Next Article in Journal
MoS2 as a Co-Catalyst for Photocatalytic Hydrogen Production: A Mini Review
Previous Article in Journal
Insight into the Progress on Natural Dyes: Sources, Structural Features, Health Effects, Challenges, and Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Review of Rosmarinic Acid: From Phytochemistry to Pharmacology and Its New Insight

1
School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
2
Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
3
School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
*
Author to whom correspondence should be addressed.
Submission received: 27 April 2022 / Revised: 13 May 2022 / Accepted: 18 May 2022 / Published: 20 May 2022

Abstract

:
Polyphenolic acids are the widely occurring natural products in almost each herbal plant, among which rosmarinic acid (RA, C18H16O8) is well-known, and is present in over 160 species belonging to many families, especially the Lamiaceae. Aside from this herbal ingredient, dozens of its natural derivatives have also been isolated and characterized from many natural plants. In recent years, with the increasing focus on the natural products as alternative treatments, a large number of pharmacological studies have been carried out to demonstrate the various biological activities of RA such as anti-inflammation, anti-oxidation, anti-diabetes, anti-virus, anti-tumor, neuroprotection, hepatoprotection, etc. In addition, investigations concerning its biosynthesis, extraction, analysis, clinical applications, and pharmacokinetics have also been performed. Although many achievements have been made in various research aspects, there still exist some problems or issues to be answered, especially its toxicity and bioavailability. Thus, we hope that in the case of natural products, the present review can not only provide a comprehensive understanding on RA covering its miscellaneous research fields, but also highlight some of the present issues and future perspectives worth investigating later, in order to help us utilize this polyphenolic acid more efficiently, widely, and safely.

1. Introduction

In recent years, with in-depth studies of the plants, natural products have increasingly attracted the attention of researchers in many fields. Rosmarinic acid (RA, C18H16O8, Figure 1) is an interesting and well-known representative. Regarding its chemical structure, this naturally-occurring phenol acid is considered as an ester, the esterification product of a caffeic acid and a 3,4-dihydroxyphenyl lactic acid. To our knowledge, it was from Rosmarinus officinalis L. that RA was first isolated and identified by two Italian scientists, Scarpati and Oriente, and was named according to the name of this herbal plant [1]. From then on, RA has been successively found in more than 160 plants belonging to Lamiaceae, Boraginaceae, Apiaceae, etc. It has also been investigated for its miscellaneous pharmacological activities including anti-oxidative activity, anti-inflammatory activity, anti-viral activity, anti-diabetic activity, anti-tumor activity, and neuroprotective activity in many in vitro and in vivo studies. Due to its higher content and similar bioactivity to phytomedicines, RA is employed as the quality indicative component for them including Perilla frutescens (L.) Britt fruits and stems, Prunella vulgaris L. spikes, and Sarcandra glabra (Thunb.) Nakai whole plants in the Chinese Pharmacopoeia, Melissa officinalis L. leaves and Eclipta prostrata (L.) L. aerial parts in the European Pharmacopoeia, and Rosmarinus officinalis L. leaves in the United States Pharmacopoeia.
As a variety of studies have been performed and some achievements have been made recently, some reviews of RA have been published [2,3,4,5,6,7,8,9]. However, these articles have mainly focused on pharmacological studies such as its neuroprotective, anti-diabetic, anticancer, and anti-inflammatory potential, which seemed to be a little simplex. Thus, there is still a lack of a comprehensive review to provide a full-scale understanding of this polyphenol acid. In the present study, we used some mainstream bibliographic databases and search engines such as the Web of Science, PubMed, Chinese National Knowledge Infrastructure (CNKI), and Google Scholar to collect a large number of the research literature and to sum up the interesting progress. Except for “rosmarinic acid” as the keyword, some other characteristic words were also employed including ”isolated” for phytochemistry, “positive drug” and “model” for pharmacology, and “pharmacokinetic” and “LC-MS” for pharmacokinetics. Aside from a summary, we also explored some oof the interesting and attractive research issues, which are proposed here and are believed to be the potential hotspots in the future.

2. Sources and Biosynthesis in the Plants

To our knowledge, RA has been found and isolated as a monomeric component from a total of 162 plants, which are listed in Table 1. It is obvious that Lamiaceae is the largest family, containing 104 plants among them. As far as the genus containing RA is concerned, Salvia is the largest, with 20 plants including S. absconditiflora Greuter & Burdet, S. deserta Schang, S. grandifolia, S. miltiorrhiza Bunge, S. plebeia and S. przewalskii Maxim, etc. With respect to chemotaxonomy, the existence of RA could provide some taxonomic basis at the level of the subfamily. According to the database of the European and Mediterranean Plant Protection Organization, among the 104 plants of Lamiaceae, 93 species come from Nepetoideae and 10 species come from Lamioideae [10]. It is obvious that RA is a characteristic natural product distinguishing Nepetoideae and other subfamilies in Lamiaceae. However, to carry out a taxonomic study in Lamiaceae more accurately, it is impossible to depend solely on RA. Characteristic terpenoids should play the same important roles.
Many of these 154 plants have been used as the sources of traditional Chinese medicinal materials for a long time such as Perilla frutescens (L.) Britt, Prunella vulgaris L., Salvia miltiorrhiza Bunge, Sarcandra glabra (Thunb.) Nakai, Schizonepeta tenuifolia Briquet, etc. Some others also serve as folk medicinal plants in many countries and regions such as Cordia bicolor, Cordia dentate, Cordia megalantha, Hyptis atrorubens Poit., and Hyptis verticillata Jacq. in Central America and the Caribbean; Micromeria myrtifolia Boiss. & Hohen, Salvia palaestina Bentham and Sanicula europaea L. in Turkey; Baccharis chilco, Hyptis capitata Jacq., and Lepechinia meyenii (Walp.) Epling in South America; Ipomoea turpethum (L.) R.Br., Thunbergia laurifolia Lindl, and Thymus serpyllum in South Asia, etc.
It has been shown that in the plants, two amino acids are separately involved in the RA biosynthesis pathways (Figure 2). In the first pathway, L-phenylalanine is orderly transformed to cinnamic acid, 4-coumaric acid, and 4-coumaroyl-CoA by phenylalanine ammonia-lyase, cinnamate 4-hydroxylase, and 4-hydroxycinnamate-CoA ligase, respectively. In the second pathway, L-tyrosine, the precursor, is first transformed to 4-hydroxyphenylpyruvic acid by tyrosine aminotransferase, and then to 4-hydroxyphenyllactic acid by the hydroxyphenylpyruvate reductase. The products of the two biosynthesis ways, both 4-coumaroyl-CoA and 4-hydroxyphenyllactic acid, could be finally converted into RA by the rosmarinic acid synthase and cytochrome P450 monooxygenase associated with the cytochrome P450 reductase [181,182,183,184]. Therefore, it can be easily concluded that rosmarinic acid synthase is a key control point for both of the above two synthesis pathways. As a member of the BAHD acyltransferase family, it was acidic stable and its molecular mass was tested between 36 kD and 59 kD. It was also characterized with the random curl and α-helix, containing neither signal peptides nor leading peptides in the secondary structure [185,186].
Except for the medicinal parts of the herbal plants as natural RA isolation sources, some non-medicinal parts also serve, where Salvia miltiorrhiza aerial parts are a good example. When the roots and rhizomes are harvested, a mass of aerial parts will be thrown and wasted. It was reported that the RA content could reach above 20 mg/g in the aerial parts [187] and Shi et al. successfully isolated it from Salvia miltiorrhiza leaves [144]. Foeniculum Vulgare Mill. is an aromatic plant and is often extracted by distillation for its volatile oil. However, the resultant residue is considered as a waste, in which many antioxidant components exist. Parejo et al. reported the isolation of RA with seven other phytochemicals in 2004 [58]. Regarding the beach waste, Zostera noltii and Zostera marina have also been used as the sources of RA isolation [179,180]. In recent years, due to the increasing price of herbal plants, it is of great interest to look for new sources for the isolation of natural products such as RA. Non-medicinal parts and other biowaste, considered as useless and burdensome in the past, have now attracted more and more research. Therefore, the isolation of RA from these new sources would achieve the aims of saving resources, protecting the environment, realizing the efficient use and recycling of resources, and promoting the development of the industrial economy.

3. Extraction from Plants

As a naturally-occurring polyphenolic acid, RA has often been obtained from plants by different extraction methods including vibration [188], maceration with continuous stirring [189], heat reflux [189], and Soxhlet solvent extraction [190]. In these traditional extraction methods, the solvent is often a key factor responsible for the RA yield. In a study that extracted RA from Dracocephalum moldavica L. aerial parts, n-butanol was investigated as the best solvent when the Soxhlet solvent extraction method was used. Compared to the extraction efficiency of n-butanol (114.54 ± 24.70 mg/g), those of other solvents were 78.43%, 8.96%, 20.84%, and 8.26% for methanol (89.83 ± 1.38 mg/g), ethyl acetate (10.26 ± 1.29 mg/g), acetonitrile (23.87 ± 0.50 mg/g), and water (9.46 ± 0.07 mg/g), respectively [190].
During the past decades, due to their simpler operation, lower time consumption, and simultaneous preparation of more samples, some novel extraction methods have been utilized for RA extraction such as ultrasound-assisted extraction [191], microwave-assisted extraction [192], enzyme-assisted extraction [193], and pressurized-liquid extraction [194]. In a comparison study of the extraction methods used for the leaves of six plants, to obtain the highest extraction efficiency of RA, the optimal extraction parameters of different methods were as follows: 120 min at 25 °C for maceration with stirring extraction (MACs), 15 min at boiling point for heat reflux extraction (HRE), and 5 min at 50 °C and 80 °C for microwave-assisted extraction (MAE). A mixed solvent (CH3CH2OH–H2O–HCl, 70:29:1, v/v/v) was also proven to be the best for each method. In light of the RA yield, MACs was the most appropriate for Melissa officinalis L. (30.0 ± 0.2 mg/g), Mentha piperita (16.2 ± 0.6 mg/g), Rosmarinus officinalis L. (9.2 ± 0.2 mg/g), and Salvia officinalis L. (19.6 ± 0.3 mg/g) while HRE was the best for Thymus vulgaris L. (15.3 ± 1.2 mg/g) and Origanum vulgare L. (40.1 ± 1.0 mg/g) [195]. In addition, characterized with lower melting points, lower cost, lower vapor pressure, and reproducibility, ionic liquid has become an efficient and environmentally-friendly extraction solvent alternative to the conventional ones. In an ultrasound-assisted extraction study of RA from Rosmarinus officinalis leaves, 1-octyl-3-methylimidazolium bromide ([C8mim]Br) was selected as the solvent due to its high extraction efficiency. After the optimization of the extraction factors with response surface methodology, the optimal conditions included 2 h for the soaking time, 30 min for the ultrasound time, 220 W for the ultrasound power, and 1:20 for the solid–liquid ratio, under which the extraction efficiency of RA could reach to 98.91% [196]. In another study of microwave-assisted extraction for RA from Rosmarinus officinalis leaves, [C8mim]Br was also used as the solvent with 700 W for the irradiation power, 15 min for the irradiation time, and 1:12 for the solid–liquid ratio. This method exhibited a considerable RA yield (3.97 mg/g) [197].

4. Natural Derivatives in Plants

From a variety of natural plants, a large number of RA derivatives have been found and isolated, which often simultaneously exist in the same plant with RA in most cases. Supplementary Figure S1 demonstrates their chemical structures.
Among these derivatives, the alkyl rosmarinates are the simplest ones in terms of their chemical structures. Due to the C8′-carboxyl group, RA can combine with some alcohol compounds to obtain some esters in the plants such as rosmarinic acid methyl ester, ethyl ester, n-propyl ester, and n-butyl ester. These alkyl rosmarinates have demonstrated anti-oxidative, anti-inflammatory, anti-allergic, anti-bacterial, anti-cardiovascular disease, and other activities [198,199,200,201,202,203]. In addition, 3-O-methyl rosmarinic acid, 3-O-caffeoyl rosmarinic acid, 3′-O-methyl rosmarinic acid, 4′-O-methyl rosmarinic acid (shimobashiric acid B), and 3, 3′-O-diethyl rosmarinic acid are the natural products of RA substituted by a methyl, ethyl, and even caffeoyl groups on the C3-, C3′-, and C4′-hydroxyl groups. As a polyphenolic acid, RA also has some bioactive glycoside derivatives including rosmarinic acid-3-O-glucoside (salviaflaside), rosmarinic acid-3′-O-glucoside, rosmarinic acid-4-O-glucoside, rosmarinic acid-4′-O-glucoside, and rosmarinic acid-4,4′-O-diglucoside. For example, rosmarinic acid-4-O-glucoside has been studied with a pleiotropic effect against viral pneumonia: (1) To reduce the levels of inflammatory cytokine and oxidative stress in the serum and lungs of A/FM/1/47 H1N1 virus infected mice; and (2) to lower the tissue fluid into the alveoli and inhibit virus proliferation, improve ventilation, and reduce mortality [204].
Aside from these OH-substituted derivatives, there are a series of depside derivatives known as salvianolic acids. Salvianolic acid B is the most famous and representative one, which is listed as one of the chemical markers for the quality evaluation of Salvia miltiorrhiza Bge. roots and rhizomes in both the Chinese Pharmacopoeia and United States Pharmacopoeia. This phytochemical has revealed multiple bioactivities including (1) a protective effect on the brain from ischemia/reperfusion-induced injury by inhibiting reactive oxygen species (ROS)-mediated inflammation [205]; (2) a protective effect on the liver from acute and chronic injury by the inhibition of Smad2C/L phosphorylation [206]; (3) an anti-inflammatory effect on atherosclerosis through the mitogen-activated protein kinase/nuclear factor-κB (MAPKs/NF-κB) signaling pathways in vivo and in vitro [207]; (4) an anti-tumor effect against human breast cancer adenocarcinoma cells [208]; and (5) anti-diabetic effects [209].
All of these mentioned components are considered as the derivatives of biosynthesis from RA. Compared to conventional chemical extraction, inducers can be used to induce plant cells to synthesize valuable secondary metabolites, which is more economical and feasible and less likely to cause pollution. Therefore, it is urgent to explore the possible derivatization patterns and to elucidate the regulatory mechanism of secondary metabolism in these plants.

5. Analytical Technique

Characterized with the higher separation efficiency, less time and sample consumption, and a wider application range, high performance liquid chromatography (HPLC) or ultra performance liquid chromatography (UPLC) has gradually become the mainstream analytical technique in the research field of herbal plants, a complicated matrix with a variety of natural products. Due to its great conjugation system, RA has a strong absorbance in the ultraviolet region. Therefore, for the majority of research papers on the quantitative analysis of RA, an ultraviolet detector or diode-array detector was the mostly used [194,210,211,212]. Moreover, HPLC coupled with evaporative light scattering detector (ELSD) has also been applied for the quantitation of RA in Rosmarinus officinalis L. leaves [213]. However, in the biological samples, there are many endogenous interfering substances present and the content of the analyte is much lower. As a result, in the pharmacokinetic studies of RA concerning plasma, serum, or different tissues, a mass spectrometry detector with multiple-reaction monitoring mode has often been utilized [214,215,216,217].
Capillary electrophoresis (CE) is another widely-used and effective separation technique for the analysis of natural products. Many subtypes are inclusive in CE. However, capillary zone electrophoresis (CZE) and micellar electrokinetic chromatography (MEKC) are the main two used for RA quantitation. In the CZE experiments, a sodium borate solution was used as the run buffer to determine the RA in Salvia officinalis tea samples [218], in 14 Salvia species [219], in Origanum Vulgare L. [220], and in Melissa officinalis products [221]. In the MEKC studies, to obtain a satisfactory separation of RA from the other components, some additives were supplemented to the buffer such as β-cyclodextrin [222,223] and sodium dodecylsulfate [224].
However, it is well-known that some physicochemical pretreatments are necessary when the aforementioned LC or CE method is employed. In recent years, nondestructive determination methods have caused wide concern, of which some techniques related to infrared are the ones most representative. There have been some successful examples of the quantitative analysis of RA in Rosmarinus officinalis L. leaves [225], Thymus vulgaris L. or Thymus zygis L. leaves and flowers [226], and several Lamiaceae plants [227]. In these studies, the conventional HPLC method has also been used to compare the results along with partial least squares regression analysis, a chemometric model for calibration and validation.

6. Pharmacology

RA, a natural product from many plants, has been studied to possess a wide range of similar pharmacological activities with its origins such as anti-inflammation, anti-oxidation, anti-diabetes, anti-tumor, anti-virus, neuroprotection, hepatoprotection, and others in many in vivo and in vitro studies.

6.1. Anti-Inflammation

Inflammatory diseases are the pathological processes of defense responses evoked by some stimulation such as infection and trauma and are characterized by the imbalance in inflammatory mediators and cells. Inflammation also has a significant impact on human health and is involved in many other diseases. In recent decades, phytochemicals have attracted more and more attention regarding treatment.
In osteoarthritis, the degradation of cartilage extracellular matrix (ECM) might be induced by the depletion of collagen 2 and aggrecan, two of its main components. In addition, a disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4) and ADAMTS-5 are involved in this degradation. In an in vitro study of IL-1β-induced chondrocytes, the gene expression of collagen 2 and aggrecan were inhibited and ECM degradation occurred. RA incubation of 100 μM was observed to abolish this inhibition and demonstrate the inhibitory effect on IL-6 production, the gene and protein expression of ADAMTS-4 and ADAMTS-5, and even on the ECM degradation. The outcome led to the conclusion that RA may be a promising drug for osteoarthritis treatment [228]. In another in vivo study of the mice arthritis model induced by collagen, intraperitoneal injection of RA (50 mg/kg) markedly improved the arthritis index and reduced the affected paw number. Compared to those in the control group, severe leukocyte infiltration, the architecture of synovial tissues, and bone integrity loss were also more normal in the RA treatment group, manifesting a lower histopathologic index [229].
It is common knowledge that T cells are involved in atopic dermatitis (AD) pathogenesis. In the acute stage, AD skin lesions are infiltrated by CD4+ T cells, which could secrete IL-4, IL-5, and IL-13. In the chronic stage, Th1 cells secrete interferon-γ (IFN-γ). Some researchers have reported that RA (5 μM) could significantly inhibit the production of IL-4 and IFN-γ through activated CD4+ T cells. In addition, the same researchers also found after 2,4-dinitrofluorobenzene challenge, the symptoms of AD-like skin lesions were found on the NC/Nga mice such as pruritus, eruptions, and ear swelling. In this pathological state, the serum IgE level was tested as abnormally high and the characteristic dermal infiltration of inflammatory cells including CD4+ T, CD8+ T, and mast cells into ear skin lesions was observed to be markedly increased. Intraperitoneal administration of RA (50 mg/kg) also exhibited remarkable ameliorating and inhibiting effects on the above pathological phenomenon [230].
Inflammatory bowel disease is a chronic and recurrent intestinal inflammation in which ulcerative colitis is a typical one. In mice with colitis induced by dextran sulfate sodium, the oral administration of RA (60 mg/kg) significantly reduced the severity of colitis as shown by the disease activity index scores, colonic damage, and colon length. Furthermore, RA treatment also led to the decrease in some of the proinflammatory cytokines including IL-6, IL-1β, and IL-22, and the protein levels of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in the colons. These protective effects were proven to be related to the inhibition of NF-κB and signal transducer and activator of transcription 3 (STAT3) activation [231]. In another study, RA was believed to protect from ulcerative colitis by regulating macrophage polarization depending on heme oxygenase-1 [232].
Aside from the above-mentioned studies, RA has been studied in vitro or in vivo to exert protective or ameliorative properties on lipopolysaccharide (LPS)-induced mastitis [233], sodium taurocholate-stimulated acute pancreatitis [234], LPS-induced acute lung injury [235], LPS-induced neuroinflammation [236], plaque-induced gingivitis [237], concanavalin A-induced hepatic injury [238], ovalbumin-stimulated allergic rhinitis [239], etc.

