is responsible for the development stage of the cancer cells. In addition to this, follow Lipinski’s rule of five, it exerted a good binding score with the selected targets and it non-mutagenic.
Similarly, cirsimaritin from Betula pubescens and Betula pendula also had a role in lowering the viability, proliferation and clonogenicity of liver (HepG2), colon (DLD-1) and colon (DLD-1), and gastric (AGS) cancer cells. This flavonoid activated intrinsic caspases (3, 7, 8 and 9) mediated apoptosis [49].

3.2.7. Other Biological Activities

Many biological activities can be linked with the use of cirsimaritin. Indeed, cirsimaritin inhibits the amplitude of the phasic contractions of guinea-pig ileum [108]. Furthermore, Hasrat et al. [26] demonstrated that cirsimaritin induces an adenosine antagonistic effect in rats. These results suggest that the interaction between the cirsimaritin and adenosine receptor could result in inhibition of calcium and sodium transport. According to these researchers, this flavonoid-molecule could have a beneficial effect in acute renal failure.
Similarly, Abdelhalim et al. [70] reported that cirsimaritin exerted significant antinociceptive and anxiolytic effects which the anxiolytic activity may be mediated via GABAA receptor, without any signs of acute toxicity (50–200 mg/kg). The anxiolytic-like activity cirsimaritin in mice was also reported by González-Trujano et al. [109]. At a concentration of 3 mg/Kg (i.p), the number of head-dips was decreased by this substance and the lapse of open arms was increased in the hole-board and in the plus-maze tests, respectively. Furthermore, Wang et al. [110] studied the inhibition of cirsimaritin on a formyl-methionyl-leucyl-phenylalanine-(fMLP) excited respiratory burst in rat neutrophils. Those researchers observe that it is probably that cirsimaritin inhibits the fMLP-caused respiratory burst in vivo via the closure of the phos-pholipase D (PLD) signalling pathway.
The protective effect of cirsimaritin from Korean thistle on apoptosis induced by STZ is documented by Lee et al. [103]. Their results, in pancreatic b cells, proved that cirsimaritin potentially removed apoptosis via increasing anti-apoptotic BCL-2 protein expression and reducing the activation of both caspases 8 and 3, BID and the DNA repair protein poly (ADP-ribose) polymerase (PARP).
Earlier study reported by Kim et al. [102] on the hyperpigmentation activity of cirsimaritin (isolated from Lithocarpus dealbatus branches) and explained the mechanism by which cirsimaritin stimulate melanogenesis in B16F10 cells (murine melanoma cells). The author’s findings indicate that this molecule stimulates the expression of tyrosinase by activation of cAMP/PKA signaling, subsequent phosphorylation of CREB, tyrosinase, TRP1 expression, and upregulation of MITF which further triggers melanogenesis.

4. Conclusions and Perspectives

Natural compounds have considerable benefits when evaluated for their biological effects. In this order, these molecules have been studied with a view to develop new therapeutic options to treat human diseases. According to the present review, many chemical analysis techniques revealed the presence of this flavonoid in a variety of pharmacologically active plants including Artemisia judaica, Cirsium japonicum, Lithocarpus dealbatus, Microtea debilis, and other species. In addition, different pharmacological and biological properties were investigated and the results showed that cirsimaritin has: a broad spectrum of action on a variety of pathogenic microorganisms in humans (Gram-positive and -negative bacteria, and fungi); anticancer effects against human colon carcinoma (HT-29), human gastric adenocarcinoma (AGS), human osteosarcoma (SaOs-2), murine fibrosarcoma (WEHI-164) and human fetal foreskin fibroblast (HFFF-P16), MCF-7, and PC-3 cell lines; and antidiabetic, antiparasitic, antioxidant, and anti-inflammatory effects. The mechanisms of action of cirsimaritin are not well understood, but it seems, according to the research highlighted, that this compound can exhibits its effects at different levels as it has been described above. Furthermore, further pharmacodynamic investigations should be conducted on cirsimaritin aimed at determining of its exact mode of action. Moreover, pharmacokinetic studies should also be conducted, on the one hand, to validate its: absorption, availability, metabolism, and elimination; on the other hand, to validate its safety via toxicological tests. Moreover, other perspectives concerning clinical trials of cirsimaritin against chronic inflammation would be a very proposing research perspectives to develop anti-inflammatory drugs. Moreover, a combination of cirsimaritin as an anticancer agent with used drugs in chemotherapy can give important results about the potent combinatory effects.

Author Contributions

Conceptualization, A.B., T.B., J.S.-G. and G.Z.; methodology, I.J., N.E.O., R.G., I.J., M.-N.P., B.K. and S.M.; software, validation, A.B., T.B. and M.A.S.; investigation, T.B., I.J., N.E.O., M.S.M. and J.S.-G.; writing—original draft preparation, I.J., K.H. (Kaoutar Harboul) and R.G.; writing—review and editing, A.B., N.E.O., M.R., K.H. (Kaoutar Harboul), M.A.S., M.S.M., G.Z., M.-N.P., B.K., S.M. and L.-H.L.; supervision, A.B. and K.H. (Khalil Hammani); funding acquisition, A.B., T.B., J.S.-G., G.Z., M.-N.P., B.K. and S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1I1A2066868), the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. 2020R1A5A2019413), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HF20C0116), and a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HF20C0038).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bouyahya, A.; El Omari, N.; Elmenyiy, N.; Guaouguaou, F.-E.; Balahbib, A.; Belmehdi, O.; Salhi, N.; Imtara, H.; Mrabti, H.N.; El-Shazly, M.; et al. Moroccan Antidiabetic Medicinal Plants: Ethnobotanical Studies, Phytochemical Bioactive Compounds, Preclinical Investigations, Toxicological Validations and Clinical Evidences; Challenges, Guidance and Perspectives for Future Management of Diabetes Worldwide. Trends Food Sci. Technol. 2021, 115, 147–254. [Google Scholar] [CrossRef]