6.2. Anti-Oxidation

Combined challenge of ovalbumin and hydrogen peroxide would lead to a superimposed asthma with oxidative lung damage symptoms in mice. In the BALF and lung tissues of the model group, inflammatory cells including eosinophils, neutrophils, and lymphocytes and cytokines IL-4, IL-5, IL-13, and IFN-γ were all found to be elevated; ROS, nicotinamide adenine dinucleotide phosphate oxidase-2 (NOX-2), and NOX-4 levels were remarkably upregulated; and the total superoxide dismutase (SOD), total glutathione peroxidase (GSH-Px), catalase (CAT), and Cu/Zn SOD activities were observably downregulated compared to those in the blank group. RA treatment (10, 20, 40 mg/kg) exhibited alleviative and protective effects on the above symptoms and the highest dose was even more effective than dexamethasone [240]. In terms of antioxidant property in Caenorhabditis elegans, RA (60, 120, 180 μM) could significantly enhance the catalase activity, GSH-Px activity, and reduce glutathione (GSH) content and the glutathione/oxidized glutathione ratio as well as diminish the malondialdehyde (MDA) content in a dose-dependent manner, which resulted in promoting the lifespan and motoricity and reducing the fat store without threatening fertility. Furthermore, after RA treatment, the survival rate under acute oxidative and thermal stress was increased while intestinal lipofuscin accumulation was suppressed. This strong antioxidant activity was deemed to be related to regulating the insulin/insulin-like growth factor signaling (IIS) and MAPK pathways and activating the downstream antioxidant enzyme gene expression in Caenorhabditis elegans [241]. Chromium is known to cause severe toxicity in the liver and kidney tissue. In a potassium dichromate challenged rat model, RA (25 mg/kg) oral gavage of 60 days was observed to show s protective effect and reduce the oxidative damage in the two tissues. Oxidative stress evaluation demonstrated a remarkable increase in the GSH level and a notable decrease in the MDA level in the RA treatment group compared to those in the model group. Immunohistochemical studies and Rt-PCR analysis have confirmed that the result might be obtained via the Nrf2 pathway [242]. By activating the same nuclear factor erythroid-2 related factor 2 (Nrf2) pathway and increasing the downstream antioxidant enzyme activity, the oral administration of RA at 2 mg/kg could protect mouse intestines against high-fat diet-stimulated oxidative stress by preventing intestinal epithelial cell apoptosis [243].

6.3. Anti-Diabetes

Some in vitro studies have exhibited the anti-diabetic activity of RA. The polyphenolic acid was shown to have an inhibitory effect on α-glucosidase with an IC50 value of 33.0 μM, much lower than that of acarbose (131.2 μM), a marketed α-glucosidase inhibitory drug [123]. RA was also demonstrated to have a regulatory effect on glucose homeostasis. It was found that RA (5.0 µM) could activate adenosine 5′-monophosphate-activated protein kinase (AMPK) phosphorylation and increase the glucose uptake in L6 rat muscle cells, comparable to the maximum insulin (0.1 µM) and metformin (2.0 mM) [244].
In a dose-dependent manner, RA treatment (120–200 mg/kg, 7 days) showed a remarkable hypoglycemic effect in streptozotocin-induced type-1-like diabetic rats and significantly improved the glucose uptake and insulin sensitivity in high-fat diet-induced type-2-like diabetic rats. This beneficial effect against diabetes was believed to be related to both the decrease in phosphoenolpyruvate carboxykinase expression in the liver and the increase in glucose transporter-4 expression in the skeletal muscle [245]. In another study, the RA treatment (100 mg/kg, 30 days) of diabetic rats was found to have the effect of restoring the blood glucose level and regulating the levels of adiponectin and leptin. In addition, the diabetic pathology in hepatic parenchymal structures was also attenuated by the introduction of RA through histological and ultrastructural observations [246]. Na+/glucose cotransporter-1 (SGLT1) is considered as an important glucose transporter from intestinal lumen to blood. RA administration (14 days) could reverse the streptozotocin-induced SGLT1 protein increase and stabilize the fasting blood glucose level in diabetic rats [247].

6.4. Anti-Tumor

Breast cancer stem-like cells play an important role in the initiation, maintenance, and metastasis of breast cancer. RA coincubation (270 μM, 810 μM) could decrease their viability, inhibit their migration, and induce their apoptosis. RT-PCR analysis and immunoblot analysis showed that the two concentrations of RA treatment notably lowered the levels of mRNA and the protein of phosphorylation of smoothened and Glioma-associated oncogene homolog 1. Furthermore, RA treatment also led to the downregulation of B-cell lymphoma-2 (Bcl-2) expression and the upregulation of Bax expression. Therefore, the anti-tumor effect of RA might be related to the Bcl-2 and hedgehog signaling pathways [248]. Cao et al. employed intragastric administration of RA (150, 300 mg/kg) for 10 days to treat H22 tumor-bearing mice. It was unveiled that RA could effectively inhibit the tumor growth by regulating the secretion of inflammation and angiogenesis cytokines (IL-1β, IL-6, tumor necrosis factor-α (TNF-α), vascular endothelial growth factor, and transforming growth factor-β) and suppressing NF-κB p65 expression in the microenvironment [249].
Regarding the 1,2-dimethylhydrazine (DMH)-induced colon carcinogenesis in rats, many pathological phenomena have been easily found and tested in the liver and colon such as a large number of colonic tumors, decreased lipid peroxidation, antioxidant status and glutathione-S-transferase activity, md elevated cytochrome P450 content and p-nitrophenol hydroxylase activity, which were significantly reversed by RA (5 mg/kg). The pronounced effects indicated the possibility of RA as a chemopreventive agent against colon cancer [250]. In another DMH-stimulated rat model with colon carcinogenesis, oral supplementation with RA (5 mg/kg) also demonstrated a pronounced anti-tumor activity, probably due to the reduction in aberrant crypt foci formation and multiplicity, the suppression of fecal and colonic mucosal bacterial enzyme activities, and the improvement in circulatory thiobarbituric acid reactive substances (TBARS), enzymic, and non-enzymic antioxidant status [251].
In addition, RA also showed an anti-tumor effect on 7,12-dimethylbenz(a)anthracene-induced skin carcinogenesis [252], a cytotoxic effect on ARH-77 human (multiple myeloma) cells [253], prostate cancer cells [254], and human Hep-G2 liver carcinoma cells [255], and an inhibitory effect on the metastatic properties of colorectal cancer cells [256].

6.5. Anti-Virus

Enterovirus 71 (EV71) is a nonenveloped single-stranded RNA virus and easily causes hand, foot, and mouth disease, and even neurological complications or fatality in children. However, there is no specific and pointed treatment. Recently, phytomedicines and phytochemicals have been the alternative to chemical drugs for anti-virus. The Melissa officinalis extract was investigated to possess anti-EV71 activity and RA was identified and proven to be the responsible bioactive component therein, in which the alleviations of p38 kinase and epidermal growth factor receptor substrate 15 hyperphosphorylation were deduced to be involved [257]. In EV71-infected human rhabdomyosarcoma cells, RA was tested with a low IC50 value (4.33 μM) and a high therapeutic index (340) when the infection multiplicity was 1. Further investigation showed that RA could protect the cells from the cytopathic effects and apoptosis at the early stage of viral infection. In EV71-challenged neonatal mice, RA (20 mg/kg) also manifested the similar protective effect at the early stage, prolonging survival time and reducing mortality [258]. The results of another study were consistent with these findings and revealed the possible mechanism associated with virus-P-selectin glycoprotein ligand-1 and virus-heparan sulfate interactions [259]. The above findings indicated RA as a potential EV71 inhibitor in the initial stages of viral infection.
Japanese encephalitis virus (JEV) is a crucial cause of acute encephalopathy in children, targeting the central nervous system. With intraperitoneal treatment of RA (25 mg/kg), the significant reduction in the mortality of JEV-infected mice was observed, along with the dramatic decreases in viral loads and proinflammatory cytokines including IL-12, TNF-α, IFN-γ, monocyte chemotactic protein 1 (MCP-1), and IL-6. These findings suggest the potential of RA as a candidate for JEV treatment [260]. In primary human hepatocytes infected by the hepatitis B virus (HBV), RA exhibited an inhibitory effect on HBV replication and a potentiation effect on the anti-HBV activity of lamivudine [261]. Furthermore, like oseltamivir, RA (IC50 = 0.40 μM) showed high neuraminidase (NA)-inhibiting activity from an in vitro study of the anti-influenza virus, which was confirmed by the high binding affinity, hot-spot residues, and II-bond formations of the RA/NA complex from the in silico study [262].

6.6. Neuroprotection

6-Hydroxydopamine (6-OHDA) is known to be a neurotoxin used to create similar symptoms as Parkinson’s disease (PD). In MES23.5 dopaminergic cells co-incubated with 6-OHDA, RA (0.1 mM) could protect them from induced neurotoxicity through preventing the viability reduction and upregulating the ROS generation and mitochondria membrane potential [263]. In an in vivo study on the 6-OHDA-induced rat model, RA (20 mg/kg) through intragastric administration showed a neuroreparative function on the degeneration of the nigrostriatal dopaminergic system by decreasing the nigral iron level and regulating the Bcl-2/Bax gene expression [264]. Therefore, regarding PD, RA might be viewed as a therapeutic treatment for related patients in the future.
Aβ42 was used to induce an Alzheimer’s disease-like rat model, resulting in a significant increase in the levels of TBARS and 4-hydroxy-2-nonenal and decrease in the SOD, CAT, GSH-Px, and glutathione levels with the reduction in acetylcholine content and acetylcholine esterase activity. In addition, mismatch negativity response and θ power/coherence of auditory event related potentials were also decreased. Fortunately, RA (50 mg/kg, oral administration) demonstrated an attenuating effect on these observed pathological changes and the increased Aβ staining and astrocyte activation [265]. In a kainate-induced rat model, seizure intensity, apoptosis, oxidative stress markers (MDA, GSH, CAT), Timm index, and the number of Nissl-stained neurons were employed as the indicators to evaluate the beneficial effect of RA. The results supported the neuroprotective effect of RA (10 mg/kg) against temporal lobe epilepsy [266]. Intraperitoneal administration of RA (20 mg/kg) was observed to improve the working, spatial, and recognition memory deficits, and to reduce the infarct size and neurological deficits of the rats lesioned by permanent middle cerebral artery occlusion, which were speculated to be related to suppressing neuronal loss and increasing synaptophysin expression and brain-derived neurotrophic factor. Therefore, the results indicate the memory protective effect of RA [267]. As for spinal cord injury, RA was investigated to show a neuroprotective effect on this severe central nervous system injury through inhibiting the TLR4/NF-κB pathway and activating the Nrf2/HO-1 pathway, as witnessed by in vitro (55.6 μM) and in vivo (40 mg/kg, intraperitoneal administration) studies [268].

6.7. Hepatoprotection

Li et al. conducted in vitro and in vivo studies to observe the hepatoprotective effect of RA against experimental liver fibrosis. In hepatic stellate cells, RA co-incubation (32 μM) was found to inhibit cell proliferation and the expressions of transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF), and α-smooth muscle actin. In CCl4-intoxicated rats with liver fibrosis, RA (10 mg/kg) could reduce the fibrosis grade, ameliorate biochemical indicators (albumin, globulin, alanine aminotransferase, glutamate-pyruvate transaminase) and histopathological morphology, and downregulate the liver TGF-β1 and CTGF expression [269]. The findings were then witnessed and confirmed by Domitrovic and his colleagues in a mice model with CCl4-intoxicated liver fibrosis. In addition to improvements in the histological and serum markers concerning liver damage and the inhibition of TGF-β1 and CTGF expression, the amelioration of oxidative/nitrosative stress and inflammatory response (NF-κB, TNF-α, COX-2) and the upregulation of Nrf2 and heme oxygenase-1 expression were also found after RA treatment (50 mg/kg) [270].
In the bile duct ligation-induced extrahepatic cholestasis rat model, RA (20 mg/kg) exhibited a hepatoprotective effect by alleviating TGF-β1 production and hepatic collagen deposition and ameliorating hepatic inflammation. Resolution of oxidative burden and downregulation of high mobility group box-1/toll-like receptor-4 (HMGB1/TLR4), NF-κB, AP-1, and TGF-β1/Smad signaling were investigated to be involved in RA hepatoprotection [271]. Furthermore, Lou et al. used the partial hepatectomy model to explore the effects of RA on liver regeneration. The evaluation content included the index of the liver to body weight and the expression of proliferating cell nuclear antigen and liver transaminases. As a result, RA (200 mg/kg) could promote liver regeneration and restore lesioned liver function via the mammalian target of rapamycin/S6 protein kinase (mTOR/S6K) pathway [272]. Furthermore, in a mouse model of non-alcohol steatohepatitis induced by a methionine-choline-deficient diet, RA (10 mg/kg) exhibited a remarkable hepatoprotective potential by decreasing the plasma triglyceride, cholesterol, liver steatosis, and oxidative stress, which was deemed to be related to the activation of the silent information regulator-two 1 (SIRT1)/Nrf2, SIRT1/NF-κB, and SIRT1/peroxisome proliferator-activated receptor α (PPARα) pathways [273].

6.8. Other Activities

To study RA protection against premature ovarian failure (POF), the intraperitoneal injection of cyclophosphamide was used to induce the mouse model. With the help of fluorescence immunohistochemistry, histological analysis, Western blot analysis and polymerase chain reaction, RA (40 mg/kg) was investigated to effectively attenuate the abnormal situations of the model including injured ovarian, increased ovarian index, and serum sex hormone levels, the overexpression of the nucleotide-binding oligomerization domain receptor protein-3 (NLRP3) inflammasome, and apoptosis-related proteins in the ovarian. The findings indicate that RA might have a bright prospect in POF treatment in the future [274].
In the treatment of thoracic tumor, radiotherapy is an essential therapy method, which will unfortunately cause pulmonary fibrosis later. Zhang et al. observed that RA (120 mg/kg) could regulate NF-κB signaling and the RhoA/Rock pathway through microRNA-19b-3p, which were responsible for the alleviation of inflammatory reactions, the reduction in collagen hyperplasia, and the suppression of pulmonary fibrosis development in the X-ray irradiation-induced rat model. Thus, RA is believed to be a potential alternative to attenuating radiotherapy-caused pulmonary fibrosis [275].
Ji et al. established aa high-fat diet and VD3-induced rat model to observe the effect of RA on vascular calcification. The results showed that RA (200 mg/kg) could notably decrease the levels of alkaline phosphatase, phosphorus, calcium, MDA, increase the SOD level, and reduce the calcified nodule content and ROS production. Additionally, the levels of Nrf2, heme oxygenase-1, NAD(P)H quinone dehydrogenase, and osteoprotegerin were upregulated, while the levels of kelch-like ECH-associated protein 1, NF-κB, β-catenin, and osteogenic transcription factor were significantly downregulated. RA coincubation (80 μM) also showed similar effects in the β-glyerophosphate-induced rat aortic smooth muscle cell model. These functions in improvement were proven to be related to the regulation of the Nrf2 pathway [276].

7. Clinical Studies

Although RA has been proven to have potential for drug application in many research articles, only two papers have been published on the clinical study of RA as a pure compound.
It was reported that there were 14 female and seven male patients with mild AD inclusive in a clinical study, in which a RA (0.3%) emulsion was applied to the elbow flexures twice a day. Compared to before the treatment, erythema and transepidermal water loss of the antecubital fossa were reduced notably after treatment of four or eight weeks. Self-reports from the patients showed that dryness, pruritus, and general AD symptoms were ameliorated after RA smearing [277]. Another clinical study enrolled 29 patients with seasonal allergic rhinoconjunctivitis. The results indicated that RA oral treatment (80, 200 mg/kg) led to significant decreases in the incidence rates for itchy nose, watery eyes, itchy eyes, and total symptoms compared to the placebo. Meanwhile, the number of neutrophils and eosinophils in the nasal lavage fluid were also significantly decreased [278].

8. Applications in Food Science

It is well-known that the polyphenol natural products are characterized by their multiple phenolic hydroxyl groups and accompanying anti-oxidation. As an organic acid with four phenolic hydroxyl groups, RA has exhibited its anti-oxidant capacity not only in pharmacological studies, but also in food scientific studies.
In sea buckthorn fruit wine, concerning DPPH radical scavenging and hydroxide radical scavenging, RA showed a greater antioxidant capacity (IC50 = 8.02 mg/mL, 99.31% clearance rate) than sulfur dioxide (IC50 = 10.31 mg/mL, 98.67% clearance rate), the conventional antioxidant. Therefore, RA was speculated to be an ideal antioxidant alternative to sulfur dioxide in wine fermentation due to its safety and stability [279]. Li et al. prepared different rabbit skin gelatin–RA composite films to study their preservation effects on the pork quality during cold storage. As a result, a composite film with RA of 0.8 g/L could effectively inhibit the increase in the total number of colonies, total volatile basic nitrogen content and pH, extend the shelf life of pork from 4 to 8 days, remain at a high hardness, and reasonable chromatic aberration. Therefore, the rabbit skin gelatin–RA composite film was proposed to be a potential packaging material for the preservation and freshness of pork [280]. A total of 1% chitosan containing 30 mg/L RA was studied with total viable counts (less than 6.0 log CFU/g), potassium value (less than 60%), free fatty acids (2.5%), trimethylamine (2 mg/100 g), and H2S-producing bacteria (less than 6.0 log CFU/g) and to maintain better sensory characteristics and flavor quality of the half-smooth tongue sole fillets stored at 4 °C for 18 days. On the other hand, the results for the control group were 7.5 log CFU/g, nearly 90%, 5.0%, and 3.7 mg/100 g more than 6.0 log CFU/g, respectively. Therefore, these significant differences indicate the complex potential to improve the quality of this fish during refrigerated storage [281].
Therefore, as a polyphenolic acid with antioxidant and antibacterial activity, RA could retard the growth of microorganisms and inhibit the increase in the pH value and perioxidation in food, contributing to its capacity of keeping food quality, slowing decay, and extending the shelf life.

9. Pharmacokinetics

It is well-known that the pharmacokinetic profiles are fundamental for a potential candidate drug. Regarding RA with the pronounced bioactivities, it is exactly that. A study of this polyphenolic acid was carried out on the metabolites and the pharmacokinetic pathways in normal rats. As a result, a total of 36 metabolites including RA itself were identified in plasma, urine, and feces after oral administration. The prototype and glucuronic acid conjugation were found to be predominant in plasma. Furthermore, Phase I metabolism (primarily hydrolysis) and Phase II metabolism (sulfation, methylation, glucuronic acid conjugation, and glucose conjugation) were mainly involved in the feces and urine, respectively [282]. In another study associated with human liver microsomes, after 1 h of incubation, RA was transformed to yield 14 metabolites and several metabolic pathways were speculated including oxidation, glucuronic acid conjugation, hydroxylation, and GSH conjugation [283].
To reveal the oral absolute bioavailability of RA, the normal rats were administered with the phytochemical through intragastrical (12.5, 25, 50 mg/kg) and intravenous (0.625 mg/kg) methods. The calculated parameters showed rapid absorption and middle-speed elimination for the pharmacokinetic characters of RA after oral administration in rats. In addition, poor absolute bioavailability was demonstrated with 1.69%, 1.28%, and 0.91% for 12.5 mg/kg, 25 mg/kg, and 50 mg/kg, respectively [284]. In a hepatoprotective and metabolic study, RA treatment could significantly suppress the pathological changes in the bile rate, thiobarbituric acid (TBA), total bilirubin (TBIL), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) of rats with cholestatic liver injury. On the other hand, cholestasis resulted in PK behavior variations and the drug accumulation of RA, which were witnessed by the decrease of 14.5% for CL and the increase of 17.0% for AUC(0∞), 40.3% for Tmax, and 13.1% for Cmax, compared to those of normal rats [285]. As above-mentioned, RA often serves as an indicator for the quality evaluation of some folk herbal medicines or compound medicines. In the same way, RA also acts as a representative component in the pharmacokinetic studies of these medicines such as the Salvia miltiorrhiza polyphenolic acid solution [286], Prunella vulgaris extract [287], ZibuPiyin Recipe [288], and Xuebijing Injection [289]. Table 2 presents the specific parameters of these pharmacokinetic studies including the AUC(0∞), Tmax, Cmax, and CL.
As a potential candidate drug with various effects, RA should first be based on its toxicity. However, there have only been several in vitro studies mentioning its non-cytotoxicity at the test concentrations in normal cells such as chondrocytes (100 μM) [228], HepG2 cells (100 μM) [290], N2A mouse neuroblastoma cells (250 μM) [291], and A172 human astrocytes (83 μM) [292], with the median lethal concentration in zebrafish embryos of 296.0 μM [236]. In the two clinical studies, it was reported that there were no self-feeling adverse events and no significant abnormalities in routine blood tests [277,278]. Therefore, a systematic toxicity investigation of RA should be conducted urgently in the future including acute toxicity, chronic toxicity, LD50, therapeutic window, etc. Additionally, according to the administration approach of the phytochemical, different types of test animals should be involved, where each important tissue and organ should be observed, and each blood index should be tested. After all, safety is the first key character of a drug, especially prior to its clinical application.

10. Future Perspectives

As above-mentioned, RA is believed to be a polyphenolic acid that widely occurs in natural plants, especially from Lamiaceae. During over the past sixty years, RA has exhibited miscellaneous pharmacological activities, pharmacokinetic characteristics, and a variety of natural sources and derivatives.
Generally speaking, the structural modification of a natural product often aims to improve its bioavailability, to extend (improve) its bioactivities, or to diminish its toxicity. Up to now, many RA derivatives have been found in nature and some have revealed impressive biological effects, which could be considered as the products of RA structural modification. However, it was unordered and unscheduled in the way in which they were isolated and found biological. With the further understanding of the RA action mechanism, structural modification with specific purposes should be well-designed and carried out in the future including (1) investigating the RA chemical structure by crystallology and quantum mechanics; (2) simulating the combination of the RA and target from the protein database; (3) summarizing the action rules of the RA derivatives with different substituent groups; (4) systematically proving their bioactivities with high throughput screening.
In terms of pharmacokinetic study of RA, there have been many articles reported including the intragastrical administration of this pure component or some compound formulations, its application on the test animals or humans, and its application on normal or model animals. However, there still exist some issues worth discussing. (1) The rats, especially the normal ones, were used in the majority of pharmacokinetic studies. Now that RA has showed a variety of pharmacological activities, the corresponding model animals should be the first choices. Additionally, rats should not be the only test species. (2) A single dose of RA administration was involved in a large number of pharmacokinetic studies. Since RA will act as a candidate drug and the treatment will last for several days, it seems that a multi-dose of RA administration is necessary and the relevant pharmacokinetic studies are essential. (3) The number of clinical pharmacokinetic studies of RA is small [293,294]. At present, RA is not approved as a legal drug and is prohibited for medical application on humans. However, in traditional medicines, some herbal extracts or compound formulations enriched with RA are allowable. Their pharmacokinetic studies could provide some basis for further drawing of the RA pharmacokinetic profile. (4) Intragastrical administration was the main focus while other administration methods have been rarely investigated and would be interesting to pursue in the future.
Until now, RA has been well-acknowledged as a promising natural product with a variety of pharmacological activities such as anti-oxidation, anti-inflammation, anti-tumor, anti-virus, anti-diabetes, etc. Furthermore, some possible signaling pathways have been explored. However, to be developed as a true candidate drug, RA should be investigated with a focus on some straightforward and effective bioactivity for some diseases including the drug-delivery method, therapeutic dose, and possible action mechanism. On the other hand, based on the clear action targets and the results of the in vitro studies, computer-assisted molecular docking is becoming a virtual screening method for both drugs and their bioactivities. RA has been found to inhibit peptide deformylase, N-myristoyltransferase, human hyaluronidase enzyme, and influenza neuraminidase through in silico evaluations [262,295]. Therefore, in the future, this technology would help us discover more activities and widen the medical application range of RA.
RA is a polyphenolic acid characterized by poor lipid solubility, poor membrane permeability, and low oral absolute bioavailability, which has limited its application. Some liposomes and solid lipid nanoparticles have been revealed to be promising [296,297]. Therefore, aside from structural modification, some systematic studies concerning pharmaceutical formulations or special excipients should be carried out to avoid RA degradation in the gastrointestinal tract and to transport RA to the target tissues. With these achievements, the shortcomings of limited absorption, fast distribution, fast metabolism, and fast elimination might be overcome in the future. Meanwhile, the present research of RA in food science are around anti-oxidation and maintaining the food color and luster. However, it is important to explore the possibility of RA being used as an alternative to the traditional additives. Therefore, the study hotspots should be to compare this phytochemical and the main additives not only affecting the food quality, but also in its safe use.

11. Conclusions

Taken together, all the research findings indicate that RA is a candidate drug or a lead component naturally occurring in plants. In the present paper, we summarized the achievements from phytochemistry, pharmacology, pharmacokinetics, and other study aspects of RA and proposed some interesting issues worth investigating in the future. We hope this paper can help researchers either in fundamental research or in applied research to understand RA more comprehensively, utilize RA more efficiently, and eventually develop RA as a novel drug.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/molecules27103292/s1, Figure S1: Chemical structure of RA derivatives.

Author Contributions

Conceptualization, H.G., L.Z. and Q.W.; Resources, M.S., W.L., B.B., Y.C. and F.C.; Writing—original draft preparation, H.G. and S.Y.; Writing—review and editing, M.S. and H.G.; Funding acquisition, Q.F. and Q.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (Grant Nos. 81473313, 81873229, 81973435).