  2. Bouyahya, A.; El Omari, N.; Hakkur, M.; El Hachlafi, N.; Charfi, S.; Balahbib, A.; Guaouguaou, F.-E.; Rebezov, M.; Maksimiuk, N.; Shariati, M.A.; et al. Sources, Health Benefits, and Biological Properties of Zeaxanthin. Trends Food Sci. Technol. 2021, 118, 519–538. [Google Scholar] [CrossRef]
  3. Bouyahya, A.; Guaouguaou, F.-E.; El Omari, N.; El Menyiy, N.; Balahbib, A.; El-Shazly, M.; Bakri, Y. Anti-Inflammatory and Analgesic Properties of Moroccan Medicinal Plants: Phytochemistry, in Vitro and in Vivo Investigations, Mechanism Insights, Clinical Evidences and Perspectives. J. Pharm. Anal. 2021, 12, 35–37. [Google Scholar] [CrossRef] [PubMed]
  4. Chamkhi, I.; Benali, T.; Aanniz, T.; El Menyiy, N.; Guaouguaou, F.-E.; El Omari, N.; El-Shazly, M.; Zengin, G.; Bouyahya, A. Plant-Microbial Interaction: The Mechanism and the Application of Microbial Elicitor Induced Secondary Metabolites Biosynthesis in Medicinal Plants. Plant Physiol. Biochem. 2021, 167, 269–295. [Google Scholar] [CrossRef] [PubMed]
  5. Rauf, A.; Akram, M.; Semwal, P.; Mujawah, A.A.H.; Muhammad, N.; Riaz, Z.; Munir, N.; Piotrovsky, D.; Vdovina, I.; Bouyahya, A.; et al. Antispasmodic Potential of Medicinal Plants: A Comprehensive Review. Oxid. Med. Cell. Longev. 2021, 2021, e4889719. [Google Scholar] [CrossRef] [PubMed]
  6. Pina, L.T.; Serafini, M.R.; Oliveira, M.A.; Sampaio, L.A.; Guimarães, J.O.; Guimarães, A.G. Carvone and Its Pharmacological Activities: A Systematic Review. Phytochemistry 2022, 196, 113080. [Google Scholar] [CrossRef]
  7. Aboulaghras, S.; Sahib, N.; Bakrim, S.; Benali, T.; Charfi, S.; Guaouguaou, F.-E.; Omari, N.E.; Gallo, M.; Montesano, D.; Zengin, G. Health Benefits and Pharmacological Aspects of Chrysoeriol. Pharmaceuticals 2022, 15, 973. [Google Scholar] [CrossRef]
  8. El Omari, N.; Jaouadi, I.; Lahyaoui, M.; Benali, T.; Taha, D.; Bakrim, S.; El Menyiy, N.; El Kamari, F.; Zengin, G.; Bangar, S.P. Natural Sources, Pharmacological Properties, and Health Benefits of Daucosterol: Versatility of Actions. Appl. Sci. 2022, 12, 5779. [Google Scholar] [CrossRef]
  9. Bakrim, S.; Machate, H.; Benali, T.; Sahib, N.; Jaouadi, I.; Omari, N.E.; Aboulaghras, S.; Bangar, S.P.; Lorenzo, J.M.; Zengin, G. Natural Sources and Pharmacological Properties of Pinosylvin. Plants 2022, 11, 1541. [Google Scholar] [CrossRef]
  10. Bouyahya, A.; Chamkhi, I.; Benali, T.; Guaouguaou, F.-E.; Balahbib, A.; El Omari, N.; Taha, D.; Belmehdi, O.; Ghokhan, Z.; El Menyiy, N. Traditional Use, Phytochemistry, Toxicology, and Pharmacology of Origanum majorana L. J. Ethnopharmacol. 2021, 265, 113318. [Google Scholar] [CrossRef]
  11. Bouyahya, A.; Mechchate, H.; Benali, T.; Ghchime, R.; Charfi, S.; Balahbib, A.; Burkov, P.; Shariati, M.A.; Lorenzo, J.M.; Omari, N.E. Health Benefits and Pharmacological Properties of Carvone. Biomolecules 2021, 11, 1803. [Google Scholar] [CrossRef] [PubMed]
  12. Bouyahya, A.; Chamkhi, I.; Guaouguaou, F.-E.; Benali, T.; Balahbib, A.; El Omari, N.; Taha, D.; El-Shazly, M.; El Menyiy, N. Ethnomedicinal Use, Phytochemistry, Pharmacology, and Food Benefits of Thymus Capitatus. J. Ethnopharmacol. 2020, 259, 112925. [Google Scholar] [CrossRef] [PubMed]
  13. El Omari, N.; Bakha, M.; Imtara, H.; Guaouguaoua, F.-E.; Balahbib, A.; Zengin, G.; Bouyahya, A. Anticancer Mechanisms of Phytochemical Compounds: Focusing on Epigenetic Targets. Environ. Sci. Pollut. Res. 2021, 28, 47869–47903. [Google Scholar] [CrossRef]
  14. El Omari, N.; Bakrim, S.; Bakha, M.; Lorenzo, J.M.; Rebezov, M.; Shariati, M.A.; Aboulaghras, S.; Balahbib, A.; Khayrullin, M.; Bouyahya, A. Natural Bioactive Compounds Targeting Epigenetic Pathways in Cancer: A Review on Alkaloids, Terpenoids, Quinones, and Isothiocyanates. Nutrients 2021, 13, 3714. [Google Scholar] [CrossRef] [PubMed]
  15. El Omari, N.; El Menyiy, N.; Zengin, G.; Goh, B.H.; Gallo, M.; Montesano, D.; Naviglio, D.; Bouyahya, A. Anticancer and Anti-Inflammatory Effects of Tomentosin: Cellular and Molecular Mechanisms. Separations 2021, 8, 207. [Google Scholar] [CrossRef]
  16. El Omari, N.; Ezzahrae Guaouguaou, F.; El Menyiy, N.; Benali, T.; Aanniz, T.; Chamkhi, I.; Balahbib, A.; Taha, D.; Shariati, M.A.; Zengin, G.; et al. Phytochemical and Biological Activities of Pinus halepensis Mill., and Their Ethnomedicinal Use. J. Ethnopharmacol. 2021, 268, 113661. [Google Scholar] [CrossRef]
  17. Cheriet, T.; Ben-Bachir, B.; Thamri, O.; Seghiri, R.; Mancini, I. Isolation and Biological Properties of the Natural Flavonoids Pectolinarin and Pectolinarigenin—A Review. Antibiotics 2020, 9, 417. [Google Scholar] [CrossRef]
  18. Williamson, G.; Barron, D.; Shimoi, K.; Terao, J. In Vitro Biological Properties of Flavonoid Conjugates Found in Vivo. Free Radic. Res. 2005, 39, 457–469. [Google Scholar] [CrossRef]
  19. Caltagirone, S.; Rossi, C.; Poggi, A.; Ranelletti, F.O.; Natali, P.G.; Brunetti, M.; Aiello, F.B.; Piantelli, M. Flavonoids Apigenin and Quercetin Inhibit Melanoma Growth and Metastatic Potential. Int. J. Cancer 2000, 87, 595–600. [Google Scholar] [CrossRef]
  20. Xing, N.; Meng, X.; Wang, S. Isobavachalcone: A Comprehensive Review of Its Plant Sources, Pharmacokinetics, Toxicity, Pharmacological Activities and Related Molecular Mechanisms. Phytother. Res. 2022, 36, 3120–3142. [Google Scholar] [CrossRef]
  21. Ferreira, A.; Pousinho, S.; Fortuna, A.; Falcão, A.; Alves, G. Flavonoid Compounds as Reversal Agents of the P-Glycoprotein-Mediated Multidrug Resistance: Biology, Chemistry and Pharmacology. Phytochem. Rev. 2015, 14, 233–272. [Google Scholar] [CrossRef]