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Scarpati, M.L.; Oriente, G. Isolamento e costituzione dell’acido rosmarinico (dal rosmarinus off.). Ric. Sci. 1958, 28, 2329–2333. [Google Scholar]
  2. Elufioye, T.O.; Habtemariam, S. Hepatoprotective effects of rosmarinic acid: Insight into its mechanisms of action. Biomed. Pharmacother. 2019, 112, 108600. [Google Scholar] [CrossRef] [PubMed]
  3. Fachel, F.N.S.; Schuh, R.S.; Veras, K.S.; Bassani, V.L.; Koester, L.S.; Henriques, A.T.; Braganhol, E.; Teixeira, H.F. An overview of the neuroprotective potential of rosmarinic acid and its association with nanotechnology-based delivery systems: A novel approach to treating neurodegenerative disorders. Neurochem. Int. 2019, 122, 47–58. [Google Scholar] [CrossRef] [PubMed]
  4. Hitl, M.; Kladar, N.; Gavaric, N.; Bozin, B. Rosmarinic acid-human pharmacokinetics and health benefits. Planta Med. 2021, 87, 273–282. [Google Scholar] [CrossRef] [PubMed]
  5. Luo, C.X.; Zou, L.; Sun, H.J.; Peng, J.Y.; Gao, C.; Bao, L.C.; Ji, R.P.; Jin, Y.; Sun, S.Y. A review of the anti-inflammatory effects of rosmarinic acid on inflammatory diseases. Front. Pharmacol. 2020, 11, 153. [Google Scholar] [CrossRef]
  6. Nadeem, M.; Imran, M.; Gondal, T.A.; Imran, A.; Shahbaz, M.; Amir, R.M.; Sajid, M.W.; Qaisrani, T.B.; Atif, M.; Hussain, G.; et al. Therapeutic potential of rosmarinic acid: A comprehensive review. Appl. Sci. 2019, 9, 3139. [Google Scholar] [CrossRef] [Green Version]
  7. Ngo, Y.L.; Lau, C.H.; Chua, L.S. Review on rosmarinic acid extraction, fractionation and its anti-diabetic potential. Food Chem. Toxicol. 2018, 121, 687–700. [Google Scholar] [CrossRef]
  8. Rahbardar, M.G.; Hosseinzadeh, H. Effects of rosmarinic acid on nervous system disorders: An updated review. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 1779–1795. [Google Scholar] [CrossRef]
  9. Swamy, M.K.; Sinniah, U.R.; Ghasemzadeh, A. Anticancer potential of rosmarinic acid and its improved production through biotechnological interventions and functional genomics. Appl. Microbiol. Biotechnol. 2018, 102, 7775–7793. [Google Scholar] [CrossRef]
  10. European and Mediterranean Plant Protection Organization. Available online: http://gd.eppo.int (accessed on 10 May 2022).
  11. Akhtar, M.S.; Hossain, M.A.; Said, S.A. Isolation and characterization of antimicrobial compound from the stem-bark of the traditionally used medicinal plant Adenium obesum. J. Tradit. Complement. Med. 2016, 7, 296–300. [Google Scholar] [CrossRef] [Green Version]
  12. Tufa, T.; Damianakos, H.; Zengin, G.; Graikou, K.; Chinou, I. Antioxidant and enzyme inhibitory activities of disodium rabdosiin isolated from Alkanna sfikasiana Tan, Vold and Strid. S. Afr. J. Bot. 2019, 120, 157–162. [Google Scholar] [CrossRef]
  13. Kuruuzum-Uz, A.; Suleyman, H.; Cadirci, E.; Guvenalp, Z.; Demirezer, L.O. Investigation on anti-Inflammatory and antiulcer activities of Anchusa azurea extracts and their major constituent rosmarinic acid. Z. Nat. C 2012, 67, 360–366. [Google Scholar]
  14. Li, M.H.; Chang, W.; Lu, W.J.; Ma, G.Z. Chemical constituents of Anchusa italica Retz. and protective effects on hypoxia/reoxygenation induced oxidative stress injury in rat primary cardiomyocyte. Northwest Pharm. J. 2020, 35, 335–340. [Google Scholar]
  15. Braca, A.; Bader, A.; Siciliano, T.; Morelli, L.; De Tommasi, N. New pyrrolizidine alkaloids and glycosides from Anchusa strigosa. Planta Med. 2003, 69, 835–841. [Google Scholar]
  16. Takeda, R.; Hasegawa, J.; Shinozaki, M. The first isolation of lignans, megacerotonic acid and anthocerotonic acid, from non-vascular plants, Anthocerotae (hornworts). Tetrahedron Lett. 1990, 31, 4159–4162. [Google Scholar] [CrossRef]
  17. Lasure, A.; Vanpoel, B.; Pieters, L.; Claeys, M.; Gupta, M.; Vandenberghe, D.; Vlietinck, A.J. Complement-inhibiting properties of Apeiba tibourbou. Planta Med. 1994, 60, 276–277. [Google Scholar] [CrossRef]
  18. Olivier, D.K.; van Wyk, B.E.; van Heerden, F.R. The chemotaxonomic and medicinal significance of phenolic acids in Arctopus and Alepidea (Apiaceae subfamily Saniculoideae). Biochem. Syst. Ecol. 2008, 36, 724–729. [Google Scholar] [CrossRef]
  19. Yuzbasioglu, M.; Kuruuzum-Uz, A.; Guvenalp, Z.; Simon, A.; Toth, G.; Harput, U.S.; Kazaz, C.; Bilgili, B.; Duman, H.; Saracoglu, I.; et al. Cytotoxic compounds from endemic Arnebia purpurea. Nat. Prod. Commun. 2015, 10, 595–596. [Google Scholar] [CrossRef] [Green Version]
  20. Argoti, J.C.; Linares-Palomino, P.J.; Salido, S.; Ramirez, B.; Insuasty, B.; Altarejos, J. On-line activity screening for radical scavengers from Baccharis chilco. Chem. Biodivers. 2013, 10, 189–197. [Google Scholar] [CrossRef]
  21. Badem, M.; Sener, S.O.; Kanbolat, S.; Korkmaz, N.; Yildirmis, S.; Ozgen, U.; Aliyazicioglu, R.; Salva, E.; Kaban, K.; Kandemir, A.; et al. Evaluation of biological activities of Barbarea integrifolia and isolation of a new glucosinolate derivated compound. Z. Nat. C 2021, 76, 375–382. [Google Scholar] [CrossRef]
  22. Scognamiglio, M.; Buommino, E.; Coretti, L.; Graziani, V.; Russo, R.; Caputo, P.; Donnarumma, G.; D’Abrosca, B.; Fiorentino, A. Phytochemical investigation and antimicrobial assessment of Bellis sylvestris leaves. Phytochem. Lett. 2016, 17, 6–13. [Google Scholar] [CrossRef]
  23. Andrade, J.M.D.; Passos, C.D.; Rubio, M.A.K.; Mendonca, J.N.; Lopes, N.P.; Henriques, A.T. Combining in vitro and in silico approaches to evaluate the multifunctional profile of rosmarinic acid from Blechnum brasiliense on targets related to neurodegeneration. Chem. Biol. Interact. 2016, 254, 135–145. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, J.; Wang, Z.W.; Mi, Q. Phenolic compounds from Canna edulis Ker residue and their antioxidant activity. LWT-Food Sci. Technol. 2011, 44, 2091–2096. [Google Scholar] [CrossRef]
  25. Ly, T.N.; Shimoyamada, M.; Yamauchi, R. Isolation and characterization of rosmarinic acid oligomers in Celastrus hindsii Benth leaves and their antioxidative activity. J. Agric. Food Chem. 2006, 54, 3786–3793. [Google Scholar] [CrossRef] [PubMed]
  26. Yoshida, M.; Fuchigami, M.; Nagao, T.; Okabe, H.; Matsunaga, K.; Takata, J.; Karube, Y.; Tsuchihashi, R.; Kinjo, J.; Mihashi, K.; et al. Antiproliferative constituents from Umbelliferae plants VII. Active triterpenes and rosmarinic acid from Centella asiatica. Biol. Pharm. Bull. 2005, 28, 173–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Chen, F.Y.; Zou, Y.; Chen, J.; Huang, W.M.; Bian, Y.T.; Luo, Y.M. Studies on chemical constituents of Chloranthus fortune. Chin. Tradit. Herb. Drugs 2020, 51, 1485–1490. [Google Scholar]
  28. Ma, X.H.; Huang, M.; Deng, S.H.; Yang, J.; Ke, R.F.; Song, P.; Yang, X.Z. Chemical constituents and bioactivity of Chloranthus multistachys Pei. J. Yunnan Univ. 2017, 39, 124–129. [Google Scholar]
  29. Sun, Z.C.; Zheng, Q.X.; Ma, G.X.; Zhang, X.P.; Yuan, J.Q.; Wu, H.F.; Liu, H.L.; Yang, J.S.; Xu, X.D. Four new phenolic acids from Clerodendranthus spicatus. Phytochem. Lett. 2014, 8, 16–21. [Google Scholar] [CrossRef]
  30. Tezuka, Y.; Stampoulis, P.; Banskota, A.H.; Awale, S.; Tran, K.Q.; Saiki, I.; Kadota, S. Constituents of the Vietnamese medicinal plant Orthosiphon stamineus. Chem. Pharm. Bull. 2000, 48, 1711–1719. [Google Scholar] [CrossRef] [Green Version]
  31. Murata, T.; Sasaki, K.; Sato, K.; Yoshizaki, F.; Yamada, H.; Mutoh, H.; Umehara, K.; Miyase, T.; Warashina, T.; Aoshima, H.; et al. Matrix metalloproteinase-2 inhibitors from Clinopodium chinense var. parviflorum. J. Nat. Prod. 2009, 72, 1379–1384. [Google Scholar] [CrossRef]
  32. Saltos, M.B.V.; Puente, B.F.N.; Malafronte, N.; Braca, A. Phenolic compounds from Clinopodium tomentosum (Kunth) Govaerts (Lamiaceae). J. Brazil. Chem. Soc. 2014, 25, 2121–2124. [Google Scholar]
  33. Wei, X.M.; Cheng, J.K.; Cheng, D.L.; Gao, L.M. Chemical constituents from Clinopodium urticifolium. J. Chin. Chem. Soc. Taip. 2004, 51, 1043–1049. [Google Scholar] [CrossRef]
  34. Kumaran, A.; Karunakaran, R.J. Activity-guided isolation and identification of free radical-scavenging components from an aqueous extract of Coleus aromaticus. Food Chem. 2007, 100, 356–361. [Google Scholar] [CrossRef]
  35. Pan, L.L.; Zhao, Q.; Liu, H.Y. Chemical contituents of Coleus forskohlii. J. Yunnan Univ. Chin. Tradit. Med. 2012, 35, 11–13, 45. [Google Scholar]
  36. Tewtrakul, S.; Miyashiro, H.; Nakamura, N.; Hattori, M.; Kawahata, T.; Otake, T.; Yoshinaga, T.; Fujiwara, T.; Supavita, T.; Yuenyongsawad, S.; et al. HIV-1 integrase inhibitory substances from Coleus parvifolius. Phytother. Res. 2003, 17, 232–239. [Google Scholar] [CrossRef]
  37. Li, H.M.; Hwang, S.H.; Kang, B.G.; Hong, J.S.; Lim, S.S. Inhibitory effects of Colocasia esculenta (L.) Schott constituents on aldose reductase. Molecules 2014, 19, 13212–13224. [Google Scholar] [CrossRef] [Green Version]
  38. Fouseki, M.M.; Damianakos, H.; Karikas, G.A.; Roussakis, C.; Gupta, M.P.; Chinou, I. Chemical constituents from Cordia alliodora and C. colloccoca (Boraginaceae) and their biological activities. Fitoterapia 2016, 115, 9–14. [Google Scholar] [CrossRef]
  39. Marini, G.; Graikou, K.; Zengin, G.; Karikas, G.A.; Gupta, M.P.; Chinou, I. Phytochemical analysis and biological evaluation of three selected Cordia species from Panama. Ind. Crop. Prod. 2018, 120, 84–89. [Google Scholar] [CrossRef]
  40. Owis, A.I.; Abo-Youssef, A.M.; Osman, A.H. Leaves of Cordia boissieri A. DC. as a potential source of bioactive secondary metabolites for protection against metabolic syndrome-induced in rats. Z. Nat. C 2017, 72, 107–118. [Google Scholar] [CrossRef]
  41. Fatima, M.; Siddiqui, B.S.; Begum, S. New neolignan glucoside and new biphenyl ether lignan from the fruits of Cordia latifolia. Chem. Nat. Compd. 2017, 53, 432–435. [Google Scholar] [CrossRef]
  42. Giles-Rivas, D.; Estrada-Soto, S.; Aguilar-Guadarrama, A.B.; Almanza-Perez, J.; Garcia-Jimenez, S.; Colin-Lozano, B.; Navarrete-Vazquez, G.; Villalobos-Molina, R. Antidiabetic effect of Cordia morelosana, chemical and pharmacological studies. J. Ethnopharmacol. 2020, 251, 112543. [Google Scholar] [CrossRef] [PubMed]
  43. Al-Musayeib, N.; Perveen, S.; Fatima, I.; Nasir, M.; Hussain, A. Antioxidant, anti-glycation and anti-Inflammatory activities of phenolic constituents from Cordia sinensis. Molecules 2011, 16, 10214–10226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ticli, F.K.; Hage, L.I.S.; Cambraia, R.S.; Pereira, P.S.; Magro, A.J.; Fontes, M.R.M.; Stabeli, R.G.; Giglio, J.R.; Franca, S.C.; Soares, A.M.; et al. Rosmarinic acid, a new snake venom phospholipase A2 inhibitor from Cordia verbenacea (Boraginaceae): Antiserum action potentiation and molecular interaction. Toxicon 2005, 46, 318–327. [Google Scholar] [CrossRef] [PubMed]
  45. Damianakos, H.; Jeziorek, M.; Syklowska-Baranek, K.; Buchwald, W.; Pietrosiuk, A.; Chinou, I. Pyrrolizidine alkaloids from Cynoglossum columnae Ten. (Boraginaceae). Phytochem. Lett. 2016, 15, 234–237. [Google Scholar] [CrossRef]
  46. Sabrin, M.S.; Selenge, E.; Takeda, Y.; Batkhuu, J.; Ogawa, H.; Jamsransuren, D.; Suganuma, K.; Murata, T. Isolation and evaluation of virucidal activities of flavanone glycosides and rosmarinic acid derivatives from Dracocephalum spp. against feline calicivirus. Phytochemistry 2021, 191, 112896. [Google Scholar] [CrossRef] [PubMed]
  47. Shi, Q.Q.; Dang, J.; Wen, H.X.; Yuan, X.; Tao, Y.D.; Wang, Q.L. Anti-hepatitis, antioxidant activities and bioactive compounds of Dracocephalum heterophyllum extracts. Bot. Stud. 2016, 57, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Olennikov, D.N.; Chirikova, N.K.; Okhlopkova, Z.M.; Zulfugarov, I.S. Chemical composition and antioxidant activity of Tánara Ótó (Dracocephalum palmatum Stephan), a medicinal plant used by the North-Yakutian Nomads. Molecules 2013, 18, 14105–14121. [Google Scholar] [CrossRef] [Green Version]
  49. Zuo, M.Y.; Yang, C.; Tian, Q.; Luo, Y.; Yang, C.; Zeng, L.; Li, G.P. Chemical constituents of Dracocephalum tanguticum Maxim of genus Dracocephalum. J. Yunnan Univ. Natl. 2015, 24, 101–103. [Google Scholar]
  50. Le, T.T.; Kang, T.K.; Do, H.T.; Nghiem, T.D.; Lee, W.B.; Jung, S.H. Protection against oxidative stress-induced retinal cell death by compounds isolated from Ehretia asperula. Nat. Prod. Commun. 2022, 16, 1934578X211067986. [Google Scholar] [CrossRef]
  51. Iqbal, K.; Nawaz, S.A.; Malik, A.; Riaz, N.; Mukhtar, N.; Mohammad, P.; Choudhary, M.I. Isolation and lipoxygenase-inhibition studies of phenolic constituents from Ehretia obtusifolia. Chem. Biodivers. 2005, 2, 104–111. [Google Scholar] [CrossRef]
  52. Simpol, L.R.; Otsuka, H.; Ohtani, K.; Kasai, R.; Yamasaki, K. Nitrile glucosides and rosmarinic acid, the histamine inhibitor from Ehretia-philippinensis. Phytochemistry 1994, 36, 91–95. [Google Scholar] [CrossRef]
  53. Li, L.; Peng, Y.; Xu, L.J.; Li, M.H.; Xiao, P.G. Flavonoid glycosides and phenolic acids from Ehretia thyrsiflora. Biochem. Syst. Ecol. 2008, 36, 915–918. [Google Scholar] [CrossRef]
  54. Zhong, J.D.; Feng, F.; Li, H.M.; Li, H.Z.; Li, R.T. Chemical constituents from Elsholtiza bodinieri Vaniot. J. Kunming Univ. Sci. Technol. 2013, 38, 75–79, 100. [Google Scholar]
  55. Li, H.; Nakashima, T.; Tanaka, T.; Zhang, Y.J.; Yang, C.R.; Kouno, I. Two new maltol glycosides and cyanogenic glycosides from Elsholtzia rugulosa Hemsl. J. Nat. Med. 2008, 62, 75–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Peng, H.Y.; Xing, Y.; Gao, L.L.; Zhang, L.; Zhang, G.L. Simultaneous separation of apigenin, luteolin and rosmarinic acid from the aerial parts of the copper-tolerant plant Elsholtzia splendens. Environ. Sci. Pollut. Res. 2014, 21, 8124–8132. [Google Scholar] [CrossRef]
  57. Devkota, H.P.; Tsushiro, K.; Watanabe, T. Bioactive phenolic compounds from the flowers of Farfugium japonicum (L.) Kitam. var. giganteum (Siebold et Zucc.) Kitam. (Asteraceae). Nat. Prod. Res. 2021. [Google Scholar] [CrossRef]
  58. Parejo, I.; Viladomat, F.; Bastida, J.; Schmeda-Hirschmann, G.; Burillo, J.; Codina, C. Bioguided isolation and identification of the nonvolatile antioxidant compounds from fennel (Foeniculum vulgare Mill.) waste. J. Agric. Food Chem. 2004, 52, 1890–1897. [Google Scholar] [CrossRef]
  59. Hawas, U.W.; Gamal-Eldeen, A.M.; El-Desouky, S.K.; Kim, Y.K.; Huefner, A.; Saf, R. Induction of caspase-8 and death receptors by a new dammarane skeleton from the dried fruits of Forsythia koreana. Z. Nat. C 2013, 68, 29–38. [Google Scholar]
  60. Shahat, A.A.; Hidayathulla, S.; Khan, A.A.; Alanazi, A.M.; Al Meanazel, O.T.; Alqahtani, A.S.; Alsaid, M.S.; Hussein, A.A. Phytochemical profiling, antioxidant and anticancer activities of Gastrocotyle hispida growing in Saudi Arabia. Acta Trop. 2019, 191, 243–247. [Google Scholar] [CrossRef]
  61. Yu, Z.B.; Wu, X.; Ye, Y.H.; Zhou, Y.W. Chemical constituents of Glechoma longituba. Nat. Prod. Res. Dev. 2008, 20, 262–264. [Google Scholar]
  62. Aquino, R.; Ciavatta, M.L.; De Simone, F.; Pizza, C. A flavanone glycoside from Hamelia patens. Phytochemistry 1990, 29, 2358–2360. [Google Scholar] [CrossRef]
  63. Trute, A.; Nahrstedt, A. Identification and quantitative analysis of phenolic compounds from the dry extract of Hedera helix. Planta Med. 1997, 63, 177–179. [Google Scholar] [CrossRef] [PubMed]
  64. Jin, X.Q.; Pang, S.Q. Studies on chemical constituents and their anti-tumor activities in roots of Helicteres angustifolia. Anhui Med. Pharmaceut. J. 2016, 20, 34–37. [Google Scholar]
  65. Tra, N.T.; Ha, N.T.T.; Cham, B.T.; Anh, L.T.T.; Yen, L.T.H.; Giang, B.L.; Anh, D.T.T.; Tuyen, N.V.; Kiem, P.V. A new benzofuran derivative from the stems of Helicteres hirsuta. Nat. Prod. Commun. 2019, 14, 1934578X19858814. [Google Scholar] [CrossRef] [Green Version]
  66. Satake, T.; Kamiya, K.; Saiki, Y.; Hama, T.; Fujimoto, U.; Kitanaka, S.; Kimura, Y.; Uzawa, J.; Endang, H.; Umar, M. Studies on the constituents of fruits of Helicteres isora L. Chem. Pharm. Bull. 1999, 47, 1444–1447. [Google Scholar] [CrossRef] [Green Version]
  67. De Lucena, H.F.S.; Madeiro, S.A.L.; Siqueira, C.D.; Barbosa, J.M.; Agra, M.D.; da Silva, M.S.; Tavares, J.F. Hypenol, a new lignan from Hypenia salzmannii. Helv. Chim. Acta 2013, 96, 1121–1125. [Google Scholar] [CrossRef]
  68. Abedini, A.; Roumy, V.; Mahieux, S.; Biabiany, M.; Standaert-Vitse, A.; Riviere, C.; Sahpaz, S.; Bailleul, F.; Neut, C.; Hennebelle, T. Rosmarinic acid and its methyl ester as antimicrobial components of the hydromethanolic extract of Hyptis atrorubens Poit. (Lamiaceae). Evid. Based Complement. Altern. Med. 2013, 2013, 604536. [Google Scholar] [CrossRef] [Green Version]
  69. Almtorp, G.T.; Hazell, A.C.; Torssell, K.B.G. A lignan and pyrone and other constituents from Hyptis-capitata. Phytochemistry 1991, 30, 2753–2756. [Google Scholar] [CrossRef]
  70. Falcao, R.A.; do Nascimento, P.L.A.; de Souza, S.A.; da Silva, T.M.G.; de Queiroz, A.C.; da Matta, C.B.B.; Moreira, M.S.A.; Camara, C.A.; Silva, T.M.S. Antileishmanial phenylpropanoids from the leaves of Hyptis pectinata (L.) Poit. Evid. Based Complement. Altern. Med. 2013, 2013, 460613. [Google Scholar] [CrossRef] [Green Version]
  71. Tang, G.Q.; Liu, X.L.; Gong, X.; Lin, X.J.; Lai, X.D.; Wang, D.; Ji, S.G. Studies on the chemical compositions of Hyptis suaveolens (L.) Poit. J. Serb. Chem. Soc. 2019, 84, 245–252. [Google Scholar] [CrossRef] [Green Version]