  22. Dey, D.; Biswas, P.; Paul, P.; Mahmud, S.; Ema, T.I.; Khan, A.A.; Ahmed, S.Z.; Hasan, M.M.; Saikat, A.S.M.; Fatema, B. Natural Flavonoids Effectively Block the CD81 Receptor of Hepatocytes and Inhibit HCV Infection: A Computational Drug Development Approach. Mol. Divers. 2022, 1–14. [Google Scholar] [CrossRef] [PubMed]
  23. Thapa, A.; Chi, E.Y. Biflavonoids as Potential Small Molecule Therapeutics for Alzheimer’s Disease. In Natural Compounds as Therapeutic Agents for Amyloidogenic Diseases; Springer: Berlin/Heidelberg, Germany, 2015; pp. 55–77. [Google Scholar]
  24. Bouzid, N.; Moulis, C.; Fouraste, I. Flavones Libres de Artemisia Mesatlantica. Planta Med. 1982, 44, 157–158. [Google Scholar] [CrossRef] [PubMed]
  25. Saleh, N.A.M.; El-Negoumy, S.I.; Abou-zaid, M.M. Flavonoids of Artemisia Judaica, A. Monosperma and A. Herba-Alba. Phytochemistry 1987, 26, 3059–3064. [Google Scholar] [CrossRef]
  26. Hasrat, J.A.; De Bruyne, T.; De Backer, J.-P.; Vauquelin, G.; Vlietinck, A.J. (Phytolaccaceae) with Adenosine Antagonistic Properties in Rats: Leads for New Therapeutics in Acute Renal Failure. J. Pharm. Pharmacol. 1997, 49, 1150–1156. [Google Scholar] [CrossRef]
  27. Kelm, M.A.; Nair, M.G.; Strasburg, G.M.; De Witt, D.L. Antioxidant and Cyclooxygenase Inhibitory Phenolic Compounds from Ocimum Sanctum Linn. Phytomedicine 2000, 7, 7–13. [Google Scholar] [CrossRef]
  28. Ragasa, C.; Pendon, Z.; Veronica, S.; Rideout, J.A. Antimicrobial flavones from Coleus amboinicus. Philipp. J. Sci. 1999, 128, 347–352. [Google Scholar]
  29. Rijo, P.; Simões, M.F.; Duarte, A.; Rodríguez, B. Isopimarane Diterpenoids from Aeollanthus Rydingianus and Their Antimicrobial Activity. Phytochemistry 2009, 70, 1161–1165. [Google Scholar] [CrossRef]
  30. Marino, A.; Zengin, G.; Nostro, A.; Ginestra, G.; Dugo, P.; Cacciola, F.; Miceli, N.; Taviano, M.F.; Filocamo, A.; Bisignano, G.; et al. Antimicrobial Activities, Toxicity and Phenolic Composition of Asphodeline anatolica E. Tuzlaci Leaf Extracts from Turkey. Nat. Prod. Res. 2016, 30, 2620–2623. [Google Scholar] [CrossRef]
  31. Cazzola, R.; Cestaro, B. Antioxidant Spices and Herbs Used in Diabetes. In Diabetes: Oxidative Stress and Dietary Antioxidants; Elsevier: Amsterdam, The Netherlands, 2014; pp. 89–97. [Google Scholar]
  32. Shin, M.-S.; Park, J.Y.; Lee, J.; Yoo, H.H.; Hahm, D.-H.; Lee, S.C.; Lee, S.; Hwang, G.S.; Jung, K.; Kang, K.S. Anti-Inflammatory Effects and Corresponding Mechanisms of Cirsimaritin Extracted from Cirsium japonicum var. maackii Maxim. Bioorgan. Med. Chem. Lett. 2017, 27, 3076–3080. [Google Scholar] [CrossRef]
  33. Bower, A.M.; Real Hernandez, L.M.; Berhow, M.A.; De Mejia, E.G. Bioactive Compounds from Culinary Herbs Inhibit a Molecular Target for Type 2 Diabetes Management, Dipeptidyl Peptidase IV. J. Agric. Food Chem. 2014, 62, 6147–6158. [Google Scholar] [CrossRef] [PubMed]
  34. Yan, H.; Wang, H.; Ma, L.; Ma, X.; Yin, J.; Wu, S.; Huang, H.; Li, Y. Cirsimaritin Inhibits Influenza A Virus Replication by Downregulating the NF-ΚB Signal Transduction Pathway. Virol. J. 2018, 15, 88. [Google Scholar] [CrossRef] [PubMed]
  35. Ibañez, E.; Kubátová, A.; Señoráns, F.J.; Cavero, S.; Reglero, U.; Hawthorne, S.B. Subcritical Water Extraction of Antioxidant Compounds from Rosemary Plants. J. Agric. Food Chem. 2003, 51, 375–382. [Google Scholar] [CrossRef]
  36. Jipa, S.; Zaharescu, T.; Kappel, W.; Dǎneţ, A.F.; Popa, C.V.; Bumbac, M.; Gorghiu, L.M.; Maris, A.M. The Effects of γ-Irradiation on the Antioxidant Activity of Rosemary Extract. Optoelectron. Adv. Mater. Rapid Commun. 2009, 3, 1315–1320. [Google Scholar]
  37. Tasdemir, D.; Kaiser, M.; Brun, R.; Yardley, V.; Schmidt, T.J.; Tosun, F.; Rüedi, P. Antitrypanosomal and Antileishmanial Activities of Flavonoids and Their Analogues: In Vitro, in Vivo, Structure-Activity Relationship, and Quantitative Structure-Activity Relationship Studies. Antimicrob. Agents Chemother. 2006, 50, 1352–1364. [Google Scholar] [CrossRef]
  38. Al Ati, H.Y.; Fawzy, G.A.; El Gamal, A.A.; Khalil, A.T.; El Din El Tahir, K.; Abdel-Kader, M.S.; Gilani, A.-H. Phytochemical and Biological Evaluation of Buddleja Polystachya Growing in Saudi Arabia. Pak. J. Pharm. Sci. 2015, 28, 1533–1540. [Google Scholar]
  39. Morita, N.; Shimizu, M. Naokata Orita and Ineo Shimizu: Of Irsium of the Leaves Iavonoids Components Resources. XXI Runts (Compositae) in Japan. ÝCirsium MAKINO 1963, 83, 615–618. [Google Scholar]
  40. Shilin, Y.; Roberts, M.F.; Phillipson, J.D. Methoxylated Flavones and Coumarins from Artemisia Annua. Phytochemistry 1989, 28, 1509–1511. [Google Scholar] [CrossRef]
  41. Namba, T.; Hattori, M.; Takehana, Y.; Tsunezuka, M.; Tomimori, T.; Kizu, H.; Miyaichi, Y.; Medical, T. A flavone Artemisia capillaris. Phytochemistry 1983, 22, 1057–1058. [Google Scholar] [CrossRef]
  42. Sanz, J.F.; Barbera, O.; Alberto, M.J. Sesquiterpene lactones from Artemisia hispanica. Phytochemistry 1989, 28, 2163–2167. [Google Scholar] [CrossRef]
  43. Zhang, W.; Zhao, D.B.; Li, M.J.; Liu, X.H.; Wang, H.Q. Studies on Flavonoid Constituents from Herbs of Artemisia Ordosica II. Zhongguo Zhong Yao Za Zhi Zhongguo Zhongyao Zazhi China J. Chin. Mater. Medica 2006, 31, 1959–1961. [Google Scholar]
  44. Chandrasekharan, I.; Khan, H.A.; Ghanim, A. Flavonoids from Artemisia scoparia. Planta Med. 1981, 43, 310–311. [Google Scholar] [CrossRef]
  45. Chemesova, I.I.; Belenovskaya, L.M.; Markova, L.P. Flavonoids of Artemisia xanthochroa. Chem. Nat. Compd. 1985, 20, 748. [Google Scholar] [CrossRef]
  46. de Sousa Andrade, L.M.; de Oliveira, A.B.M.; Leal, A.L.A.B.; de Alcântara Oliveira, F.A.; Portela, A.L.; de Sousa Lima Neto, J.; de Siqueira-Júnior, J.P.; Kaatz, G.W.; da Rocha, C.Q.; Barreto, H.M. Antimicrobial Activity and Inhibition of the NorA Efflux Pump of Staphylococcus Aureus by Extract and Isolated Compounds from Arrabidaea brachypoda. Microb. Pathog. 2020, 140, 103935. [Google Scholar] [CrossRef] [PubMed]