  72. Kuhnt, M.; Rimpler, H.; Heinrich, M. Lignans and other compounds from the Mixe-Indian medicinal plant Hyptis-verticillata. Phytochemistry 1994, 36, 485–489. [Google Scholar] [CrossRef]
  73. Furukawa, M.; Makino, M.; Ohkoshi, E.; Uchiyama, T.; Fujimoto, Y. Terpenoids and phenethyl glucosides from Hyssopus cuspidatus (Labiatae). Phytochemistry 2011, 72, 2244–2252. [Google Scholar] [CrossRef] [PubMed]
  74. Arif, Z.; Khan, S.; Farheen, S.; Kazmi, M.H.; Fatima, I.; Malik, A.; Ali, M.S.; Inamullah, F.; Afaq, S.; Shaikh, S.A.; et al. Turpesteryl ester, a new antibacterial steroid from Ipomoea turpethum. Chem. Nat. Compd. 2020, 56, 270–273. [Google Scholar] [CrossRef]
  75. Niu, X.M.; Li, S.H.; Na, Z.; Mei, S.X.; Zhao, Q.S.; Sun, H.D. Studies on chemical constituents of Isodon eriocalyx var. laxiflora. Chin. Tradit. Herb. Drugs 2003, 34, 300–303. [Google Scholar]
  76. Li, L.J.; Yu, L.J.; Wu, Z.Z.; Liu, X. Chemical constituents in ethyl acetate extract from Rabdosia flexicaulis. Chin. Tradit. Herb. Drugs 2015, 46, 339–343. [Google Scholar]
  77. Zhou, W.T.; Xie, H.H.; Xu, X.Y.; Liang, Y.G.; Wei, X.Y. Phenolic constituents from Isodon lophanthoides var. graciliflorus and their antioxidant and antibacterial activities. J. Funct. Foods 2014, 6, 492–498. [Google Scholar]
  78. Kuang, Y.H.; Lin, Q.; Liang, S.; Yao, X.H.; Wang, Z.M.; Li, C.Y. Water-soluble chemical constituents from Rabdosia lophanthoides. Chin. J. Exp. Tradit. Med. Formulae 2014, 20, 110–112. [Google Scholar]
  79. Huang, H.; Chao, Q.R.; Tan, R.X.; Sun, H.D.; Wang, D.C.; Ma, J.; Zhao, S.X. A new rosmarinic acid derivative from Isodon oresbius. Planta Med. 1999, 65, 92–93. [Google Scholar] [CrossRef] [Green Version]
  80. Zheng, X.K.; Li, Q.; Feng, W.S. Studies on chemical constituents of phenolic acids in Rabdosia rubescens. Chin. Pharm. J. 2004, 39, 335–336. [Google Scholar]
  81. Khan, S.; Taning, C.N.T.; Bonneure, E.; Mangelinckx, S.; Smagghe, G.; Ahmad, R.; Fatima, N.; Asif, M.; Shah, M.M. Bioactivity-guided isolation of rosmarinic acid as the principle bioactive compound from the butanol extract of Isodon rugosus against the pea aphid, Acyrthosiphon pisum. PLoS ONE 2019, 14, e0215048. [Google Scholar] [CrossRef] [Green Version]
  82. Jiang, B.; Hou, A.J.; Li, M.L.; Li, S.H.; Han, Q.B.; Wang, S.J.; Lin, Z.W.; Sun, H.D. Cytotoxic ent-kaurane diterpenoids from Isodon sculponeata. Planta Med. 2002, 68, 921–925. [Google Scholar] [CrossRef] [PubMed]
  83. Murata, T.; Miyase, T.; Yoshizaki, F. Hyaluronidase inhibitors from Keiskea japonica. Chem. Pharm. Bull. 2012, 60, 121–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Dehaghi, N.K.; Gohari, A.R.; Sadat-Ebrahimi, S.S.; Badi, H.N.; Amanzadeh, Y. Phytochemistry and antioxidant activity of Lallemantia iberica aerial parts. Res. J. Pharmacogn. 2016, 3, 27–34. [Google Scholar]
  85. Yadikar, N.; Bobakulov, K.; Li, G.; Aisa, H.A. Seven new phenolic compounds from Lavandula angustifolia. Phytochem. Lett. 2018, 23, 149–154. [Google Scholar] [CrossRef]
  86. Parejo, I.; Caprai, E.; Bastida, J.B.; Viladomat, F.; Jauregui, O.; Codina, C. Investigation of Lepechinia graveolens for its antioxidant activity and phenolic composition. J. Ethnopharmacol. 2004, 94, 175–184. [Google Scholar] [CrossRef]
  87. Crespo, M.I.; Chaban, M.F.; Lanza, P.A.; Joray, M.B.; Palacios, S.M.; Vera, D.M.A.; Carpinella, M.C. Inhibitory effects of compounds isolated from Lepechinia meyenii on tyrosinase. Food Chem. Toxicol. 2019, 125, 383–391. [Google Scholar] [CrossRef]
  88. Esteves, P.F.; Kuster, R.M.; Barbi, N.D.; Menezes, F.D. Chemical composition and cytotoxic activity of Lepechinia speciosa (St. Hill) Epling. Lat. Am. J. Pharm. 2010, 29, 38–44. [Google Scholar]
  89. Revoltella, S.; Baraldo, G.; Waltenberger, B.; Schwaiger, S.; Kofler, P.; Moesslacher, J.; Huber-Seidel, A.; Pagitz, K.; Kohl, R.; Jansen-Duerr, P.; et al. Identification of the NADPH oxidase 4 inhibiting principle of Lycopus europaeus. Molecules 2018, 23, 653. [Google Scholar] [CrossRef] [Green Version]
  90. Woo, E.R.; Piao, M.S. Antioxidative constituents from Lycopus lucidus. Arch. Pharm. Res. 2004, 27, 173–176. [Google Scholar] [CrossRef]
  91. Neamah, S.I.; Sarhan, I.A.; Al-Shaye’a, O.N. Extraction and evaluation of the anti-inflammatory activity of six compounds of Marrubium vulgare L. Biosci. Res. 2018, 15, 2393–2400. [Google Scholar]
  92. Murata, T.; Miyase, T.; Yoshizaki, F. Hyaluronidase inhibitory rosmarinic acid derivatives from Meehania urticifolia. Chem. Pharm. Bull. 2011, 59, 88–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tagashira, M.; Ohtake, Y. A new antioxidative 1,3-benzodioxole from Melissa officinalis. Planta Med. 1998, 64, 555–558. [Google Scholar] [CrossRef]
  94. Ji, Z.Y.; Yang, Y.X.; Zhuang, F.F.; Yan, F.L.; Wang, C.H. Chemical constituents from Melissa officinalis leaves. J. Chin. Med. Mater. 2015, 38, 510–513. [Google Scholar]
  95. Aksit, H.; Celik, S.M.; Sen, O.; Erenler, R.; Demirtas, I.; Telci, I.; Elmastas, M. Complete isolation and characterization of polar portion of Mentha dumetorum water extract. Rec. Nat. Prod. 2014, 8, 277–280. [Google Scholar]
  96. She, G.M.; Xu, C.; Liu, B.; Shi, R.B. Polyphenolic Acids from mint (the aerial of Mentha haplocalyx Briq.) with DPPH radical scavenging activity. J. Food Sci. 2010, 75, C359–C362. [Google Scholar] [CrossRef] [PubMed]
  97. Guvenalp, Z.; Ozbek, H.; Karadayi, M.; Gulluce, M.; Kuruuzum-Uz, A.; Salih, B.; Demirezer, O. Two antigenotoxic chalcone glycosides from Mentha longifolia subsp longifolia. Pharm. Biol. 2015, 53, 888–896. [Google Scholar] [CrossRef] [PubMed]
  98. Fecka, I.; Kowalczyk, A.; Cisowski, W. Optimization of the separation of flavonoid glycosides and rosmarinic acid from Mentha piperita on HPTLC plates. JPC-J. Planar Chromatogr. 2004, 17, 22–25. [Google Scholar] [CrossRef]
  99. Inoue, T.; Sugimoto, Y.; Masuda, H.; Kamei, C. Antiallergic effect of flavonoid glycosides obtained from Mentha piperita L. Biol. Pharm. Bull. 2002, 25, 256–259. [Google Scholar] [CrossRef] [Green Version]
  100. Zheng, H.J.; Gao, H.Y.; Chen, G.T.; Yang, X.K.; Wu, B.; Wu, L.J. Chemical constituents of the active parts of Mentha spicata L. (Ⅱ). J. Shenyang Pharmaceut. Univ. 2006, 23, 212–215, 255. [Google Scholar]
  101. Wang, F.; Xiang, R.Y.; Lin, C.Z.; Zhu, C.C. Chemical constituents from Mesona chinensis. J. Chin. Med. Mater. 2017, 40, 2839–2843. [Google Scholar]
  102. Akkol, E.K.; Dereli, F.T.G.; Ilhan, M. Assessment of antidepressant effect of the aerial parts of Micromeria myrtifolia Boiss. & Hohen on mice. Molecules 2019, 24, 1869. [Google Scholar] [CrossRef] [Green Version]
  103. Liang, C.Q.; Zhou, X.L.; Wang, P.C.; Tan, X.D.; Luo, Q.; Chen, X.; Pan, Z.H. Chemical constituents from stems and leaves of Microsorium fortune. J. Chin. Med. Mater. 2017, 40, 2089–2092. [Google Scholar]
  104. De Tommasi, N.; De Simone, F.; De Feo, V.; Pizza, C. Phenylpropanoid glycosides and rosmarinic acid from Momordica balsamina. Planta Med. 1991, 57, 201. [Google Scholar] [CrossRef] [PubMed]
  105. Goldansaz, S.M.; Festa, C.; Pagano, E.; De Marino, S.; Finamore, C.; Parisi, O.A.; Borrelli, F.; Sonboli, A.; D’Auria, M.V. Phytochemical and biological studies of Nepeta asterotricha Rech. f. (Lamiaceae): Isolation of nepetamoside. Molecules 2019, 24, 1684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Takeda, Y.; Ooiso, Y.; Masuda, T.; Honda, G.; Otsuka, H.; Sezik, E.; Yesilada, E. Iridoid and eugenol glycosides from Nepeta cadmea. Phytochemistry 1999, 49, 787–791. [Google Scholar] [CrossRef]
  107. Rabee, M.; Andersen, Ø.M.; Fossen, T.; Enerstvedt, K.H.; Abu Ali, H.; Rayyan, S. Acylated flavone O-glucuronides from the aerial parts of Nepeta curviflora. Molecules 2020, 25, 3782. [Google Scholar] [CrossRef] [PubMed]
  108. Ruiz-Vargas, J.A.; Morales-Ferra, D.L.; Ramirez-Avila, G.; Zamilpa, A.; Negrete-Leon, E.; Acevedo-Fernandez, J.J.; Pena-Rodriguez, L.M. α-Glucosidase inhibitory activity and in vivo antihyperglycemic effect of secondary metabolites from the leaf infusion of Ocimum campechianum mill. J. Ethnopharmacol. 2019, 243, 112081. [Google Scholar] [CrossRef]
  109. Kelm, M.A.; Nair, M.G.; Strasburg, G.M.; DeWitt, D.L. Antioxidant and cyclooxygenase inhibitory phenolic compounds from Ocimum sanctum Linn. Phytomedicine 2000, 7, 7–13. [Google Scholar] [CrossRef]
  110. Chatzopoulou, A.; Karioti, A.; Gousiadou, C.; Vivancos, V.L.; Kyriazopoulos, P.; Golegou, S.; Skaltsa, H. Depsides and other polar constituents from Origanum dictamnus L. and their in vitro antimicrobial activity in clinical strains. J. Agric. Food Chem. 2010, 58, 6064–6068. [Google Scholar] [CrossRef]
  111. Basli, A.; Delaunay, J.C.; Pedrot, E.; Bernillon, S.; Madani, K.; Monti, J.P.; Merillon, J.M.; Chibane, M.; Richard, T. New cyclolignans from Origanum glandulosum active against beta-amyloid aggregation. Rec. Nat. Prod. 2014, 8, 208–216. [Google Scholar]
  112. Erenler, R.; Sen, O.; Aksit, H.; Demirtas, I.; Yaglioglu, A.S.; Elmastas, M.; Telci, I. Isolation and identification of chemical constituents from Origanum majorana and investigation of antiproliferative and antioxidant activities. J. Sci. Food Agr. 2016, 96, 822–836. [Google Scholar] [CrossRef] [PubMed]
  113. Elmastas, M.; Celik, S.M.; Genc, N.; Aksit, H.; Erenler, R.; Gulcin, I. Antioxidant activity of an anatolian herbal tea Origanum minutiflorum: Isolation and characterization of its secondary metabolites. Int. J. Food Prop. 2018, 21, 374–384. [Google Scholar] [CrossRef] [Green Version]
  114. Erenler, R.; Meral, B.; Sen, O.; Elmastas, M.; Aydin, A.; Eminagaoglu, O.; Topcu, G. Bioassay-guided isolation, identification of compounds from Origanum rotundifolium and investigation of their antiproliferative and antioxidant activities. Pharm. Biol. 2017, 55, 1646–1653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Koukoulitsa, C.; Karioti, A.; Bergonzi, M.C.; Pescitelli, G.; Di Bari, L.; Skaltsa, H. Polar constituents from the aerial parts of Origanum vulgare L. ssp hirtum growing wild in Greece. J. Agric. Food Chem. 2006, 54, 5388–5392. [Google Scholar] [CrossRef] [PubMed]
  116. Lee, K.H.; Yang, M.C.; Kim, K.H.; Kwon, H.C.; Choi, S.U.; Lee, K.R. A new phenolic amide from the roots of Paris verticillata. Molecules 2008, 13, 41–45. [Google Scholar] [CrossRef] [Green Version]
  117. Lim, H.J.; Woo, K.W.; Lee, K.R.; Lee, S.K.; Kim, H.P. Inhibition of proinflammatory cytokine generation in lung inflammation by the leaves of Perilla frutescens and its constituents. Biomol. Ther. 2014, 22, 62–67. [Google Scholar] [CrossRef] [Green Version]
  118. Ha, T.J.; Lee, J.H.; Lee, M.H.; Lee, B.W.; Kwon, H.S.; Park, C.H.; Shim, K.B.; Kim, H.T.; Baek, I.Y.; Jang, D.S. Isolation and identification of phenolic compounds from the seeds of Perilla frutescens (L.) and their inhibitory activities against alpha-glucosidase and aldose reductase. Food Chem. 2012, 135, 1397–1403. [Google Scholar] [CrossRef]
  119. Gu, L.H.; Wu, T.; Wang, Z.T. TLC bioautography-guided isolation of antioxidants from fruit of Perilla frutescens var. acuta. LWT-Food Sci. Technol. 2009, 42, 131–136. [Google Scholar] [CrossRef]
  120. Senol, F.S.; Slusarczyk, S.; Matkowski, A.; Perez-Garrido, A.; Giron-Rodriguez, F.; Ceron-Carrasco, J.P.; den-Haan, H.; Pena-Garcia, J.; Perez-Sanchez, H.; Domaradzki, K.; et al. Selective in vitro and in silico butyrylcholinesterase inhibitory activity of diterpenes and rosmarinic acid isolated from Perovskia atriplicifolia Benth. and Salvia glutinosa L. Phytochemistry 2017, 133, 33–44. [Google Scholar] [CrossRef]
  121. Kubínová, R.; Švajdlenka, E.; Schneiderová, K.; Hanáková, Z.; Dall’ Acqua, S.; Farsa, O. Polyphenols and diterpenoids from Plectranthus forsteri ‘Marginatu’. Biochem. Syst. Ecol. 2013, 49, 39–42. [Google Scholar] [CrossRef]
  122. Ji, H.S.; Li, H.; Mo, E.J.; Kim, U.H.; Kim, Y.H.; Park, H.Y.; Jeong, T.S. Low-density lipoprotein-antioxidant flavonoids and a phenolic ester from Plectranthus hadiensis var. tomentosus. Appl. Biol. Chem. 2019, 62, 58. [Google Scholar] [CrossRef]
  123. Kubínová, R.; Pořízková, R.; Navrátilová, A.; Farsa, O.; Hanáková, Z.; Bačinská, A.; Čížek, A.; Valentová, M. Antimicrobial and enzyme inhibitory activities of the constituents of Plectranthus madagascariensis (Pers.) Benth. J. Enzym. Inhib. Med. Chem. 2014, 29, 749–752. [Google Scholar] [CrossRef] [PubMed]
  124. Kubínová, R.; Gazdová, M.; Hanáková, Z.; Jurkaninová, S.; Dall’ Acqua, S.; Cvačka, J.; Humpa, O. New diterpenoid glucoside and flavonoids from Plectranthus scutellarioides (L.) R. Br. S. Afr. J. Bot. 2019, 120, 286–290. [Google Scholar] [CrossRef]
  125. Hu, H.B.; Wang, G.W.; Liu, J.X.; Cao, H.; Zheng, X.D. Studies on phenolic compounds from Polygomun aviculane. China J. Chin. Mater. Med. 2006, 31, 740–742. [Google Scholar]
  126. Wang, Z.J.; Zhao, Y.Y.; Wang, B.; Al, T.M.; Chen, Y.Y. Depsides from Prunella vulgaris. Chin. Chem. Lett. 2000, 11, 997–1000. [Google Scholar]
  127. Kim, H.I.; Quan, F.S.; Kim, J.E.; Lee, N.R.; Kim, H.J.; Jo, S.J.; Lee, C.M.; Jang, D.S.; Inn, K.S. Inhibition of estrogen signaling through depletion of estrogen receptor alpha by ursolic acid and betulinic acid from Prunella vulgaris var. lilacina. Biochem. Biophys. Res. Commun. 2014, 451, 282–287. [Google Scholar] [CrossRef]
  128. Lee, I.K.; Kim, D.H.; Lee, S.Y.; Kim, K.R.; Choi, S.U.; Hong, J.K.; Lee, J.H.; Park, Y.H.; Lee, K.R. Triterpenoic acids of Prunella vulgaris var. lilacina and their cytotoxic activities in vitro. Arch. Pharm. Res. 2008, 31, 1578–1583. [Google Scholar]
  129. Chanu, M.B.; Labala, R.K.; Sheikh, Y.; Borah, J.C.; Ghosh, S.K.; Sahoo, D.; Singh, O.J.; Shakya, A.; Thongam, B. Bioassay guided isolation of alpha-glucosidase inhibitory compound, in vivo postprandial anti hyperglycemia and docking study of the isolated compound from the leaves of the methanolic extract of Quercus serrata. Biosci. Biotech. Res. Commun. 2018, 11, 647–657. [Google Scholar] [CrossRef]
  130. Hyun, H.B.; Shrestha, S.; Boo, K.H.; Cho, S.K. Evaluation of antioxidant potential of ethyl acetate fraction of Rosmarinus officinalis L. and its major components. J. Korean. Soc. Appl. Biol. Chem. 2015, 58, 715–722. [Google Scholar] [CrossRef]
  131. Bai, N.S.; He, K.; Roller, M.; Lai, C.S.; Shao, X.; Pan, M.H.; Ho, C.T. Flavonoids and phenolic compounds from Rosmarinus officinalis. J. Agric. Food Chem. 2010, 58, 5363–5367. [Google Scholar] [CrossRef]
  132. Koysu, P.; Genc, N.; Elmastas, M.; Aksit, H.; Erenler, R. Isolation, identification of secondary metabolites from Salvia absconditiflora and evaluation of their antioxidative properties. Nat. Prod. Res. 2019, 33, 3592–3595. [Google Scholar] [CrossRef] [PubMed]
  133. Qu, G.W.; Yue, X.D.; An, F.S.; Dai, S.J.; Li, G.S.; Li, B.F. Chemical constituents contained in Salvia castanea. China J. Chin. Mater. Med. 2012, 37, 1985–1989. [Google Scholar]
  134. Zhang, H.J.; Li, L.N. Salvianolic Acid I: A new depside from Salvia cavalerici. Planta Med. 1994, 60, 70–72. [Google Scholar] [CrossRef] [PubMed]
  135. Ertas, A.; Cakirca, H.; Yener, I.; Akdeniz, M.; Firat, M.; Topcu, G.; Kolak, U. Bioguided isolation of secondary metabolites from Salvia cerino-pruinosa Rech. f. var. cerino-pruinosa. Rec. Nat. Prod. 2021, 15, 585–592. [Google Scholar]
  136. Gao, J.F.; Ding, L.; Zhang, P.; Liu, J.X. Chemical constituents of Salvia chinensis. China J. Chin. Mater. Med. 2013, 38, 1556–1559. [Google Scholar]
  137. Qian, T.X.; Li, L.N. Isosalvianolic acid-C, a depside possessing a dibenzooxepin skeleton. Phytochemistry 1992, 31, 1068–1070. [Google Scholar]
  138. Tezuka, Y.; Kasimu, R.; Li, J.X.; Basnet, P.; Tanaka, K.; Namba, T.; Kadota, S. Constituents of roots of Salvia deserta Schang (Xinjiang-Danshen). Chem. Pharm. Bull. 1998, 46, 107–112. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, X.L.; Rena, K.; Zaoranmu, N.; Du, N.S. Studies on the chemical constituents of the flowers of Salvia deserta Schang. J. Xinjiang Med. Univ. 2003, 26, 583–585. [Google Scholar]
  140. Ai, C.B.; Deng, Q.H.; Song, W.Z.; Li, L.N. Salvianolic acidJ, a depside from Salvia-flava. Phytochemistry 1994, 37, 907–908. [Google Scholar] [CrossRef]
  141. Kang, J.; Tang, Y.B.; Liu, Q.; Guo, N.; Zhang, J.; Xiao, Z.Y.; Chen, R.Y.; Shen, Z.F. Isolation, modification, and aldose reductase inhibitory activity of rosmarinic acid derivatives from the roots of Salvia grandifolia. Fitoterapia 2016, 112, 197–204. [Google Scholar] [CrossRef]
  142. Xia, G.H.; Bi, D.W.; Li, H.Z.; Wang, L.Q. Triterpenes and phenolic acids from roots of Salvia kiaometiensis. Chin. Tradit. Herb. Drugs 2019, 50, 1043–1048. [Google Scholar]
  143. Gohari, A.R.; Saeidnia, S.; Malmir, M.; Hadjiakhoondi, A.; Ajani, Y. Flavones and rosmarinic acid from Salvia limbata. Nat. Prod. Res. 2010, 24, 1902–1906. [Google Scholar] [CrossRef] [PubMed]
  144. Shi, G.Y.; Guo, Q.M.; Zhou, F.Q. Study on chemical constituents of leaves of Salvia miltiorrhiza Bge. J. Shanxi Univ. 2015, 38, 692–695. [Google Scholar]
  145. Tung, N.H.; Hung, L.Q.; Oanh, H.V.; Huong, D.T.L.; Thuong, P.T.; Long, D.D.; Hai, N.T. Bioactive phenolic compounds from the roots of Danshen (Salvia miltiorrhiza). Nat. Prod. Commun. 2018, 13, 1305–1307. [Google Scholar] [CrossRef] [Green Version]
  146. Lu, Y.R.; Foo, L.Y. Rosmarinic acid derivatives from Salvia officinalis. Phytochemistry 1999, 51, 91–94. [Google Scholar] [CrossRef]