  47. Weimann, C.; Göransson, U.; Pongprayoon-Claeson, U.; Claeson, P.; Bohlin, L.; Rimpler, H.; Heinrich, M. Spasmolytic Effects of Baccharis Conferta and Some of Its Constituents. J. Pharm. Pharmacol. 2001, 54, 99–104. [Google Scholar] [CrossRef] [PubMed]
  48. Graver, R.E.J.; Veitch, N.C. An 8-hydroxylated external flavone and its 8-O-glucoside from Becium grandiflorum. Phytochemistry 1998, 47, 7–10. [Google Scholar]
  49. Szoka, L.; Nazaruk, J.; Stocki, M.; Isidorov, V. Santin and Cirsimaritin from Betula Pubescens and Betula Pendula Buds Induce Apoptosis in Human Digestive System Cancer Cells. J. Cell. Mol. Med. 2021, 25, 11085–11096. [Google Scholar] [CrossRef]
  50. Sen, A.; Turan, S.O.; Bitis, L. Bioactivity-Guided Isolation of Anti-Proliferative Compounds from Endemic Centaurea kilaea. Pharm. Biol. 2016, 55, 541–546. [Google Scholar] [CrossRef]
  51. Al-Wahaibi, L.H.; Mahmood, A.; Khan, M.; Alkhathlan, H.Z. Phytochemical Analysis and Bioactivity Screening of Three Medicinal Plants of Saudi Arabia. Trop. J. Pharm. Res. 2020, 19, 371–376. [Google Scholar] [CrossRef]
  52. Diaa, Y.; August, W.F. Constituents of the Aerial Parts of Ruta Montana. Planta Med. 1995, 61, 279. [Google Scholar]
  53. Lee, D.; Kim, K.H.; Lee, J.; Hwang, G.S.; Lee, H.L.; Hahm, D.H.; Huh, C.K.; Lee, S.C.; Lee, S.; Kang, K.S. Protective Effect of Cirsimaritin against Streptozotocin-Induced Apoptosis in Pancreatic Beta Cells. J. Pharm. Pharmacol. 2017, 69, 875–883. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, J.S.; Paje, L.A.; Rodriguez, J.P.; Kang, K.S.; Hahm, D.H.; Shim, J.S.; Choi, Y.J.; Lee, S. Validation of an HPLC/UV Analysis Method for Cirsimaritin in Cirsium japonicum var. Maackii. Korean J. Pharmacogn. 2020, 51, 217–221. [Google Scholar] [CrossRef]
  55. Zhu, M.; Phillipson, J.D.; Greengrass, P.M.; Bowery, N.G. Chemical and Biological Investigation of the Root Bark of Clerodendrum mandarinorum. Planta Med. 1996, 62, 393–396. [Google Scholar] [CrossRef] [PubMed]
  56. Dawé, A.; Mbiantcha, M.; Yakai, F.; Jabeen, A.; Ali, M.S.; Lateef, M.; Ngadjui, B.T. Flavonoids and Triterpenes from Combretum Fragrans with Anti-Inflammatory, Antioxidant and Antidiabetic Potential. Z. Nat.-Sect. C J. Biosci. 2018, 73, 211–219. [Google Scholar] [CrossRef] [PubMed]
  57. Fattahi, M.; Nazeri, V.; Torras-Claveria, L.; Sefidkon, F.; Cusido, R.M.; Zamani, Z.; Palazon, J. Identification and Quantification of Leaf Surface Flavonoids in Wild-Growing Populations of Dracocephalum kotschyi by LC-DAD-ESI-MS. Food Chem. 2013, 141, 139–146. [Google Scholar] [CrossRef]
  58. Tahtah, Y.; Wubshet, S.G.; Kongstad, K.T.; Heskes, A.M.; Pateraki, I.; Møller, B.L.; Jäger, A.K.; Staerk, D. High-Resolution PTP1B Inhibition Profiling Combined with High-Performance Liquid Chromatography-High-Resolution Mass Spectrometry-Solid-Phase Extraction-Nuclear Magnetic Resonance Spectroscopy: Proof-of-Concept and Antidiabetic Constituents in Crude Extra. Fitoterapia 2016, 110, 52–58. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, Y.; Ho, D.K.; Cassady, J.M. Isolation of Potential Cancer Chemopreventive Agents. J. Nat. Prod. 1992, 5, 7–13. [Google Scholar]
  60. Lin, S.; Zhang, Q.W.; Zhang, N.N.; Zhang, Y.X. Determination of Flavonoids in Buds of Herba Artemisiae Scopariae by HPLC. Zhongguo Zhong Yao Za Zhi Zhongguo Zhongyao Zazhi China J. Chin. Mater. Medica 2005, 30, 591–594. [Google Scholar]
  61. Isobe, T.; Doe, M.; Morimoto, Y.; Nagata, K.; Ohsaki, A. The Anti-Helicobacter Pylori Flavones in a Brazilian Plant, Hyptis fasciculata, and the Activity of Methoxyflavones. Biol. Pharm. Bull. 2006, 29, 1039–1041. [Google Scholar] [CrossRef] [Green Version]
  62. Yu, Z.; Zhu, H.; Yang, X.; Sun, Q.; Hao, X. Study on Chemical Constituents from Incarvillea Arguta and Their Accelerating PC-12 Cell Differentiation. Zhongguo Zhong Yao Za Zhi Zhongguo Zhongyao Zazhi China J. Chin. Mater. Medica 2005, 30, 1335–1338. [Google Scholar]
  63. Bai, N.; He, K.; Roller, M.; Lai, C.S.; Shao, X.; Pan, M.H.; Bily, A.; Ho, C.T. Flavonoid Glycosides from Microtea debilis and Their Cytotoxic and Anti-Inflammatory Effects. Fitoterapia 2011, 82, 168–172. [Google Scholar] [CrossRef] [PubMed]
  64. Berim, A.; Park, J.J.; Gang, D.R. Unexpected Roles for Ancient Proteins: Flavone 8-Hydroxylase in Sweet Basil Trichomes Is a Rieske-Type, PAO-Family Oxygenase. Plant J. 2014, 80, 385–395. [Google Scholar] [CrossRef] [PubMed]
  65. Vieira, R.F.; Grayer, R.J.; Paton, A.; Simon, J.E. Genetic Diversity of Ocimum gratissimum L. Based on Volatile Oil Constituents, Flavonoids and RAPD Markers. Biochem. Syst. Ecol. 2001, 29, 287–304. [Google Scholar] [CrossRef]
  66. Bosabalidis, A.; Gabrieli, C.; Niopas, I. Flavone Aglycones in Glandular Hairs of Origanum × intercedens. Phytochemistry 1998, 49, 1549–1553. [Google Scholar] [CrossRef]
  67. Khaliq, S.; Volk, F.J.; Frahm, A.W. Phytochemical Investigation of Perovskia abrotanoides. Planta Med. 2007, 73, 77–83. [Google Scholar] [CrossRef] [PubMed]
  68. Zhong, J.; Huang, C.G.; Yu, Y.J.; Li, Z.Q.; Wang, W.; Huang, X.Z.; Liu, W.X.; Yuan, Y.; Jiang, Z.Y. Chemical Constituents from Perovskia Atriplicifolia. Zhongguo Zhong Yao Za Zhi Zhongguo Zhongyao Zazhi China J. Chin. Mater. Medica 2015, 40, 1108–1113. [Google Scholar] [CrossRef]
  69. De Azevedo Maia, G.L.; Dos Santos Falcão-Silva, V.; Aquino, P.G.V.; De Araújo-Júnior, J.X.; Tavares, J.F.; Da Silva, M.S.; Rodrigues, L.C.; De Siqueira-Júnior, J.P.; Barbosa-Filho, J.M. Flavonoids from Praxelis clematidea R.M. King and Robinson Modulate Bacterial Drug Resistance. Molecules 2011, 16, 4828–4835. [Google Scholar] [CrossRef] [PubMed]
  70. Abdelhalim, A.; Karim, N.; Chebib, M.; Aburjai, T.; Khan, I.; Johnston, G.A.R.; Hanrahan, J.R. Antidepressant, Anxiolytic and Antinociceptive Activities of Constituents from Rosmarinus Officinalis. J. Pharm. Pharm. Sci. 2015, 18, 448–459. [Google Scholar] [CrossRef]
  71. Cavero, S.; Jaime, L.; Martín-Álvarez, P.J.; Señoráns, F.J.; Reglero, G.; Ibañez, E. In Vitro Antioxidant Analysis of Supercritical Fluid Extracts from Rosemary (Rosmarinus officinalis L.). Eur. Food Res. Technol. 2005, 221, 478–486. [Google Scholar] [CrossRef]
  72. Pérez-Sánchez, A.; Borrás-Linares, I.; Barrajón-Catalán, E.; Arráez-Román, D.; González-Álvarez, I.; Ibáñez, E.; Segura-Carretero, A.; Bermejo, M.; Micol, V. Evaluation of the Intestinal Permeability of Rosemary (Rosmarinus officinalis L.) Extract Polyphenols and Terpenoids in Caco-2 Cell Monolayers. PLoS ONE 2017, 12, e0172063. [Google Scholar] [CrossRef] [Green Version]
  73. Xu, J.-H.; Lo, Y.M.; Chang, W.-C.; Huang, D.-W.; Wu, J.S.-B.; Jhang, Y.-Y.; Huang, W.-C.; Ko, C.-Y.; Shen, S.-C. Identification of Bioactive Components from Ruellia tuberosa L. on Improving Glucose Uptake in TNF-α-Induced Insulin-Resistant Mouse FL83B Hepatocytes. Evid. Based Complement. Alternat. Med. 2020, 2020, 6644253. [Google Scholar] [CrossRef] [PubMed]
  74. Srivedavyasasri, R.; Hayes, T.; Ross, S.A. Phytochemical and Biological Evaluation of Salvia apiana. Nat. Prod. Res. 2017, 31, 2058–2061. [Google Scholar] [CrossRef] [PubMed]
  75. Kanetis, L.; Exarchou, V.; Charalambous, Z.; Goulas, V. Edible Coating Composed of Chitosan and Salvia fruticosa Mill. Extract for the Control of Grey Mould of Table Grapes. J. Sci. Food Agric. 2017, 97, 452–460. [Google Scholar] [CrossRef] [PubMed]
  76. Exarchou, V.; Kanetis, L.; Charalambous, Z.; Apers, S.; Pieters, L.; Gekas, V.; Goulas, V. HPLC-SPE-NMR Characterization of Major Metabolites in Salvia fruticosa Mill. Extract with Antifungal Potential: Relevance of Carnosic Acid, Carnosol, and Hispidulin. J. Agric. Food Chem. 2015, 63, 457–463. [Google Scholar] [CrossRef]
  77. Kavvadias, D.; Monschein, V.; Sand, P.; Riederer, P.; Schreier, P. Constituents of Sage (Salvia officinalis) with in Vitro Affinity to Human Brain Benzodiazepine Receptor. Planta Med. 2003, 69, 113–117. [Google Scholar] [CrossRef]
  78. Miski, M.; Ulubelen, A.; Johansson, C.; Mabry, T.J. Antibacterial Activity Studies of Flavonoids from Salvia palaestina. J. Nat. Prod. 1983, 46, 874–875. [Google Scholar] [CrossRef]
  79. Cottiglia, F.; Casu, L.; Bonsignore, L.; Casu, M.; Floris, C.; Sosa, S.; Altinier, G.; Della Loggia, R. Topical Anti-Inflammatory Activity of Flavonoids and a New Xanthone from Santolina insularis. Z. Nat.-Sect. C J. Biosci. 2005, 60, 63–66. [Google Scholar] [CrossRef]
  80. Malmir, M.; Gohari, A.R.; Saeidnia, S.; Silva, O. A New Bioactive Monoterpene-Flavonoid from Satureja khuzistanica. Fitoterapia 2015, 105, 107–112. [Google Scholar] [CrossRef]
  81. Shafiq, N.; Riaz, N.; Ahmed, S.; Ashraf, M.; Ejaz, S.A.; Ahmed, I.; Saleem, M.; Touseef, M.I.; Tareen, R.B.; Jabbar, A. Bioactive Phenolics from Seriphidium stenocephalum. J. Asian Nat. Prod. Res. 2013, 15, 286–293. [Google Scholar] [CrossRef]
  82. Beer, M.F.; Frank, F.M.; Germán Elso, O.; Ernesto Bivona, A.; Cerny, N.; Giberti, G.; Luis Malchiodi, E.; Susana Martino, V.; Alonso, M.R.; Patricia Sülsen, V.; et al. Trypanocidal and Leishmanicidal Activities of Flavonoids Isolated from Stevia satureiifolia var. Satureiifolia. Pharm. Biol. 2016, 54, 2188–2195. [Google Scholar] [CrossRef] [Green Version]
  83. Ren, X.; Bao, Y.; Zhu, Y.; Liu, S.; Peng, Z.; Zhang, Y.; Zhou, G. Isorhamnetin, Hispidulin, and Cirsimaritin Identified in Tamarix Ramosissima Barks from Southern Xinjiang and Their Antioxidant and Antimicrobial Activities. Molecules 2019, 24, 390. [Google Scholar] [CrossRef] [PubMed]
  84. Polatoğlu, K.; Karakoç, Ö.C.; Demirci, F.; Gökçe, A.; Gören, N. Chemistry and Biological Activities of Tanacetum chiliophyllum var. oligocephalum Extracts. J. AOAC Int. 2013, 96, 1222–1227. [Google Scholar] [CrossRef] [PubMed]
  85. Stefkov, G.; Kulevanova, S.; Miova, B.; Dinevska-Kjovkarovska, S.; Mølgaard, P.; Jäger, A.K.; Josefsen, K. Effects of Teucrium polium spp. Capitatum Flavonoids on the Lipid and Carbohydrate Metabolism in Rats. Pharm. Biol. 2011, 49, 885–892. [Google Scholar] [CrossRef]
  86. Ben Sghaier, M.; Skandrani, I.; Nasr, N.; Franca, M.G.D.; Chekir-Ghedira, L.; Ghedira, K. Flavonoids and Sesquiterpenes from Tecurium ramosissimum Promote Antiproliferation of Human Cancer Cells and Enhance Antioxidant Activity: A Structure-Activity Relationship Study. Environ. Toxicol. Pharmacol. 2011, 32, 336–348. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, R.F.; Yang, X.W.; Ma, C.M.; Liu, H.Y.; Shang, M.Y.; Zhang, Q.Y.; Cai, S.Q.; Park, J.H. Trollioside, a New Compound from the Flowers of Trollius chinensis. J. Asian Nat. Prod. Res. 2004, 6, 139–144. [Google Scholar] [CrossRef] [PubMed]
  88. Nyiligira, E.; Viljoen, A.M.; Van Heerden, F.R.; Van Zyl, R.L.; Van Vuuren, S.F.; Steenkamp, P.A. Phytochemistry and in Vitro Pharmacological Activities of South African Vitex (Verbenaceae) Species. J. Ethnopharmacol. 2008, 119, 680–685. [Google Scholar] [CrossRef] [PubMed]
  89. Hussain, W.; Amir, A.; Rasool, N. Computer-Aided Study of Selective Flavonoids against Chikungunya Virus Replication Using Molecular Docking and DFT-Based Approach. Struct. Chem. 2020, 31, 1363–1374. [Google Scholar] [CrossRef]
  90. Kiran, G.; Karthik, L.; Devi, M.S.; Sathiyarajeswaran, P.; Kanakavalli, K.; Kumar, K.M.; Kumar, D.R. In Silico Computational Screening of Kabasura Kudineer-Official Siddha Formulation and JACOM against SARS-CoV-2 Spike Protein. J. Ayurveda Integr. Med. 2020, 13, 100324. [Google Scholar] [CrossRef]
  91. Tasdemir, D.; Lack, G.; Brun, R.; Rüedi, P.; Scapozza, L.; Perozzo, R. Inhibition of Plasmodium f Alciparum Fatty Acid Biosynthesis: Evaluation of FabG, FabZ, and FabI as Drug Targets for Flavonoids. J. Med. Chem. 2006, 49, 3345–3353. [Google Scholar] [CrossRef]
  92. Quintanilla-Licea, R.; Vargas-Villarreal, J.; Verde-Star, M.J.; Rivas-Galindo, V.M.; Torres-Hernández, Á.D. Antiprotozoal Activity against Entamoeba Histolytica of Flavonoids Isolated from Lippia Graveolens Kunth. Molecules 2020, 25, 2464. [Google Scholar] [CrossRef]