  147. Cioffi, G.; Bader, A.; Malafronte, A.; Dal Piaz, F.; De Tommasi, N. Secondary metabolites from the aerial parts of Salvia palaestina Bentham. Phytochemistry 2008, 69, 1005–1012. [Google Scholar] [CrossRef]
  148. Nugroho, A.; Kim, M.H.; Choi, J.; Baek, N.I.; Park, H.J. In vivo sedative and gastroprotective activities of Salvia plebeia extract and its composition of polyphenols. Arch. Pharm. Res. 2012, 35, 1403–1411. [Google Scholar] [CrossRef]
  149. Gong, X.; Yang, S.S. Isolation, identification and antioxidant properties of flavonoids from Salvia plebeia. Chin. Wild Plant. Resour. 2013, 32, 24–27. [Google Scholar]
  150. Yang, Y.; Bing, Z.; Sun, L.N.; Wu, Z.J.; Chen, W.S. Chemical constituents of Salvia przewalskii Maxim. Asian J. Chem. 2013, 25, 1747–1748. [Google Scholar]
  151. Wu, Z.J.; Ouyang, M.A.; Yang, C.R. Polyphenolic constituents of Salvia przewalskii. Acta Bot. Yunnanica 1999, 21, 512–516. [Google Scholar]
  152. Wu, Z.J.; Ouyang, M.A.; Yang, C.R. Polyphenolic constituents of Salvia sonchifolia. Acta Bot. Yunnanica 1999, 21, 393–398. [Google Scholar]
  153. Moharram, F.A.; Marzouk, M.S.; El-Shenawy, S.M.; Gaara, A.H.; El Kady, W.M. Polyphenolic profile and biological activity of Salvia splendens leaves. J. Pharm. Pharmacol. 2012, 64, 1678–1687. [Google Scholar] [CrossRef] [PubMed]
  154. Çulhaoğlu, B.; Hatipoğlu, S.D.; Dönmez, A.A.; TopÇu, G. Antioxidant and anticholinesterase activities of lupane triterpenoids and other constituents of Salvia trichoclada. Med. Chem. Res. 2015, 24, 3831–3837. [Google Scholar] [CrossRef]
  155. Rungsimakan, S.; Rowan, M.G. Terpenoids, flavonoids and caffeic acid derivatives from Salvia viridis L. cvar. Blue Jeans. Phytochemistry 2014, 108, 177–188. [Google Scholar] [CrossRef] [PubMed]
  156. Zhang, Z.F.; Chen, H.S.; Li, J.R.; Jiang, J.D.; Li, Z.R. Studies on polyphenolic chemical constituents from root of Salvia yunnansis. China J. Chin. Mater. Med. 2007, 32, 1886–1890. [Google Scholar]
  157. Arda, N.; Goren, N.; Kuru, A.; Pengsuparp, T.; Pezzuto, J.M.; Qiu, S.X.; Cordell, G.A. Saniculoside N from Sanicula europaea L. J. Nat. Prod. 1997, 60, 1170–1173. [Google Scholar] [CrossRef]
  158. Zhou, L.Y.; Liu, H.Y.; Xie, B.B.; Liu, Z.H.; Chen, C.X. Two new glycosides from Sanicula lamelligera. Z. Nat. B 2006, 61, 607–610. [Google Scholar] [CrossRef]
  159. Huang, M.J.; Li, Y.L.; Zeng, G.Y.; Yuan, W.M.; Tan, J.B.; Tan, G.S.; Zhou, Y.J. Chemical constituents of Sarcandra glabra. Cent. South Pharm. 2007, 5, 459–461. [Google Scholar]
  160. Moghadam, S.E.; Ebrahimi, S.N.; Gafner, F.; Ochola, J.B.; Marubu, R.M.; Lwande, W.; Haller, B.F.; Salehi, P.; Hamburger, M. Metabolite profiling for caffeic acid oligomers in Satureja biflora. Ind. Crop. Prod. 2015, 76, 892–899. [Google Scholar] [CrossRef]
  161. Lee, I.K.; Kim, M.A.; Lee, S.Y.; Hong, J.K.; Lee, J.H.; Lee, K.R. Phytochemical constituents of Schizonepeta tenuifolia Briquet. Nat. Prod. Sci. 2008, 14, 100–106. [Google Scholar] [CrossRef] [Green Version]
  162. Deveci, E.; Tel-Cayan, G.; Duru, M.E.; Ozturk, M. Phytochemical contents, antioxidant effects, and inhibitory activities of key enzymes associated with Alzheimer’s disease, ulcer, and skin disorders of Sideritis albiflora and Sideritis leptoclada. J. Food Biochem. 2019, 43, e13078. [Google Scholar] [CrossRef] [PubMed]
  163. Garcia, J.M.; Prieto, L.J.; Guevara, A.; Malagon, D.; Osorio, C. Chemical studies of yellow tamarillo (Solanum betaceum Cav.) fruit flavor by using a molecular sensory approach. Molecules 2016, 21, 1729. [Google Scholar] [CrossRef] [PubMed]
  164. Taiwo, B.J.; Obuotor, E.M.; Onawunmi, G.O.; Ogundaini, A.O. Radical scavenging compounds from the aerial parts of Solenostemon monostachys Briq (Lamiaceae). Afr. J. Tradit. Complement. Altern. Med. 2015, 12, 140–144. [Google Scholar] [CrossRef] [Green Version]
  165. Trifan, A.; Skalicka-Wozniak, K.; Granica, S.; Czerwinska, M.E.; Kruk, A.; Marcourt, L.; Wolfender, J.L.; Wolfram, E.; Esslinger, N.; Grubelnik, A.; et al. Syrnphytum officinale L.: Liquid-liquid chromatography isolation of caffeic acid oligomers and evaluation of their influence on pro-inflammatory cytokine release in LPS-stimulated neutrophils. J. Ethnopharmacol. 2020, 262, 113169. [Google Scholar] [CrossRef] [PubMed]
  166. Boonyarikpunchai, W.; Sukrong, S.; Towiwat, P. Antinociceptive and anti-inflammatory effects of rosmarinic acid isolated from Thunbergia laurifolia Lindl. Pharmacol. Biochem. Behav. 2014, 124, 67–73. [Google Scholar] [CrossRef]
  167. Dall’ Acqua, S.; Peron, G.; Ferrari, S.; Gandin, V.; Bramucci, M.; Quassinti, L.; Martonfi, P.; Maggi, F. Phytochemical investigations and antiproliferative secondary metabolites from Thymus alternans growing in Slovakia. Pharm. Biol. 2017, 55, 1162–1170. [Google Scholar] [CrossRef] [Green Version]
  168. Khouya, T.; Ramchoun, M.; Amrani, S.; Harnafi, H.; Rouis, M.; Couchie, D.; Simmet, T.; Alem, C. Anti-inflammatory and anticoagulant effects of polyphenol-rich extracts from Thymus atlanticus: An in vitro and in vivo study. J. Ethnopharmacol. 2020, 252, 112475. [Google Scholar] [CrossRef]
  169. Erenler, R.; Sen, O.; Yildiz, I.; Aydin, A. Antiproliferative Activities of chemical constituents isolated from Thymus praecox subsp grossheimii (Ronniger) Jalas. Rec. Nat. Prod. 2016, 10, 766–770. [Google Scholar]
  170. Sevindik, H.G.; Ozgen, U.; Atila, A.; Er, H.O.; Kazaz, C. Phtytochemical studies and quantitative HPLC analysis of rosmarinic acid and luteolin 5-O-β-D-glucopyranoside on Thymus praecox subsp grossheimii var. grossheimii. Chem. Pharm. Bull. 2015, 63, 720–725. [Google Scholar] [CrossRef] [Green Version]
  171. Lee, I.C.; Bae, J.S.; Kim, T.; Kwon, O.J.; Kim, T.H. Polyphenolic constituents from the aerial parts of Thymus quinquecostatus var. japonica collected on Ulleung island. J. Korean Soc. Appl. Biol. Chem. 2011, 54, 811–816. [Google Scholar] [CrossRef]
  172. Aziz, S.; Irshad, M.; Habib-ur-Rehman. Isolation of a new antibacterial polyphenol from Thymus serpyllum. Chem. Nat. Compd. 2014, 49, 1023–1027. [Google Scholar] [CrossRef]
  173. Kontogiorgis, C.; Ntella, M.; Mpompou, L.; Karallaki, F.; Athanasios, P.; Hadjipavlou-Litina, D.; Lazari, D. Study of the antioxidant activity of Thymus sibthorpii Bentham (Lamiaceae). J. Enzym. Inhib. Med. Chem. 2016, 31, 154–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ozgen, U.; Mavi, A.; Terzi, Z.; Kazaz, C.; Asci, A.; Kaya, Y.; Secen, H. Relationship between chemical structure and antioxidant activity of luteolin and its glycosides isolated from Thymus sipyleus subsp sipyleus var. sipyleus. Rec. Nat. Prod. 2011, 5, 12–21. [Google Scholar]
  175. Engelbertz, J.; Lechtenberg, M.; Studt, L.; Hensel, A.; Verspohl, E.J. Bioassay-guided fractionation of a thymol-deprived hydrophilic thyme extract and its antispasmodic effect. J. Ethnopharmacol. 2012, 141, 848–853. [Google Scholar] [CrossRef] [PubMed]
  176. Lin, Y.L.; Chang, Y.Y.; Kuo, Y.H.; Shiao, M.S. Anti-lipid-peroxidative principles from Tournefortia sarmentosa. J. Nat. Prod. 2002, 65, 745–747. [Google Scholar] [CrossRef]
  177. Guo, X.J.; Ding, X.; Dong, Y.C.; Xu, G.H. Chemical constituents of Veronica sibirica L. Pennell. J. Med. Sci. Yanbian Univ. 2018, 41, 14–23. [Google Scholar]
  178. Li, G.Z.; Meng, Q.Y.; Wang, L.J.; Luo, B.; Ge, Z.H.; Liu, W.J. Chemical constituents from Ziziphora clinopodioides. Chin. Tradit. Herbal Drugs 2015, 46, 2534–2539. [Google Scholar]
  179. Wang, J.Y.; Pan, X.R.; Han, Y.; Guo, D.S.; Guo, Q.Q.; Li, R.G. Rosmarinic acid from eelgrass shows nematicidal and antibacterial activities against pine wood nematode and its carrying bacteria. Mar. Drugs 2012, 10, 2729–2740. [Google Scholar] [CrossRef] [Green Version]
  180. Achamlale, S.; Rezzonico, B.; Grignon-Dubois, M. Rosmarinic acid from beach waste: Isolation and HPLC quantification in Zostera detritus from Arcachon lagoon. Food Chem. 2009, 113, 878–883. [Google Scholar] [CrossRef]
  181. De Eknamkul, W.; Ellis, B.E. Tyrosine aminotransferase: The entrypoint enzyme of the tyrosine-derived pathway in rosmarinic acid biosynthesis. Phytochemistry 1987, 26, 1941–1946. [Google Scholar] [CrossRef]
  182. Petersen, M.; Häusler, E.; Karwatzki, B.; Meinhard, J. Proposed biosynthetic pathway for rosmarinic acid in cell cultures of Coleus blumei Benth. Planta 1993, 189, 10–14. [Google Scholar] [CrossRef]
  183. Petersen, M. Cytochrome P450-Dependent hydroxylation in the biosynthesis of rosmarinic acid in Coleus. Phytochemistry 1997, 45, 1165–1172. [Google Scholar] [CrossRef]
  184. Razzaque, A.; Ellis, B.E. Rosmarinic acid production in Coleus cell cultures. Planta 1977, 137, 287–291. [Google Scholar] [CrossRef] [PubMed]
  185. Berger, A.; Meinhard, J.; Petersen, M. Rosmarinic acid synthase is a new member of the superfamily of BAHD acyltransferases. Planta 2006, 224, 1503–1510. [Google Scholar] [CrossRef]
  186. Yang, J.D.; Ma, J.Y.; Li, Q.; Zhu, T.T.; Ji, Q.; Zhang, L. Bioinformatics analysis of rosmarinic acid synthase based on genome of Salvia miltiorrhiza. Genom. Appl. Biol. 2017, 36, 1611–1622. [Google Scholar]
  187. Sha, X.X.; Su, S.L.; Shen, F.; Jiang, S.; Yan, H.; Guo, S.; Qian, D.W.; Duan, J.A. Distribution of salvianolic acids in aerial parts of Salvia miltiorrhiza during different growing periods and accumulation dynamic analysis. Chin. Tradit. Herbal Drugs 2015, 46, 3414–3419. [Google Scholar]
  188. Oliveira, G.D.A.R.; de Oliveira, A.E.; da Conceição, E.C.; Leles, M.I.G. Multiresponse optimization of an extraction procedure of carnosol and rosmarinic and carnosic acids from rosemary. Food Chem. 2016, 211, 465–473. [Google Scholar] [CrossRef]
  189. Hernández-Hernández, E.; Ponce-Alquicira, E.; Jaramillo-Flores, M.E.; Guerrero Legarreta, I. Antioxidant effect rosemary (Rosmarinus officinalis L.) and oregano (Origanum vulgare L.) extracts on TBARS and colour of model raw pork batters. Meat Sci. 2009, 81, 410–417. [Google Scholar] [CrossRef]
  190. Dastmalchi, K.; Damien Dorman, H.J.; Laakso, I.; Hiltunen, R. Chemical composition and antioxidative activity of Moldavian balm (Dracocephalum moldavica L.) extracts. LWT-Food Sci. Technol. 2007, 40, 1655–1663. [Google Scholar] [CrossRef]
  191. Shekarchi, M.; Hajimehdipoor, H.; Saeidnia, S.; Gohari, A.R.; Hamedani, M.P. Comparative study of rosmarinic acid content in some plants of Labiatae family. Pharmacogn. Mag. 2012, 8, 37–41. [Google Scholar]
  192. Chatterjee, A.; Tandon, S.; Ahmad, A. Comparative extraction and downstream processing techniques for quantitative analysis of rosmarinic acid in Rosmarinus officinalis. Asian J. Chem. 2014, 26, 4313–4318. [Google Scholar] [CrossRef]
  193. Miron, T.L.; Herrero, M.; Ibáñez, E. Enrichment of antioxidant compounds from lemon balm (Melissa officinalis) by pressurized liquid extraction and enzyme-assisted extraction. J. Chromatogr. A 2013, 1288, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Sánchez-Camargo, A.P.; Mendiola, J.A.; Valdés, A.; Castro-Puyana, M.; García-Cañas, V.; Cifuentes, A.; Herrero, M.; Ibáñez, E. Supercritical antisolvent fractionation of rosemary extracts obtained by pressurized liquid extraction to enhance their antiproliferative activity. J. Supercrit. Fluids 2016, 107, 581–589. [Google Scholar] [CrossRef]
  195. Sik, B.; Hanczne, E.L.; Kapcsandi, V.; Ajtony, Z. Conventional and nonconventional extraction techniques for optimal extraction processes of rosmarinic acid from six Lamiaceae plants as determined by HPLC-DAD measurement. J. Pharm. Biomed. 2020, 184, 113173. [Google Scholar] [CrossRef] [PubMed]
  196. Zu, G.; Zhang, R.R.; Yang, L.; Ma, C.H.; Zu, Y.G.; Wang, W.J.; Zhao, C.J. Ultrasound-assisted extraction of carnosic acid and rosmarinic acid using ionic liquid solution from Rosmarinus officinalis. Int. J. Mol. Sci. 2012, 13, 11027–11043. [Google Scholar] [CrossRef] [Green Version]
  197. Liu, T.T.; Sui, X.Y.; Zhang, R.R.; Yang, L.; Zu, Y.G.; Zhang, L.; Zhang, Y.; Zhang, Z.H. Application of ionic liquids based microwave-assisted simultaneous extraction of carnosic acid, rosmarinic acid and essential oil from Rosmarinus officinalis. J. Chromatogr. A 2011, 1218, 8480–8489. [Google Scholar] [CrossRef]
  198. Lim, S.H.; Nam, K.H.; Kim, K.; Yi, S.A.; Lee, J.; Han, J.W. Rosmarinic acid methyl ester regulates ovarian cancer cell migration and reverses cisplatin resistance by inhibiting the expression of Forkhead Box M1. Pharmaceuticals 2020, 13, 302. [Google Scholar] [CrossRef]
  199. Liu, R.X.; Heiss, E.H.; Waltenberger, B.; Blažević, T.; Schachner, D.; Jiang, B.H.; Krystof, V.; Liu, W.H.; Schwaiger, S.; Peña-Rodríguez, L.M.; et al. Constituents of mediterranean spices counteracting vascular smooth muscle cell proliferation: Identification and characterization of rosmarinic acid methyl ester as a novel inhibitor. Mol. Nutr. Food Res. 2018, 62, 1700860. [Google Scholar] [CrossRef]
  200. Panya, A.; Laguerre, M.; Bayrasy, C.; Lecomte, J.; Villeneuve, P.; McClements, D.J.; Decker, E.A. An investigation of the versatile antioxidant mechanisms of action of rosmarinate alkyl esters in oil-in-water emulsions. J. Agric. Food Chem. 2012, 60, 2692–2700. [Google Scholar] [CrossRef]
  201. Suriyarak, S.; Gibis, M.; Schmidt, H.; Velleneuve, P.; Weiss, J. Antimicrobial mechanism and activity of dodecyl rosmarinate against Staphylococcus carnosus LTH1502 as influenced by addition of salt and change in pH. J. Food Protect. 2014, 77, 444–452. [Google Scholar] [CrossRef]
  202. Thammason, H.; Khetkam, P.; Pabuprapap, W.; Suksamrarn, A.; Kunthalert, D. Ethyl rosmarinate inhibits lipopolysaccharide-induced nitric oxide and prostaglandin E-2 production in alveolar macrophages. Eur. J. Pharmacol. 2018, 824, 17–23. [Google Scholar] [CrossRef] [PubMed]
  203. Zhu, F.X.; Xu, Z.M.; Yonekura, L.; Yang, R.H.; Tamura, H. Antiallergic activity of rosmarinic acid esters is modulated by hydrophobicity, and bulkiness of alkylside chain. Biosci. Biotechnol. Biochem. 2015, 79, 1178–1182. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, J.X.; Zhang, Y.; Hu, Q.P.; Li, J.Q.; Liu, Y.T.; Wu, Q.G.; Wu, J.G.; Lai, X.P.; Zhang, Z.D.; Li, X.; et al. Anti-inflammatory effects of rosmarinic acid-4-O-β-D-glucoside in reducing acute lung injury in mice infected with influenza virus. Antivir. Res. 2017, 144, 34–43. [Google Scholar] [CrossRef] [PubMed]
  205. Lim, C.; Kim, C.H.; Lim, S.H.; Cho, S. Salvianolic acid B attenuated ischemia/reperfusion-induced brain injury in mice by inhibiting reactive oxygen species-mediated inflammation. Rec. Nat. Prod. 2021, 15, 25–34. [Google Scholar] [CrossRef]
  206. Tao, X.M.; Li, D.; Zhang, C.; Wen, G.H.; Wu, C.; Xu, Y.Y.; Kan, Y.; Lu, W.P.; Ding, H.Y.; Yang, Y. Salvianolic acid B protects against acute and chronic liver injury by inhibiting Smad2C/L phosphorylation. Exp. Ther. Med. 2021, 21, 34. [Google Scholar] [CrossRef] [PubMed]
  207. Zhang, Y.F.; Feng, X.T.; Du, M.; Ding, J.; Liu, P. Salvianolic acid B attenuates the inflammatory response in atherosclerosis by regulating MAPKs/NF-kappa B signaling pathways in LDLR-/- mice and RAW264.7 cells. Int. J. Immunopathol. Pharmacol. 2021, 36, 03946320221079468. [Google Scholar] [CrossRef]
  208. Katary, M.A.; Abdelsayed, R.; Alhashim, A.; Abdelhasib, M.; Elmarakby, A.A. Salvianolic acid B slows the progression of breast cancer cell growth via enhancement of apoptosis and reduction of oxidative stress, inflammation, and angiogenesis. Int. J. Mol. Sci. 2019, 20, 5653. [Google Scholar] [CrossRef] [Green Version]
  209. Huang, M.A.; Wang, P.J.; Xu, S.Y.; Xu, W.; Xu, W.; Chu, K.D.; Lu, J.J. Biological activities of salvianolic acid B from Salvia miltiorrhiza on type 2 diabetes induced by high-fat diet and streptozotocin. Pharm. Biol. 2015, 53, 1058–1065. [Google Scholar] [CrossRef] [Green Version]
  210. Bilgin, M.; Elhussein, E.A.A.; Ozyurek, M.; Guclu, K.; Sahin, S. Optimizing the extraction of polyphenols from Sideritis montana L. using response surface methodology. J. Pharmaceut. Biomed. 2018, 158, 137–143. [Google Scholar] [CrossRef]
  211. Duletić-Laušević, S.; Aradski, A.A.; Kolarević, S.; Vuković-Gačić, B.; Oalđe, M.; Živković, J.; Šavikin, K.; Marin, P.D. Antineurodegenerative, antioxidant and antibacterial activities and phenolic components of Origanum majorana L. (Lamiaceae) extracts of different origin. J. Appl. Bot. Food Qual. 2018, 91, 126–134. [Google Scholar]
  212. Piatczak, E.; Owczarek, A.; Lisiecki, P.; Gonciarz, W.; Kozlowska, W.; Szemraj, M.; Chmiela, M.; Kiss, A.K.; Olszewska, M.A.; Grzegorczyk-Karolak, I. Identification and quantification of phenolic compounds in Salvia cadmica Boiss. and their biological potential. Ind. Crops Prod. 2021, 160, 113113. [Google Scholar] [CrossRef]
  213. Li, P.H.; Liu, A.L.; Li, Y.H.; Yuan, B.; Xiao, W.J.; Liu, Z.H.; Zhang, S.; Lin, H.Y. Development and validation of an analytical method based on HPLC-ELSD for the simultaneous determination of rosmarinic acid, carnosol, carnosic acid, oleanolic acid and ursolic acid in rosemary. Molecules 2019, 24, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Li, X.C.; Yu, C.; Sun, W.K.; Liu, G.Y.; Jia, J.Y.; Wang, Y.P. Simultaneous determination of magnesium lithospermate B, rosmarinic acid, and lithospermic acid in beagle dog serum by liquid chromatography/tandem mass spectrometry. Rapid Commun. Mass Spectrom. 2004, 18, 2878–2882. [Google Scholar] [CrossRef] [PubMed]