  93. Kolak, U.; Hacibekiroǧlu, I.; Öztürk, M.; Özgökçe, F.; Topçu, G.; Ulubelen, A. Antioxidant and Anticholinesterase Constituents of Salvia Poculata. Turk. J. Chem. 2009, 33, 813–823. [Google Scholar] [CrossRef]
  94. Liu, C.Z.; Murch, S.J.; El-Demerdash, M.; Saxena, P.K. Artemisia Judaica L.: Micropropagation and Antioxidant Activity. J. Biotechnol. 2004, 110, 63–71. [Google Scholar] [CrossRef] [PubMed]
  95. Burki, S.; Mehjabeen; Burki, Z.G.; Shah, Z.A.; Imran, M.; Khan, M. Phytochemical Screening, Antioxidant, and in Vivo Neuropharmacological Effect of Monotheca buxifolia (Falc.) Barks Extract. Pak. J. Pharm. Sci. 2018, 31, 1519–1528. [Google Scholar] [PubMed]
  96. Kuo, C.-F.; Su, J.-D.; Chiu, C.-H.; Peng, C.-C.; Chang, C.-H.; Sung, T.-Y.; Huang, S.-H.; Lee, W.-C.; Chyau, C.-C. Anti-Inflammatory Effects of Supercritical Carbon Dioxide Extract and Its Isolated Carnosic Acid from Rosmarinus officinalis Leaves. J. Agric. Food Chem. 2011, 59, 3674–3685. [Google Scholar] [CrossRef]
  97. Moghaddam, G.; Ebrahimi, S.A.; Rahbar-Roshandel, N.; Foroumadi, A. Antiproliferative Activity of Flavonoids: Influence of the Sequential Methoxylation State of the Flavonoid Structure: Antiproliferative flavonoids from dracocephalum kotschyi. Phytother. Res. 2012, 26, 1023–1028. [Google Scholar] [CrossRef]
  98. Pathak, G.; Singh, S.; Kumari, P.; Raza, W.; Hussain, Y.; Meena, A. Cirsimaritin, a Lung Squamous Carcinoma Cells (NCIH-520) Proliferation Inhibitor. J. Biomol. Struct. Dyn. 2020, 39, 3312–3323. [Google Scholar] [CrossRef]
  99. Quan, Z.; Gu, J.; Dong, P.; Lu, J.; Wu, X.; Wu, W.; Fei, X.; Li, S.; Wang, Y.; Wang, J.; et al. Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress and Mitochondrial Dysfunction Contribute to Cirsimaritin-Induced Apoptosis in Human Gallbladder Carcinoma GBC-SD Cells. Cancer Lett. 2010, 295, 252–259. [Google Scholar] [CrossRef]
  100. Patel, D.K. Health Beneficial Aspect and Therapeutic Potential of Cirsimaritin in the Medicine for the Treatment of Human Health Complications. Curr. Bioact. Compd. 2022, 18, 27–38. [Google Scholar] [CrossRef]
  101. Awad, B.M.; Abd-Alhaseeb, M.M.; Habib, E.S.; Ibrahim, A.K.; Ahmed, S.A. Antitumor Activity of Methoxylated Flavonoids Separated from Achillea fragrantissima Extract in Ehrlich’s Ascites Carcinoma Model in Mice. J. Herbmed Pharmacol. 2020, 9, 28–34. [Google Scholar] [CrossRef]
  102. Kim, H.; Kim, I.; Dong, Y.; Lee, I.-S.; Kim, J.; Kim, J.-S.; Woo, J.-T.; Cha, B.-Y. Melanogenesis-Inducing Effect of Cirsimaritin through Increases in Microphthalmia-Associated Transcription Factor and Tyrosinase Expression. Int. J. Mol. Sci. 2015, 16, 8772–8788. [Google Scholar] [CrossRef] [Green Version]
  103. Lee, D.; Jung, Y.; Baek, J.Y.; Shin, M.-S.; Lee, S.; Hahm, D.-H.; Lee, S.C.; Shim, J.S.; Kim, S.N.; Kang, K.S. Cirsimaritin Contributes to the Estrogenic Activity of Cirsium Japonicum var. Maackii through the Activation of Estrogen Receptor α: Estrogenic Compounds of Cirsium Japonicum var. Maackii. Bull. Korean Chem. Soc. 2017, 38, 1486–1490. [Google Scholar] [CrossRef]
  104. Manurung, K.; Sulastri, D.; Zubir, N.; Ilyas, S. In Silico Anticancer Activity and in Vitro Antioxidant of Flavonoids in Plectranthus amboinicus. Pharmacogn. J. 2020, 12, 1573–1577. [Google Scholar] [CrossRef]
  105. Plochmann, K.; Korte, G.; Koutsilieri, E.; Richling, E.; Riederer, P.; Rethwilm, A.; Schreier, P.; Scheller, C. Structure–Activity Relationships of Flavonoid-Induced Cytotoxicity on Human Leukemia Cells. Arch. Biochem. Biophys. 2007, 460, 1–9. [Google Scholar] [CrossRef] [PubMed]
  106. Yeon Park, J.; Young Kim, H.; Shibamoto, T.; Su Jang, T.; Cheon Lee, S.; Suk Shim, J.; Hahm, D.-H.; Lee, H.-J.; Lee, S.; Sung Kang, K. Beneficial Effects of a Medicinal Herb, Cirsium japonicum var. Maackii, Extract and Its Major Component, Cirsimaritin on Breast Cancer Metastasis in MDA-MB-231 Breast Cancer Cells. Bioorgan. Med. Chem. Lett. 2017, 27, 3968–3973. [Google Scholar] [CrossRef] [PubMed]
  107. Zehra, B.; Ahmed, A.; Sarwar, R.; Khan, A.; Farooq, U.; Abid Ali, S.; Al-Harrasi, A. Apoptotic and Antimetastatic Activities of Betulin Isolated from Quercus incana against Non-Small Cell Lung Cancer Cells. Cancer Manag. Res. 2019, 11, 1667–1683. [Google Scholar] [CrossRef]
  108. Abdalla, S.S.; Zarga, M.A. Effects of Cirsimaritin, a Flavone Isolated from Artemisia judaica, on Isolated Guinea-Pig Ileum. Planta Med. 1987, 53, 322–324. [Google Scholar] [CrossRef]
  109. González-Trujano, M.E.; Hernández-Sánchez, L.Y.; Muñoz Ocotero, V.; Dorazco-González, A.; Guevara Fefer, P.; Aguirre-Hernández, E. Pharmacological Evaluation of the Anxiolytic-like Effects of Lippia graveolens and Bioactive Compounds. Pharm. Biol. 2017, 55, 1569–1576. [Google Scholar] [CrossRef]
  110. Wang, J.-P.; Chang, L.-C.; Hsu, M.-F.; Chen, S.-C.; Kuo, S.-C. Inhibition of Formyl-Methionyl-Leucyl-Phenylalanine-Stimulated Respiratory Burst by Cirsimaritin Involves Inhibition of Phospholipase D Signaling in Rat Neutrophils. Naunyn. Schmiedebergs Arch. Pharmacol. 2002, 366, 307–314. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of Cirsimaritin (drawn by Chem-Draw).
Figure 1. Chemical structure of Cirsimaritin (drawn by Chem-Draw).
Antioxidants 11 01842 g001
Figure 2. Different biological and pharmacological properties induced by cirsimaritin.
Figure 2. Different biological and pharmacological properties induced by cirsimaritin.
Antioxidants 11 01842 g002
Figure 3. Anti-inflammatory mechanisms induced by cirsimaritin.
Figure 3. Anti-inflammatory mechanisms induced by cirsimaritin.
Antioxidants 11 01842 g003
Figure 4. Antidiabetic mechanisms of cirsimaritin.
Figure 4. Antidiabetic mechanisms of cirsimaritin.
Antioxidants 11 01842 g004
Figure 5. Anticancer mechanisms of cirsimaritin.
Figure 5. Anticancer mechanisms of cirsimaritin.
Antioxidants 11 01842 g005
Table 1. Plant sources of cirsimaritin.
Table 1. Plant sources of cirsimaritin.
PlantsPart UsedBotanical FamiliesType of ExtractConcentration/FractionReferences
Aeollanthus rydingianusAerial partsLamiaceaeMe2CO Extract58 mg[29]
Artemisia annuaLeaves
Stems
AsteraceaeMethanolic extract1.45 mg[40]
Artemisia capillarisSpikesAsteraceaeMethanolic extractnot specified[41]
Artemisia hispanicaAerial partsAsteraceaeMethanolic extractnot specified[42]
Artemisia judaicaLeaves
Stems
AsteraceaeEthanolic extractnot specified[25]
Artemisia meatlanticaeAerial partsAsteraceaeEther extractnot specified[24]
Artemisia monospermaLeaves
Stems
AsteraceaeEthanolic extractnot specified[25]
Artemisia ordosica IInot specifiedAsteraceaenot specifiednot specified[43]
Artemisia scopariaDried inflorescenceAsteraceaeChloroform extractnot specified[44]
Artemisia xanthochroaEpigeal partsAsteraceaeEthanolic extractnot specified[45]
Arrabidaea brachypodaFlowersBignoniaceaeEthanolic extractnot specified[46]
Asphodeline anatolicaLeavesLiliaceaeAcetone and methanol extractnot specified[30]
Baccharis confertaAerial partsAsteraceaeEthanolic extractnot specified[47]
Becium grandiflorumLeavesLamiaceaeMethanolic extractnot specified[48]
Betula pendulaBudsBetulaceaeCarbon dioxide supercritical extraction3.79 mg/g[49]
Betula pubescensBudsBetulaceaeCarbon dioxide supercritical extraction4.21 mg/g[49]
Buddleja polystachyaAerial partsBuddlejaceaeEthanolic extract (cold maceration)not specified[38]
Centuarea kilaeaAerial partsAsteraceaeChloroform extract10.2 mg[50]
Centaurea pseudosinaicaEntire plant (leaves, flowers, stems)AsteraceaeEthanolic extract0.52 g[51]
Centaurea scopariaAerial partsAsteraceaeEthanolic extract10 mg[52]
Cirsium martimumLeavesAsteraceaeMethanolic extractnot specified[39]
Cirsium japonicumAerial parts
Leaves
AsteraceaeEthanolic
Extract
6.24 mg/g[32]
6.24 mg/g[53]
37.13 mg/g[54]
Clerodendrum mandarinorumRoot barkLamiaceaeEthanolic
Extract
50 mg[55]
Combretum fragransLeavesCombretaceaeMethanolic extractnot specified[56]
Dracocephalum kotschyiLeavesLamiaceaeDiethyl Ether extract97.3–637.6 µg/g[57]
Eremophila lucidaLeavesMyoporaceaeEthyl acetate extractnot specified[58]
Eriodictyon californicumLeavesHydrophyllaceaeEthanol extract3.85 mg[59]
Herba artemisiae ScopariaeBudsCompositaeEthyl acetate extract (ultrasonic)not specified[60]
Hyptis fasciculataAerial partsLabiataeChloroform and methanol extract19 mg[61]
Incarvillea argutanot specifiedBignoniaceaenot specifiednot specified[62]
Microtea debilisWhole plant
Aerial parts
MicroteaceaeAqueous & ethanol extractEthanol extract0.7 mg/mL
65 mg
[26]
[63]
Ocimum basilicumTricomsLamiaceaeCrude protein extract with HClnot specified[64]
Ocimum gratissimumAbove-ground biomassLamiaceaeClevenger apparatus10% of the total flavonoids[65]
Origanum intercedensLeavesLamiaceaeChloroform extractnot specified[66]
Osimum sanctumLeaves
Stems
LabiataeChloroform extract1 mg[27]
Perovskia abrotanoiesAerial partsLamiaceaeMethanolic extract10 mg[67]
Perovskia atriplicifoliaLeavesLamiaceaeEthanolic
Extract
not specified[68]
Praxelis clematideaAerial partsAsteraceaeEthanolic
Extract (exhaustive maceration)
not specified[69]
Rosmarinus officinalisLeavesLamiaceaeEthyl acetate extractnot specified[70]
Subcritical extraction with water1.72%[35]
Supercritical fluid extraction0.54–17.59%[71]
Ethanolic extractnot specified[36]
Super critical fluid extractionnot specified[72]
Ruellia tuberosaLeaves & stemsAcanthaceaeMethanolic extract805 µg/g[73]
Salvia apianaAerial partsLamiaceaeAqueous ethanolic extractnot specified[74]
Salvia fruticosaAerial partsLamiaceaeAcetonic extract (Soxtec system)not specified[75]
Ethyl acetate extract (Soxtec extraction)10.4 mg/g[76]
Salvia officinalisLeavesLamiaceaeMethanolic extraction (ultrasonic bath)194 mg[77]
[31]
Salvia palaestinaLeavesLamiaceaeBenzene extract (Soxhlet)30 mg[78]
Santolina insularisLeavesAsteraceaeMethanolic extract6.9 mg[79]
Satureja khuzistanicaAerial parts LamiaceaeEthyl acetate extraction5 mg[80]
Seriphidium stenocephalumnot specifiedAsteraceaeMethanolic extract15 mg[81]
Stevia satureiifoliaAerial partsAsteraceaeDichloromethane extract1.9%[82]
Tamarix ramosissimaBarkTamaricaceaeEthanolic extract13.35 µg/mg[83]
Tanacetum chiliophyllumStemsCompositaeEthyl acetate extract36 mg[84]
Teucrium poliumAerial parts LamiaceaeAlcohol extractionnot specified[85]
Teucrium ramosissimumLeavesLamiaceaeChloroformic extractnot specified[86]
Trollius chinensisFlowersRanunculaceaeEthanolic extract14 mg[87]
Vitex rehmanniiAerial partsVerbenaceaeAcetone extract5 mg[88]
Table 2. Antibacterial and antifungal effects of cirsimaritin.
Table 2. Antibacterial and antifungal effects of cirsimaritin.
Methods UsedStrains TestedKey ResultsReferences
Disk diffusion assayEscherichia coliMIC = 31.25 μg/mL, MBC = 125 μg/mL[78]
Klebsiella pneumoniaMIC = 31.25 μg/mL, MBC = 125 μg/mL
Pseudomonas aeruginosaMIC = 45 μg/mL, MBC = 90 μg/mL
Proteus vulgarisMIC = 31.25 μg/mL, MBC = 125 μg/mL
Staphylococcus aureusMIC = 31.25 μg/mL, MBC = 125 μg/mL
Staphylococcus epidermisMIC = 62.5 μg/mL, MBC = 125 μg/mL
Disk diffusion methodAspergillus nigerΦ = 13 mm at 40 µg[28]
Basilus subtilisΦ = 0 mm at 40 µg
Candida albicansΦ = 12 mm at 40 µg
Escherichia coliΦ = 0 mm at 40 µg
Pseudomonas aeruginosaΦ = 13 mm at 40 µg
Staphylococcus aureusΦ = 11 mm at 40 µg
Trichophyton mentagrophytesΦ = 14 mm at 40 µg
Agar diffusion methodCandida albicansNo activity[29]
Escherichia coliNo activity
Enterococcus hiraeGrowth zone inhibition
Mycobacterium smegmatisNo activity
Pseudomonas aeruginosaNo activity
Staphylococcus aureusGrowth zone inhibition
Micro-dilution techniqueAspergilus fumigatusMIC = 1.95 μg/mL[51]
Bacillus subtilisMIC = 0.03 μg/mL
Candida albicansMIC = 1.95 μg/mL
Escherichia coliMIC = 11.25 μg/mL
Geotrichum candidumMIC = 0.48 μg/mL
Pseudomonas aeruginosaMIC = 50.0 μg/mL
Streptococcus pneumoniaeMIC = 7.81 μg/mL
Syncephalastrum racemosumMIC = 12.5 μg/mL
Micro-dilution methodBacillus cereusMIC = 5 mg/mL, MBC = 20 mg/mL[83]
Escherichia coliMIC = 10 mg/mL, MBC = 25 mg/mL
Listeria monocytogenesMIC = 5 mg/mL, MBC = 10 mg/mL
Pseudomonas aeruginosaMIC > 10 mg/mL, MBC = NA
Salmonella typhimuriumMIC > 10 mg/mL, MBC = NA
Shigella castellaniMIC = 5 mg/mL, MBC = 15 mg/mL
Staphylococcus aureusMIC = 5 mg/mL, MBC = 15 mg/mL
MIC: Minimum Inhibitory Concentrations (mg/mL or μg/mL); MBC: Minimum bactericidal Concentrations (mg/mL or μg/mL); Φ: The diameter of inhibition zones (mm); NA: No activity.