  215. Li, S.; Xie, X.M.; Li, D.X.; Yu, Z.G.; Tong, L.; Zhao, Y.L. Simultaneous determination and tissue distribution studies of four phenolic acids in rat tissue by UFLC-MS/MS after intravenous administration of salvianolic acid for injection. Biomed. Chromatogr. 2018, 32, e4128. [Google Scholar] [CrossRef]
  216. Liu, Z.; Wu, X.Q.; Si, Z.Y.; Kong, D.S.; Yang, D.W.; Zhou, F.Q.; Wang, Z.J. Simultaneous determination of nine constituents by validated UFLC-MS/MS in the plasma of cough variant asthma rats and its application to pharmacokinetic study after oral administration of Huanglong cough oral liquid. J. Pharm. Biomed. 2021, 193, 113726. [Google Scholar] [CrossRef]
  217. Qu, C.; Xu, D.Q.; Yue, S.J.; Shen, L.F.; Zhou, G.S.; Chen, Y.Y.; Wang, X.P.; Bai, J.Q.; Liu, F.; Tang, Y.P.; et al. Pharmacodynamics and pharmacokinetics of Danshen in isoproterenol-induced acute myocardial ischemic injury combined with Honghua. J. Ethnopharmacol. 2020, 247, 112284. [Google Scholar] [CrossRef]
  218. Baskan, S.; Oztekin, N.; Erim, F.B. Determination of carnosic acid and rosmarinic acid in sage by capillary electrophoresis. Food Chem. 2007, 101, 1748–1752. [Google Scholar] [CrossRef]
  219. Adimcilar, V.; Kalaycioglu, Z.; Aydogdu, N.; Dirmenci, T.; Kahraman, A.; Erim, F.B. Rosmarinic and carnosic acid contents and correlated antioxidant and antidiabetic activities of 14 Salvia species from Anatolia. J. Pharm. Biomed. 2019, 175, 112763. [Google Scholar] [CrossRef]
  220. Tang, Z.X.; Zeng, Y.K.; Zhou, Y.; He, P.G.; Fang, Y.Z.; Zang, S.L. Determination of active ingredients of Origanum Vulgare L. and its medicinal preparations by capillary electrophoresis with electrochemical detection. Anal. Lett. 2006, 39, 2861–2875. [Google Scholar] [CrossRef]
  221. Acosta, G.; Arce, S.; Martinez, L.D.; Llabot, J.; Gomez, M.R. Monitoring of phenolic compounds for the quality control of Melissa officinalis products by capillary electrophoresis. Phytochem. Anal. 2012, 23, 177–183. [Google Scholar] [CrossRef]
  222. Cao, J.L.; Wei, J.C.; Tian, K.; Su, H.X.; Wan, J.B.; Li, P. Simultaneous determination of seven phenolic acids in three Salvia species by capillary zone electrophoresis with β-cyclodextrin as modifier. J. Sep. Sci. 2014, 37, 3738–3744. [Google Scholar] [CrossRef] [PubMed]
  223. Cheung, H.Y.; Zhang, Q.F. Enhanced analysis of triterpenes, flavonoids and phenolic compounds in Prunella vulgaris L. by capillary zone electrophoresis with the addition of running buffer modifiers. J. Chromatogr. A 2008, 1213, 231–238. [Google Scholar] [CrossRef] [PubMed]
  224. Gao, J.; Deng, G.H.; Chen, S.Y.; Wang, H.; Qin, Y.L. Determination of rosmarinic acid in Salvia miltiorrhiza and Compound Salvia tablets by micellar electrokiectic capillary chromatography. Lishizhen Med. Mater. Med. Res. 2012, 23, 1095–1096. [Google Scholar]
  225. Kirchler, C.G.; Pezzei, C.K.; Bec, K.B.; Henn, R.; Ishigaki, M.; Ozaki, Y.; Huck, C.W. Critical evaluation of NIR and ATR-IR spectroscopic quantifications of rosmarinic acid in Rosmarini folium supported by quantum chemical calculations. Planta Med. 2017, 83, 1076–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Pezzei, C.K.; Schönbichler, S.A.; Hussain, S.; Kirchler, C.G.; Huck-Pezzei, V.A.; Popp, M.; Krolitzek, J.; Bonn, G.K.; Huck, C. Near-infrared and mid-infrared spectroscopic techniques for a fast and nondestructive quality control of Thymi herba. Planta Med. 2018, 84, 420–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Saltas, D.; Pappas, C.S.; Daferera, D.; Tarantilis, P.A.; Polissiou, M.G. Direct determination of rosmarinic acid in Lamiaceae herbs using diffuse reflectance infrared fourier transform spectroscopy (DRIFTS) and chemometrics. J. Agric. Food Chem. 2013, 61, 3235–3241. [Google Scholar] [CrossRef]
  228. Hu, Z.N.; Huang, L.J.; Chen, W.P. The inhibitory effects of rosmarinic acid on catabolism induced by IL-1β in rat chondrocyte. Acta Biochim. Pol. 2018, 65, 535–538. [Google Scholar] [CrossRef]
  229. Youn, J.; Lee, K.H.; Won, J.; Huh, S.J.; Yun, H.S.; Cho, W.G.; Paik, D.J. Beneficial effects of rosmarinic acid on suppression of collagen induced arthritis. J. Rheumatol. 2003, 30, 1203–1207. [Google Scholar]
  230. Jang, A.H.; Kim, T.H.; Kim, G.D.; Kim, J.E.; Kim, H.J.; Kim, S.S.; Jin, Y.H.; Park, Y.S.; Park, C.S. Rosmarinic acid attenuates 2,4-dinitrofluorobenzene-induced atopic dermatitis in NC/Nga mice. Int. Immunopharmacol. 2011, 11, 1271–1277. [Google Scholar] [CrossRef]
  231. Jin, B.R.; Chung, K.S.; Cheon, S.Y.; Lee, M.; Hwang, S.; Noh Hwang, S.; Rhee, K.J.; An, H.J. Rosmarinic acid suppresses colonic inflammation in dextran sulphate sodium (DSS)-induced mice via dual inhibition of NF-κB and STAT3 activation. Sci. Rep. 2017, 7, 46252. [Google Scholar] [CrossRef]
  232. Mai, P.; Chen, C.; Xiao, X.H.; Ma, X.; Shi, Y.P.; Miao, G.Y.; Zhang, L.P. Rosmarinic acid protects against ulcerative colitis by regulating macrophage polarization depending on heme oxygenase-1 in mice. Eur. J. Inflamm. 2020, 18, 2058739220959916. [Google Scholar] [CrossRef]
  233. Jiang, K.F.; Ma, X.F.; Guo, S.; Zhang, T.; Zhao, G.; Wu, H.C.; Wang, X.Y.; Deng, G.Z. Anti-inflammatory effects of rosmarinic acid in lipopolysaccharide-induced mastitis in mice. Inflammation 2018, 41, 437–448. [Google Scholar] [CrossRef] [PubMed]
  234. Fan, Y.T.; Yin, G.J.; Xiao, W.Q.; Qiu, L.; Yu, G.; Hu, Y.L.; Xing, M.; Wu, D.Q.; Cang, X.F.; Wan, R.; et al. Rosmarinic acid attenuates sodium taurocholate-induced acute pancreatitis in rats by inhibiting nuclear factor-κB activation. Am. J. Chin. Med. 2015, 43, 1117–1135. [Google Scholar] [CrossRef] [PubMed]
  235. Chu, X.; Ci, X.X.; He, J.K.; Jiang, L.X.; Wei, M.M.; Cao, Q.J.; Guan, M.F.; Xie, X.X.; Deng, X.M. Effects of a natural prolyl oligopeptidase inhibitor, rosmarinic acid, on lipopolysaccharide induced acute lung injury in mice. Molecules 2012, 17, 3586–3598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Fasolo, J.M.M.A.; Vizuete, A.F.K.; Rico, E.P.; Rambo, R.B.S.; Toson, N.S.B.; Santos, E.; de Oliveira, D.L.; Goncalves, C.A.S.; Schapoval, E.E.S.; Heriques, A.T. Anti-inflammatory effect of rosmarinic acid isolated from Blechnum brasiliense in adult zebrafish brain. Comp. Biochem. Phys. C 2021, 239, 108874. [Google Scholar] [CrossRef]
  237. Van Dyke, T.E.; Braswell, L.; Offenbacher, S. Inhibition of gingivitis by topical application of ebselen and rosmarinic acid. Agents Actions 1986, 19, 376–377. [Google Scholar] [CrossRef]
  238. Wang, Y.Y.; Meng, J.; Men, L.; An, B.R.; Jin, X.X.; He, W.J.; Lu, S.C.; Li, N. Rosmarinic acid protects mice from concanavalin A-induced hepatic injury through AMPK signaling. Biol. Pharm. Bull. 2020, 43, 1749–1759. [Google Scholar] [CrossRef]
  239. Oh, H.A.; Park, C.S.; Ahn, H.J.; Park, Y.S.; Kim, H.M. Effect of Perilla frutescens var. acuta Kudo and rosmarinic acid on allergic inflammatory reactions. Exp. Biol. Med. 2011, 236, 99–106. [Google Scholar] [CrossRef]
  240. Liang, Z.M.; Wu, L.Q.; Deng, X.; Liang, Q.L.; Xu, Y.F.; Deng, R.H.; Lv, L.; Ji, M.; Hao, Z.H.; He, J.K. The antioxidant rosmarinic acid ameliorates oxidative lung damage in experimental allergic asthma via modulation of NADPH oxidases and antioxidant enzymes. Inflammation 2020, 43, 1902–1912. [Google Scholar] [CrossRef]
  241. Lin, C.X.; Xiao, J.; Xi, Y.; Zhang, X.Y.; Zhong, Q.Q.; Zheng, H.J.; Cao, Y.; Chen, Y.J. Rosmarinic acid improved antioxidant properties and healthspan via the IIS and MAPK pathways in Caenorhabditis elegans. Biofactors 2019, 45, 774–787. [Google Scholar] [CrossRef]
  242. Khalaf, A.A.; Ibrahim, E.I.; Ibrahim, M.A.; Tohamy, A.F.; Aboseada, M.A.; Hassan, H.M.; Zaki, A.R. Rosmarinic acid attenuates chromium-induced hepatic and renal oxidative damage and DNA damage in rats. J. Biochem. Mol. Toxic. 2020, 34, e22579. [Google Scholar] [CrossRef] [PubMed]
  243. Cai, X.; Yang, F.; Zhu, L.H.; Xia, Y.; Wu, Q.Y.; Xue, H.Q.; Lu, Y.H. Rosmarinic acid, the main effective constituent of Orthosiphon stamineus, inhibits intestinal epithelial apoptosis via regulation of the Nrf2 pathway in mice. Molecules 2019, 24, 3027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Vlavcheski, F.; Naimi, M.; Murphy, B.; Hudlicky, T.; Tsiani, E. Rosmarinic acid, a rosemary extract polyphenol, increases skeletal muscle cell glucose uptake and activates AMPK. Molecules 2017, 22, 1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Runtuwene, J.; Cheng, K.C.; Asakawa, A.; Amitani, H.; Amitani, M.; Morinaga, A.; Inui, A. Rosmarinic acid ameliorates hyperglycemia and insulin sensitivity in diabetic rats, potentially by modulating the expression of PEPCK and GLUT4. Drug Des. Dev. Ther. 2016, 10, 2193–2202. [Google Scholar]
  246. Jayanthy, G.; Subramanian, S. RA abrogates hepatic gluconeogenesis and insulin resistance by enhancing IRS-1 and AMPK signalling in experimental type 2 diabetes. RSC Adv. 2015, 5, 44053–44067. [Google Scholar] [CrossRef]
  247. Azevedo, M.F.; Lima, C.F.; Fernandes-Ferreira, M.; Almeida, M.J.; Wilson, J.M.; Pereira-Wilson, C. Rosmarinic acid, major phenolic constituent of Greek sage herbal tea, modulates rat intestinal SGLT1 levels with effects on blood glucose. Mol. Nutr. Food Res. 2011, 55, S15–S25. [Google Scholar] [CrossRef] [Green Version]
  248. Li, H.; Zhang, Y.F.; Chen, H.H.; Huang, E.; Zhuang, H.L.; Li, D.; Ni, F. Rosmarinic acid inhibits stem-like breast cancer through Hedgehog and Bcl-2/Bax signaling pathways. Pharmacogn. Mag. 2019, 15, 600–606. [Google Scholar] [CrossRef]
  249. Cao, W.; Hu, C.; Wu, L.L.; Xu, L.B.; Jiang, W.Z. Rosmarinic acid inhibits inflammation and angiogenesis of hepatocellular carcinoma by suppression of NF-κB signaling in H22 tumor-bearing mice. J. Pharmacol. Sci. 2016, 132, 131–137. [Google Scholar] [CrossRef] [Green Version]
  250. Venkatachalam, K.; Gunasekaran, S.; Jesudoss, V.A.S.; Namasivayam, N. The effect of rosmarinic acid on 1,2-dimethylhydrazine induced colon carcinogenesis. Exp. Toxicol. Pathol. 2013, 65, 409–418. [Google Scholar] [CrossRef]
  251. Karthikkumar, V.; Sivagami, G.; Vinothkumar, R.; Rajkumar, D.; Nalini, N. Modulatory efficacy of rosmarinic acid on premalignant lesions and antioxidant status in 1,2-dimethylhydrazine induced rat colon carcinogenesis. Environ. Toxicol. Pharmacol. 2012, 34, 949–958. [Google Scholar] [CrossRef]
  252. Sharmila, R.; Manoharan, S. Anti-tumor activity of rosmarinic acid in 7,12-dimethylbenz(a)anthracene (DMBA) induced skin carcinogenesis in Swiss albino mice. Indian J. Exp. Biol. 2012, 50, 187–194. [Google Scholar] [PubMed]
  253. Canturk, Z.; Dikmen, M.; Artagan, O.; Ozarda, M.G.; Ozturk, N. Cytotoxic effects of resveratrol, rutin and rosmarinic acid on ARH-77 human (multiple myeloma) cell line. Nat. Prod. Commun. 2016, 11, 1441–1444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Jang, Y.G.; Hwang, K.A.; Choi, K.C. Rosmarinic acid, a component of rosemary tea, Induced the cell cycle arrest and apoptosis through modulation of HDAC2 expression in prostate cancer cell lines. Nutrients 2018, 10, 1784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Jin, B.X.; Liu, J.N.; Gao, D.N.; Xu, Y.; He, L.; Zang, Y.J.; Li, N.; Lin, D.D. Detailed studies on the anticancer action of rosmarinic acid in human Hep-G2 liver carcinoma cells: Evaluating its effects on cellular apoptosis, caspase activation and suppression of cell migration and invasion. J. BUON 2020, 25, 1383–1389. [Google Scholar] [PubMed]
  256. Han, Y.H.; Kee, J.Y.; Hong, S.H. Rosmarinic acid activates AMPK to inhibit metastasis of colorectal cancer. Front. Pharmacol. 2018, 9, 68. [Google Scholar] [CrossRef] [PubMed]
  257. Chen, S.G.; Leu, Y.L.; Cheng, M.L.; Ting, S.C.; Liu, C.C.; Wang, S.D.; Yang, C.H.; Hung, C.Y.; Sakurai, H.; Chen, K.H.; et al. Anti-enterovirus 71 activities of Melissa officinalis extract and its biologically active constituent rosmarinic acid. Sci. Rep. 2017, 7, 12264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Lin, W.Y.; Yu, Y.J.; Jinn, T.R. Evaluation of the virucidal effects of rosmarinic acid against enterovirus 71 infection via in vitro and in vivo study. Virol. J. 2019, 16, 94. [Google Scholar] [CrossRef]
  259. Hsieh, C.F.; Jheng, J.R.; Lin, G.H.; Chen, Y.L.; Ho, J.Y.; Liu, C.J.; Hsu, K.Y.; Chen, Y.S.; Chan, Y.K.F.; Yu, H.M.; et al. Rosmarinic acid exhibits broad anti-enterovirus A71 activity by inhibiting the interaction between the five-fold axis of capsid VP1 and cognate sulfated receptors. Emerg. Microbes. Infect. 2020, 9, 1194–1205. [Google Scholar] [CrossRef]
  260. Swarup, V.; Ghosh, J.; Ghosh, S.; Saxena, A.; Basu, A. Antiviral and anti-inflammatory effects of rosmarinic acid in an experimental murine model of Japanese encephalitis. Antimicrob. Agents Chemother. 2007, 51, 3367–3370. [Google Scholar] [CrossRef] [Green Version]
  261. Tsukamoto, Y.; Ikeda, S.; Uwai, K.; Taguchi, R.; Chayama, K.; Sakaguchi, T.; Narita, R.; Yao, W.L.; Takeuchi, F.; Otakaki, Y.; et al. Rosmarinic acid is a novel inhibitor for Hepatitis B virus replication targeting viral epsilon RNA-polymerase interaction. PLoS ONE 2018, 13, e0197664. [Google Scholar] [CrossRef] [Green Version]
  262. Mahalapbutr, P.; Sangkhawasi, M.; Kammarabutr, J.; Chamni, S.; Rungrotmongkol, T. Rosmarinic acid as a potent influenza neuraminidase inhibitor: In vitro and in silico study. Curr. Top. Med. Chem. 2020, 20, 2046–2055. [Google Scholar] [CrossRef] [PubMed]
  263. Ren, P.; Jiang, H.; Li, R.G.; Wang, J.; Song, N.; Xu, H.M.; Xie, J.X. Rosmarinic acid inhibits 6-OHDA-induced neurotoxicity by anti-oxidation in MES23.5 cells. J. Mol. Neurosci. 2009, 39, 220–225. [Google Scholar] [CrossRef] [PubMed]
  264. Wang, J.Y.; Xu, H.M.; Jiang, H.; Du, X.X.; Sun, P.; Xie, J.X. Neurorescue effect of rosmarinic acid on 6-hydroxydopamine-lesioned nigral dopamine neurons in rat model of Parkinson’s disease. J. Mol. Neurosci. 2012, 47, 113–119. [Google Scholar] [CrossRef] [PubMed]
  265. Gok, D.K.; Hidisoglu, E.; Ocak, G.A.; Er, H.; Acun, A.D.; Yargicoglu, P. Protective role of rosmarinic acid on amyloid beta 42-induced echoic memory decline: Implication of oxidative stress and cholinergic impairment. Neurochem. Int. 2018, 118, 1–13. [Google Scholar]
  266. Khamse, S.; Sadr, S.S.; Roghani, M.; Hasanzadeh, G.; Mohammadian, M. Rosmarinic acid exerts a neuroprotective effect in the kainate rat model of temporal lobe epilepsy: Underlying mechanisms. Pharm. Biol. 2015, 53, 1818–1825. [Google Scholar] [CrossRef] [PubMed]
  267. Fonteles, A.A.; de Souza, C.M.; de sousa, J.C.N.; Menezes, A.P.F.; do Carmo, M.R.S.; Fernandes, F.D.P.; de Araújo, P.R.; de Andrade, G.M. Rosmarinic acid prevents against memory deficits in ischemic mice. Behav. Brain Res. 2016, 297, 91–103. [Google Scholar] [CrossRef]
  268. Ma, Z.J.; Lu, Y.B.; Yang, F.G.; Li, S.P.; He, X.G.; Gao, Y.C.; Zhang, G.Z.; Ren, E.H.; Wang, Y.G.; Kang, X.W. Rosmarinic acid exerts a neuroprotective effect on spinal cord injury by suppressing oxidative stress and inflammation via modulating the Nrf2/HO-1 and TLR4/NF-κ B pathways. Toxicol. Appl. Pharmacol. 2020, 397, 115014. [Google Scholar] [CrossRef]
  269. Li, G.S.; Jiang, W.L.; Tian, J.W.; Qu, G.W.; Zhu, H.B.; Fu, F.H. In vitro and in vivo antifibrotic effects of rosmarinic acid on experimental liver fibrosis. Phytomedicine 2010, 17, 282–288. [Google Scholar] [CrossRef]
  270. Domitrovic, R.; Skoda, M.; Marchesi, V.V.; Cvijanovic, O.; Pugel, E.P.; Stefan, M.B. Rosmarinic acid ameliorates acute liver damage and fibrogenesis in carbon tetrachloride-intoxicated mice. Food Chem. Toxicol. 2013, 51, 370–378. [Google Scholar] [CrossRef]
  271. Lin, S.Y.; Wang, Y.Y.; Chen, W.Y.; Liao, S.L.; Chou, S.T.; Yang, C.P.; Chen, C.J. Hepatoprotective activities of rosmarinic acid against extrahepatic cholestasis in rats. Food Chem. Toxicol. 2017, 108, 214–223. [Google Scholar] [CrossRef]
  272. Lou, K.H.; Yang, M.; Duan, E.D.; Zhao, J.H.; Yu, C.; Zhang, R.P.; Zhang, L.C.; Zhang, M.; Xiao, Z.C.; Hu, W.Y.; et al. Rosmarinic acid stimulates liver regeneration through the mTOR pathway. Phytomedicine 2016, 23, 1574–1582. [Google Scholar] [CrossRef] [PubMed]
  273. Komeili-Movahhed, T.; Bassirian, M.; Changizi, Z.; Moslehi, A. SIRT1/NF κB pathway mediates anti-inflammatory and anti-apoptotic effects of rosmarinic acid on in a mouse model of nonalcoholic steatohepatitis (NASH). J. Recept. Signal Transduct. 2021. [Google Scholar] [CrossRef] [PubMed]
  274. Yao, Y.; Xu, Y.L.; Wang, Y. Protective roles and mechanisms of rosmarinic acid in cyclophosphamide-induced premature ovarian failure. J. Biochem. Mol. Toxicol. 2020, 34, e22591. [Google Scholar] [CrossRef] [PubMed]
  275. Zhang, T.T.; Ma, S.S.; Liu, C.; Hu, K.; Xu, M.; Wang, R.S. Rosmarinic acid prevents radiation-induced pulmonary fibrosis through attenuation of ROS/MYPT1/TGFβ1 signaling via miR-19b-3p. Dose-Response 2020, 18, 155932582096841. [Google Scholar] [CrossRef] [PubMed]
  276. Ji, R.P.; Sun, H.J.; Peng, J.Y.; Ma, X.D.; Bao, L.C.; Fu, Y.F.; Zhang, X.X.; Luo, C.X.; Gao, C.; Jin, Y.; et al. Rosmarinic acid exerts an antagonistic effect on vascular calcification by regulating the Nrf2 signaling pathway. Free Radic. Res. 2019, 53, 187–197. [Google Scholar] [CrossRef] [PubMed]
  277. Lee, J.S.; Jung, E.S.; Koh, J.; Kim, Y.S.; Park, D. Effect of rosmarinic acid on atopic dermatitis. J. Dermatol. 2008, 35, 768–771. [Google Scholar] [CrossRef]
  278. Osakabe, N.; Takano, H.; Sanbongi, C.; Yasuda, A.; Yanagisawa, R.; Inoue, K.; Yoshikawa, T. Anti-inflammatory and anti-allergic effect of rosmarinic acid (RA); inhibition of seasonal allergic rhinoconjunctivitis (SAR) and its mechanism. Biofactors 2004, 21, 127–131. [Google Scholar] [CrossRef]
  279. Wang, Y.; Liu, F.X.; Wang, W.Q.; Su, J.Y.; Xue, G.; Pi, Q. Application of rosemary acid in sea buckthorn fruit wine. China Brew. 2018, 37, 121–123. [Google Scholar]
  280. Li, Y.; Zhang, X.J.; Ma, L.; Guo, T.; Yu, Y.; Dai, H.J.; Zhou, H.Y.; Zhang, Y.H. Application of rabbit skin gelatin/rosmarinic acid composite film in pork quality preservation during cold storage. Food Sci. 2019, 40, 281–287. [Google Scholar]
  281. Li, N.; Mei, J.; Shen, Y.; Xie, J. Quality improvement of half-smooth tongue sole (Cynoglossus Semilaevis) fillets by chitosan coatings containing rosmarinic acid during storage. CYTA-J. Food 2018, 16, 1018–1029. [Google Scholar] [CrossRef] [Green Version]
  282. Guo, S.B.; Xu, L.L.; Jiang, L.J.; Wang, F.; Wang, Z.J.; Zhang, J.Y.; Liu, B. Profiling and identification of in vivo metabolism of rosmarinic acid in rats. China J. Chin. Mater. Med. 2019, 44, 4704–4712. [Google Scholar]
  283. Su, J.; Jia, F.Y.; Lu, J.J.; Chen, W.X.; Sun, H.; Liu, T.; Wu, X. Characterization of the metabolites of rosmarinic acid in human liver microsomes using liquid chromatography combined with electrospray ionization tandem mass spectrometry. Biomed. Chromatogr. 2020, 34, e4806. [Google Scholar] [CrossRef] [PubMed]
  284. Wang, J.X.; Li, G.Y.; Rui, T.Q.; Kang, A.; Li, G.C.; Fu, T.M.; Li, J.S.; Di, L.Q.; Cai, B.C. Pharmacokinetics of rosmarinic acid in rats by LC-MS/MS: Absolute bioavailability and dose proportionality. RSC Adv. 2017, 7, 9057–9063. [Google Scholar] [CrossRef] [Green Version]
  285. Min, J.B.; Chen, H.; Gong, Z.P.; Liu, X.; Wu, T.; Li, W.R.; Fang, J.S.; Huang, T.L.; Zhang, Y.F.; Zhao, W.; et al. Pharmacokinetic and pharmacodynamic properties of rosmarinic acid in rat cholestatic liver injury. Molecules 2018, 23, 2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Lu, P.; Xing, Y.; Xue, Z.F.; Ma, Z.; Zhang, B.; Peng, H.; Zhou, Q.; Liu, H.F.; Liu, Z.D.; Li, J.W. Pharmacokinetics of salvianolic acid B, rosmarinic acid and Danshensu in rat after pulmonary administration of Salvia miltiorrhiza polyphenolic acid solution. Biomed. Chromatogr. 2019, 33, e4561. [Google Scholar] [CrossRef]
  287. Yang, Y.M.; Ying, S.; Li, T.; Zhen, J.; Chen, D.M.; Wang, J.M. A sensitive LC-MS/MS-based bioanalytical method for quantification of salviaflaside and rosmarinic acid in rat plasma and its application in a pharmacokinetic study. Biomed. Chromatogr. 2018, 32, e4259. [Google Scholar] [CrossRef]
  288. Zhang, L.; Xu, H.Y.; Zhan, L.B. Pharmacokinetic assessments of liquiritin, protocatechuic aldehyde and rosmarinic acid in rat plasma by UPLC-MS-MS after administration of ZibuPiyin Recipe. J. Chromatogr. Sci. 2018, 56, 139–146. [Google Scholar] [CrossRef]
  289. Ouyang, H.Z.; He, J. Simultaneous determination of nine constituents of Xuebijing Injection in rat plasma and their pharmacokinetics by LC-MS/MS. China J. Chin. Mater. Med. 2018, 43, 3553–3561. [Google Scholar]
  290. Ozgun, G.S.; Ozgun, E. The cytotoxic concentration of rosmarinic acid increases MG132-induced cytotoxicity, proteasome inhibition, autophagy, cellular stresses, and apoptosis in HepG2 cells. Hum. Exp. Toxicol. 2019, 39, 514–523. [Google Scholar] [CrossRef]
  291. Ghaffari, H.; Venkataramana, M.; Ghassam, B.J.; Nayaka, S.C.; Nataraju, A.; Geetha, N.P.; Prakash, H.S. Rosmarinic acid mediated neuroprotective effects against H2O2-induced neuronal cell damage in N2A cells. Life Sci. 2014, 113, 7–13. [Google Scholar] [CrossRef]
  292. Costa, P.; Sarmento, B.; Gonçalves, S.; Romano, A. Protective effects of Lavandula viridis L’Hér extracts and rosmarinic acid against H2O2-induced oxidative damage in A172 human astrocyte cell line. Ind. Crops Prod. 2013, 50, 361–365. [Google Scholar] [CrossRef]
  293. Baba, S.; Osakabe, N.; Natsume, M.; Yasuda, A.; Muto, Y.; Hiyoshi, K.; Takano, H.; Yoshikawa, T.; Terao, J. Absorption, metabolism, degradation and urinary excretion of rosmarinic acid after intake of Perilla frutescens extract in humans. Eur. J. Nutr. 2005, 44, 1–9. [Google Scholar] [CrossRef] [PubMed]
  294. Noguchi-Shinohara, M.; Ono, K.; Hamaguchi, T.; Iwasa, K.; Nagai, T.; Kobayashi, S.; Nakamura, H.; Yamada, M. Pharmacokinetics, safety and tolerability of Melissa officinalis extract which contained rosmarinic acid in healthy individuals: A randomized controlled trial. PLoS ONE 2015, 10, e0126422. [Google Scholar] [CrossRef] [PubMed]
  295. Li, H.Z.; Ren, Z.Q.; Reddy, N.V.; Hou, T.Y.; Zhang, Z.J. In silico evaluation of antimicrobial, antihyaluronidase and bioavailability parameters of rosmarinic acid in Perilla frutescens leaf extracts. SN Appl. Sci. 2020, 2, 1547. [Google Scholar] [CrossRef]
  296. Baranauskaite, J.; Duman, G.; Corapcioglu, G.; Baranauskas, A.; Taralp, A.; Ivanauskas, L.; Bernatoniene, J. Liposomal incorporation to improve dissolution and stability of rosmarinic acid and carvacrol extracted from oregano (O. onites L.). Biomed. Res. Int. 2018, 2018, 6147315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. Madureira, A.R.; Campos, D.A.; Oliveira, A.; Sarmento, B.; Pintado, M.M.; Gomes, A.M. Insights into the protective role of solid lipid nanoparticles on rosmarinic acid bioactivity during exposure to simulated gastrointestinal conditions. Colloids Surf. B Biointerfaces 2016, 139, 277–284. [Google Scholar] [CrossRef]
Figure 1. The chemical structure of RA.
Figure 1. The chemical structure of RA.
Molecules 27 03292 g001
Figure 2. Biosynthesis pathway of RA in the plants.
Figure 2. Biosynthesis pathway of RA in the plants.
Molecules 27 03292 g002
Table 1. Plants containing RA.
Table 1. Plants containing RA.
No.PlantFamilyPartReference
1Adenium obesumApocynaceaeStem Barks[11]
2Alkanna sfikasiana Tan, Vold and StridBoraginaceaeRoots[12]
3Anchusa azurea Miller var. azureaBoraginaceaeRoots[13]
4Anchusa italica Retz.Boraginaceae-[14]
5Anchusa strigosa Banks et SolBoraginaceaeRoots[15]
6Anthoceros punctatusAnthocerotaceae-[16]
7Apeiba tibourbou Aubl.TiliaceaeLeaves[17]
8Arctopus monacanthusApiaceaeRoots[18]
9Arnebia purpurea S. Erik & H. SumbulBoraginaceaeRoots[19]
10Baccharis chilco KunthAsteraceaeAerial parts[20]
11Barbarea integrifoliaBrassicaceaeAerial parts[21]
12Bellis sylvestrisAsteraceaeLeaves[22]
13Blechnum brasilienseBlechnaceaeLeaves[23]
14Canna edulis KerCannceaeRhizomes[24]
15Celastrus hindsii BenthCelastraceaeLeaves[25]
16Centella asiaticaApiaceaeAerial parts[26]
17Chloranthus fortune (A. Gray) Solms-LaubChloranthaceaeWhole plants[27]
18Chloranthus multistachys PeiChloranthaceae-[28]
19Clerodendranthus spicatus (Thunb.) C.Y. WuLamiaceaeWhole plants[29]
Aerial parts[30]
20Clinopodium chinense var. parviflorumLamiaceaeAerial parts[31]
21Clinopodium tomentosum (Kunth) GovaertsLamiaceaeAerial parts[32]
22Clinopodium urticifoliumLamiaceaeWhole plants[33]
23Coleus aromaticus Benth.LamiaceaeLeaves[34]
24Coleus forskohlii (Willd) Briq.LamiaceaeWhole plants[35]
25Coleus parvifolius Benth.LamiaceaeAerial parts[36]
26Colocasia esculenta (L.) SchottAraceaeLeaves[37]
27Cordia alliodoraBoraginaceaeRoot barks[38]
28Cordia bicolorBoraginaceaeLeaves[39]
29Cordia boissieri A. DC.BoraginaceaeLeaves[40]
30Cordia dentataBoraginaceaeLeaves[39]
31Cordia latifolia Roxb.BoraginaceaeFruits[41]
32Cordia megalanthaBoraginaceaeLeaves[39]
33Cordia morelosana StandleyBoraginaceaeAerial parts[42]
34Cordia sinensisBoraginaceaeWhole plants[43]
35Cordia verbenaceaBoraginaceaeLeaves[44]
36Cynoglossum columnae Ten.BoraginaceaeRoots[45]
37Dracocephalum fruticulosum Steph. Ex Willd.LamiaceaeAerial parts[46]
38Dracocephalum heterophyllumLamiaceaeWhole plants[47]
39Dracocephalum nutans L.LamiaceaeAerial parts[46]
40Dracocephalum palmatum StephanLamiaceaeAerial parts[48]
41Dracocephalum tanguticum Maxim.LamiaceaeWhole plants[49]
42Ehretia asperulaBoraginaceaeLeaves[50]
43Ehretia obtusifoliaBoraginaceaeWhole plants[51]
44Ehretia philippinensisBoraginaceaeBarks[52]
45Ehretia thyrsifloraBoraginaceaeLeaves[53]
46Elsholtiza bodinieri VaniotLamiaceaeWhole plants[54]
47Elsholtzia rugulosa Hemsl.LamiaceaeAerial parts[55]
48Elsholtzia splendens NakaiLamiaceaeFlowers and leaves[56]
49Farfugium japonicum (L.) Kitam. Var. giganteum (Siebold et Zucc.) Kitam.AsteraceaeFlowers[57]
50Foeniculum vulgare Mill.ApiaceaeAerial parts[58]
51Forsythia koreana NakaiOleaceaeFruits[59]
52Gastrocotyle hispidaBoraginaceaeAerial parts[60]
53Glechoma longitubaLamiaceaeWhole plants[61]
54Hamelia patens Jacq.RubiaceaeAerial parts[62]
55Hedera helix L.Araliaceae-[63]
56Helicteres angustifolia Linn.SterculiaceaeRoots[64]
57Helicteres hirsuta LourSterculiaceaeStems[65]
58Helicteres isora L.SterculiaceaeFruits[66]
59Hypenia salzmannii (Benth.) HarleyLamiaceaeLeaves[67]
60Hyptis atrorubens Poit.LamiaceaeLeaves and stems[68]
61Hyptis capitata Jacq.LamiaceaeAerial parts[69]
62Hyptis pectinata (L.) PoitLamiaceaeLeaves[70]
63Hyptis suaveolens (L.) PoitLamiaceaeAerial parts[71]
64Hyptis verticillata Jacq.LamiaceaeAerial parts[72]
65Hyssopus cuspidatusLamiaceaeWhole plants[73]
66Ipomoea turpethum (L.) R.Br.ConvolvulaceaeWhole plants[74]
67Isodon eriocalyx (Dunn) Hara var. laxiflora C. Y. Wu et H. W. LiLamiaceaeLeaves[75]
68Isodon flexicaulis C. Y. Wu et H. W. LiLamiaceaeAerial parts[76]
69Isodon lophanthoides var. graciliflorusLamiaceaeAerial parts[77]
Leaves[78]
70Isodon oresbius (W. W. Smith) KudoLamiaceaeAerial parts[79]
71Isodon rubescens (Hemsl.) HaraLamiaceae-[80]
72Isodon rugosus (Wall. Ex Benth.) CoddLamiaceaeAerial parts[81]
73Isodon sculponeata (Vaniot) Hara.LamiaceaeLeaves[82]
74Keiskea japonica Miq.LamiaceaeAerial parts[83]
75Lallemantia iberica (Bieb.) Fisch & C.A. MeyLamiaceaeAerial parts[84]
76Lavandula angustifolia Mill.LamiaceaeAerial parts[85]
77Lepechinia graveolens (Reg.) Epling.Lamiaceae-[86]
78Lepechinia meyenii (Walp.) EplingLamiaceae-[87]
79Lepechinia speciosa (St. Hill) EplingLamiaceae-[88]
80Lycopus europaeus L.LamiaceaeWhole plants[89]
81Lycopus lucidus Turcz.LamiaceaeAerial parts[90]
82Marrubium vulgare L.LamiaceaeLeaves[91]
83Meehania urticifolia (Miq.) MakinoLamiaceaeWhole plants[92]
84Melissa officinalis L.LamiaceaeAerial parts[93]
Leaves[94]
85Mentha dumetorumLamiaceaeAerial parts[95]
86Mentha haplocalyx Briq.LamiaceaeAerial parts[96]
87Mentha longifolia (L.) Hudson subsp. longifoliaLamiaceaeAerial parts[97]
88Mentha piperita L.LamiaceaeLeaves[98]
Aerial parts[99]
89Mentha spicata L.LamiaceaeWhole plants[100]
90Mesona chinensis Benth.LamiaceaeWhole plants[101]
91Micromeria myrtifolia Boiss. & HohenLamiaceaeAerial parts[102]
92Microsorum fortunei (T. Moore) ChingPolypodiaceaeLeaves and stems[103]
93Momordica balsaminaCucurbitaceaeAerial parts[104]
94Nepeta asterotricha Rech. F.LamiaceaeAerial parts[105]
95Nepeta cadmea Boiss.LamiaceaeAerial parts[106]
96Nepeta curviflora Boiss.LamiaceaeAerial parts[107]
97Ocimum campechianum Mill.LamiaceaeLeaves[108]
98Ocimum sanctum Linn.LamiaceaeLeaves and stems[109]
99Origanum dictamnus L.LamiaceaeAerial parts[110]
100Origanum glandulosum DesfLamiaceaeAerial parts[111]
101Origanum majorana L.LamiaceaeAerial parts[112]
102Origanum minutiflorumLamiaceaeAerial parts[113]
103Origanum rotundifolium Boiss.LamiaceaeAerial parts[114]
104Origanum vulgare L. ssp. HirtumLamiaceaeAerial parts[115]
105Paris veriticillata Bieb.LiliaceaeRoots[116]
106Perilla frutescens (L.) Britton var. acuta KudoLamiaceaeLeaves[117]
Seeds[118]
107Perilla frutescens var. acutaLamiaceaeFruits[119]
108Perovskia atriplicifolia Benth.LamiaceaeRoots[120]
109Plectranthus forsteri ‘Marginatus’LamiaceaeAerial parts[121]
110Plectranthus hadiensis var. tomentosusLamiaceaeAerial parts[122]
111Plectranthus madagascariensis (Pers.) BenthLamiaceaeAerial parts[123]
112Plectranthus scutellarioides (L.) R. Br.LamiaceaeAerial parts[124]
113Polygomun aviculanePolygonaceaeAerial parts[125]
114Prunella vulgaris L.LamiaceaeSpikes[126]
115Prunella vulgaris var. lilacinaLamiaceaeSpikes[127]
Aerial parts[128]
116Quercus serrata MurrayFagaceaeLeaves[129]
117Rosmarinus officinalis L.LamiaceaeSprigs[130]
Leaves[131]
118Salvia absconditiflora Greuter & BurdetLamiaceaeAerial parts[132]
119Salvia castanea Diels f. tomentosa Stib.LamiaceaeRhizomes[133]
120Salvia cavaleriei Levi.LamiaceaeWhole plants[134]
121Salvia cerino-pruinosa Rech. F. var. cerino-pruinosaLamiaceaeAerial parts[135]
122Salvia chinensis Benth.LamiaceaeAerial parts[136]
Whole plants[137]
123Salvia deserta SchangLamiaceaeRoots[138]
Flowers[139]
124Salvia flava ForrestLamiaceaeWhole plants[140]
125Salvia grandifolia W. W. SmithLamiaceaeRoots[141]
126Salvia kiaometiensis Lévl.LamiaceaeRoots[142]
127Salvia limbata C.A. MeyerLamiaceaeAerial parts[143]
128Salvia miltiorrhiza BungeLamiaceaeLeaves[144]
Roots[145]
129Salvia officinalisLamiaceae-[146]
130Salvia palaestina BenthamLamiaceaeAerial parts[147]
131Salvia plebeia R. Br.LamiaceaeLeaves[148]
Whole plants[149]
132Salvia przewalskii MaximLamiaceaeRoots and rhizomes[150]
Roots[151]
133Salvia sonchifolia C.Y. WuLamiaceaeRoots[152]
134Salvia splendens Sellow ex Roem & SchultLamiaceaeLeaves[153]
135Salvia trichoclada BenthamLamiaceaeWhole plants[154]
136Salvia viridis L. cvar. Blue JeansLamiaceaeAerial parts[155]
137Salvia yunaansisLamiaceaeRoots[156]
138Sanicula europaea L.ApiaceaeAerial parts[157]
139Sanicula lamelligera HanceApiaceaeWhole plants[158]
140Sarcandra glabra (Thunb.) Nakai.ChloranthaceaeWhole plants[159]
141Satureja bifloraLamiaceaeAerial parts[160]
142Schizonepeta tenuifolia BriquetLamiaceaeAerial parts[161]
143Sideritis albifloraLamiaceaeAerial parts[162]
144Sideritis leptocladaLamiaceaeAerial parts[162]
145Solanum betaceum Cav.SolanaceaeFruits[163]
146Solenostemon monostachys BriqLamiaceaeAerial parts[164]
147Symphytum officinale L.BoraginaceaeRoots[165]
148Thunbergia laurifolia LindlAcanthaceaeLeaves[166]
149Thymus alternans KlokovLamiaceaeAerial parts[167]
150Thymus atlanticus (Ball) RoussineLamiaceaeLeaves[168]
151Thymus praecox subsp grossheimii (Ronniger) JalasLamiaceaeAerial parts[169]
152Thymus praecox subsp grossheimii (Ronniger) Jalas var. grossheimiiLamiaceaeAerial parts[170]
153Thymus quinquecostatus var. japonicaLamiaceaeAerial parts[171]
154Thymus serpyllumLamiaceaeWhole plants[172]
155Thymus sibthorpii BenthamLamiaceaeAerial parts[173]
156Thymus sipyleus subsp. Sipyleus var. sipyleusLamiaceaeAerial parts[174]
157Thymus vulgaris L.LamiaceaeLeaves[175]
158Tournefortia sarmentosa Lam.BoraginaceaeStems[176]
159Veronica sibirica L.PennellScrophulariaceaeRhizomes[177]
160Ziziphora clinopodioides Lam.LamiaceaeAerial parts[178]
161Zostera marinaPotamogetonaceaeLeaves[179]
162Zostera noltiiPotamogetonaceaeLeaves[180]
-: not mentioned.
Table 2. The pharmacokinetic characteristics of RA in different test drugs and animals.
Table 2. The pharmacokinetic characteristics of RA in different test drugs and animals.
No.DrugAnimalAdministration ModePharmacokinetic CharactersReference
1RANormal ratsIntragastrical administration,
12.5 mg/kg
AUC(0∞) = 866.51 ng/mL·h, Tmax = 0.139 h,
Cmax = 215.21 ng/mL, CL = 15.00 L/(h·kg)
[284]
Intragastrical administration,
25 mg/kg
AUC(0∞) = 1308.62 ng/mL·h, Tmax = 0.181 h,
Cmax = 361.57 ng/mL, CL = 19.20 L/(h·kg)
Intragastrical administration,
50 mg/kg
AUC(0∞) = 1866.58 ng/mL·h, Tmax = 0.306 h,
Cmax = 790.96 ng/mL, CL = 27.60 L/(h·kg)
Intravenous administration,
0.625 mg/kg
AUC(0∞) = 2556.14 ng/mL·h,
Cmax = 6166.89 ng/mL, CL = 6.00 L/(h·kg)
2RACholestatic liver injured ratsIntragastrical administration,
100 mg/kg
AUC(0∞) = 23.984 mg/mL·h, Tmax = 0.988 h,
Cmax = 2.876 mg/mL, CL = 4.169 L/(h·kg)
[285]
Normal ratsAUC(0∞) = 20.500 mg/mL·h, Tmax = 0.704 h,
Cmax = 2.542 mg/mL, CL = 4.876 L/(h·kg)
3Salvia miltiorrhiza
polyphenolic acid solution
Normal ratsPulmonary administration,
10 mg/kg
AUC(0∞) = 200.01 ng/mL·h, Tmax = 0.07 h,
Cmax = 370.78 ng/mL, CL = 0.05 L/(h·kg)
[286]
Intravenous administration,
10 mg/kg
AUC(0∞) = 209.34 ng/mL·h, Tmax = 0.03 h,
Cmax = 1344.10 ng/mL, CL = 0.05 L/(h·kg)
4Prunella vulgaris extractNormal ratsIntragastrical administration,
10 mL/kg (1.25 mg/mL for RA)
AUC(0∞) = 737.7 ng/mL·h, Tmax = 1.5 h,
Cmax = 120.8 ng/mL, CL = 21.0 L/(h·kg)
[287]
5ZibuPiyin RecipeNormal ratsIntragastrical administration,
3.951 g/kg (0.03 mg/g for RA)
AUC(0∞) = 3099.4 μg/mL·h, Tmax = 1.7 h,
Cmax = 222.7 ng/mL
[288]
6Xuebijing InjectionNormal ratsIntravenous administration,
6 mL/kg (12.56 μg/mL for RA)
AUC(0∞) = 4.10 ng/mL·h, Tmax = 0.08 h,
Cmax = 173.19 ng/mL
[289]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Guan, H.; Luo, W.; Bao, B.; Cao, Y.; Cheng, F.; Yu, S.; Fan, Q.; Zhang, L.; Wu, Q.; Shan, M. A Comprehensive Review of Rosmarinic Acid: From Phytochemistry to Pharmacology and Its New Insight. Molecules 2022, 27, 3292. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27103292

AMA Style

Guan H, Luo W, Bao B, Cao Y, Cheng F, Yu S, Fan Q, Zhang L, Wu Q, Shan M. A Comprehensive Review of Rosmarinic Acid: From Phytochemistry to Pharmacology and Its New Insight. Molecules. 2022; 27(10):3292. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27103292

Chicago/Turabian Style

Guan, Huaquan, Wenbin Luo, Beihua Bao, Yudan Cao, Fangfang Cheng, Sheng Yu, Qiaoling Fan, Li Zhang, Qinan Wu, and Mingqiu Shan. 2022. "A Comprehensive Review of Rosmarinic Acid: From Phytochemistry to Pharmacology and Its New Insight" Molecules 27, no. 10: 3292. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27103292

Article Metrics

Back to TopTop