Table 3. Antioxidant activities of cirsimaritin.
Table 3. Antioxidant activities of cirsimaritin.
Used MethodKey ResultsReferences
DPPH radical scavenging activityEC50 = 11.3 µg/mL[35]
β-carotene bleaching, superoxide anion radical, and ABTS cation radical scavenging activity assaysNo antioxidant activity[93]
ABTS assayTEAC (µM) = 2.04[86]
CUPRAC assayTEAC (µM) = 4.7
RP (Reducing power) assayTEAC (µM) = 0.95
FRAP assayTEAC (µM) = 0.625
DPPH Scavenging activityInhibition efficiency (%) = 80–100 at a concentration of 100 µg/mL[53]
FRAP assay AC = 203.39 to 681.27 µmol Fe2+/100 g DW at a concentration of 97.38–637.66 µg/g DW[57]
DPPH assay Significantly higher capacity to detoxify oxygen radicals[94]
DPPH scavenging% inhibition (at 500 µg/mL) = 89.55[95]
Superoxide scavenging% inhibition (at 500 µg/mL) = 82.10
Hydrogen peroxide scavenging% inhibition (at 500 µg/mL) = 80.55
DPPH assayIC50 = 55.9 µM[56]
Table 4. Anticancer activity of cirsimaritin.
Table 4. Anticancer activity of cirsimaritin.
OriginBiological Model (In Vitro or In Vivo)Experimental ApproachResults and Mechanism of ActionReferences
Synthetic compoundhuman cancer cell lines namely COLO-205, MDA-MB-231, HaCaT, K562, A431, A549, MCF-7, PC-3, NCIH- 520, normal cell lines WRL-68, HEK 293 and L132 and in primary macrophagesMTT assay
Inhibitory potential and binding interaction with the selected targets were analyzed through in vitro and in silico analysis
Inhibited the growth of NCIH-520 cell-line (IC50 23.29 μM)
Induced apoptosis
Inhibited the activity of ODC and CATD
Exhibited a good binding in silico score with the selected targets and it non-mutagenic
[98]
Synthetic compoundgallbladder carcinoma cell lines GBC-SD and GBCSD18H cells, gastric carcinoma cell line BGC-823 cells, and hepatoma cell line SMMC-7721 cellsCytotoxicity assay
Cell apoptosis assay
Cell mitochondrial membrane potential assay
Subcellular fractionation
Western blot
Small interference RNA
RT)-PCR
Detection of intracellular ROS
Inhibited the growth of tumor cells
Induced mitochondrial apoptosis in GBC-SD cells
Triggered endoplasmic reticulum (ER) stress Down-regulated the phosphorylation of Akt
[99]
Centaurea kilaeaone normal cell line (L-929, mouse
fibroblast)
three human cancer cell lines (Hela, cervix carcinoma; MCF-7, breast carcinoma; PC-3, prostate carcinoma
MTT assayInhibited the growth of MCF-7 and PC-3[50]
Teucrium ramosissimumEhrlich’s ascites carcinoma model in mice(5, 10, 20 mg/kg/d, orally)Reduced tumor weight compared to EAC-control and cisplatin groups
Induced tumor cell necrosis
Reduced significantly the level of TNF-α in serum
[101]
Teucrium ramosissimumhuman chronic myelogenous K562 cellsMTT assayExhibited an antiproliferative effect of human cancer cells IC50 = 1.015 × 107 mol/mL[86]
Lithocarpus dealbatusMurine melanoma B16F10 cells (CRL-6415)Cell Morphology and Cell Viability Measurement
Measurement of Cellular Tyrosinase Activity
Melanin Content Measurement
Western Blotting
Stimulated melanogenesis in B16F10 cells
Activated of CREB as well as upregulation of MITF and tyrosinase expression
activated by cAMP signaling
[102]
Cirsium japonicum var. maackiihuman breast cancer (MCF-7) cell-basedTransactivation assay
Proliferative activity
Exerted beneficial effects on MCF-7 cells
Increased estrogenic activity
[103]
Plectranthus amboinicusCancer P-Glycoprotein-1, Cyclin Dependent Kinase-2, and Phosphoinositide-3-Kinase receptorsIn silico anticancer TestExhibited an important strong anti-cancer effect[104]
Dracocephalum kotschyi Boiss.AGS, HT-29, HL60, SaOs-2, WEHI-164 and HFFF-P16 cellsMTT assayExhibited and antiproliferative activity of malignant cells [97]
IsolatedHuman T lymphoblasts (Jurkat Clone E6-1)Cytotoxicity experiments
Flow cytometry
Induced cytotoxicity
EC50 = 66.8 µM (24 h)
EC50 = 44.4 µM (48 h)
[105]
Cirsium japonicumBreast cancerCell proliferation assay
Tube-formation assay
Western blot analysis
Inhibited the viability of HUVECs in a dose-dependent manner
Inhibited angiogenesis by downregulation of VEGF, p-Akt and p-ERK in MDA-MB-231 cells
[106]
Betula pubescens and Betula pendulagastric (AGS), colon (DLD-1) and liver (HepG2) cancer cellsCell viability assay
DNA biosynthesis
Colony formation assay
Apoptosis assay
Western immunoblot
Immunofluorescence microscopy
Induced apoptosis
Activated caspase-3, caspase-7, caspase-8 and caspase-9 expression
Upregulated p53 expression
[49]
Quercus incananon-small cell lung carcinoma (NCI-H460) and normal mouse fibroblast (NIH-3T3) cell lines.mRNA extraction and qRT-PCR
Colony formation assay
Flow cytometry analysis
Cell cycle analysis
Western blot analysis
Induced antiproliferative against NIH 3T3
(IC50 = 26.23 ± 0.053 μM) and in NCI-H460 (IC50 = 38.84 ± 0.037 μM)
[107]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Benali, T.; Jaouadi, I.; Ghchime, R.; El Omari, N.; Harboul, K.; Hammani, K.; Rebezov, M.; Shariati, M.A.; Mubarak, M.S.; Simal-Gandara, J.; et al. The Current State of Knowledge in Biological Properties of Cirsimaritin. Antioxidants 2022, 11, 1842. https://doi.org/10.3390/antiox11091842

AMA Style

Benali T, Jaouadi I, Ghchime R, El Omari N, Harboul K, Hammani K, Rebezov M, Shariati MA, Mubarak MS, Simal-Gandara J, et al. The Current State of Knowledge in Biological Properties of Cirsimaritin. Antioxidants. 2022; 11(9):1842. https://doi.org/10.3390/antiox11091842

Chicago/Turabian Style

Benali, Taoufiq, Imane Jaouadi, Rokia Ghchime, Nasreddine El Omari, Kaoutar Harboul, Khalil Hammani, Maksim Rebezov, Mohammad Ali Shariati, Mohammad S. Mubarak, Jesus Simal-Gandara, and et al. 2022. "The Current State of Knowledge in Biological Properties of Cirsimaritin" Antioxidants 11, no. 9: 1842. https://doi.org/10.3390/antiox11091842

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop