Next Article in Journal
Self-Assembled Matrine-PROTAC Encapsulating Zinc(II) Phthalocyanine with GSH-Depletion-Enhanced ROS Generation for Cancer Therapy
Previous Article in Journal
Emulsion Liquid Membranes Based on Os–NP/n–Decanol or n–Dodecanol Nanodispersions for p–Nitrophenol Reduction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Tumor Effects and Toxicity Reduction Mechanisms of Prunella vulgaris: A Comprehensive Review

1
College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
2
Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
3
College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, China
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(8), 1843; https://doi.org/10.3390/molecules29081843
Submission received: 20 March 2024 / Revised: 12 April 2024 / Accepted: 16 April 2024 / Published: 18 April 2024

Abstract

:
Purpose: To investigate and systematically describe the mechanism of action of Prunella vulgaris (P. vulgaris) against digestive system tumors and related toxicity reduction. Methods: This study briefly describes the history of medicinal food and the pharmacological effects of P. vulgaris, focusing on the review of the anti-digestive tumor effects of the active ingredients of P. vulgaris and the mechanism of its toxicity reduction. Results: The active ingredients of P. vulgaris may exert anti-tumor effects by inducing the apoptosis of cancer cells, inhibiting angiogenesis, inhibiting the migration and invasion of tumor cells, and inhibiting autophagy. In addition, P. vulgaris active ingredients inhibit the release of inflammatory factors and macrophages and increase the level of indicators of oxidative stress through the modulation of target genes in the pathway to achieve the effect of toxicity reduction. Conclusion: The active ingredients in the medicine food homology plant P. vulgaris not only treat digestive system tumors through different mechanisms but also reduce the toxic effects. P. vulgaris is worthy of being explored more deeply.

Graphical Abstract

1. Introduction

Digestive system carcinoma, including esophageal carcinoma, colorectal carcinoma, gastric carcinoma, pancreatic carcinoma, gallbladder carcinoma, liver carcinoma and so on, have a high incidence and mortality [1]. While significant advancements in medical technology have made great progress in cancer treatment, enhancing cancer survival rates and prognostic efficacy remains a formidable challenge [2]. Conventional treatment methods such as surgery, radiotherapy and targeted therapy all cause greater toxic side effects. Chinese medicine has a systematic theory and practical history in cancer and has better therapeutic effects while also reducing the toxic side effects caused by radiotherapy, making it a promising clinical treatment method for tumors [3].
Gastric carcinoma is the third deadliest type of cancer [4]. Its pathogenesis is complex and closely related to age, high salt intake and a lack of fruit and vegetables. Helicobacter pylori infection is the main cause of gastric cancer, which has declined with improved food preservation techniques and treatment of Helicobacter pylori, but poor prognosis has continued to increase the rate of gastric cancer [5]. Liver carcinoma typically arises from underlying diseases, mainly hepatitis B virus, fatty liver, poor diet and fungal toxins [6], and it is the only one of the grievous carcinoma whose incidence is increasing every year [7]. Its prognosis is poor, with only 5–15% of early-stage patients being suitable for surgical resection, usually without cirrhosis due to the reduced regenerative capacity of the liver, and the efficacy of treatment with kinase inhibitors in patients with advanced disease is extremely low [8]. Colorectal carcinoma is a common malignancy of adenocarcinoma origin, whose incidence increases with age; in about 80% of cases, patients are diagnosed with localized tumors, for which the causative factors are mainly age and family history. Surgery is the main treatment for localized disease, while post-operative chemotherapy serves to reduce recurrence, especially if the lesion has spread to the lymph nodes. Notably, a combination of chemo-radiotherapy serves as the main therapeutic approach for rectal carcinoma other than surgery and is often administered preoperatively [9]. Gallbladder carcinoma is usually diagnosed late in the advanced stages, characterized by substantial tumor size leading to obstruction of and infiltration into adjacent tissues and organs. In most cases, diagnosis occurs incidentally during surgical intervention for gallstones, highlighting the lack of routine screening or pro-phase diagnostic protocols for early detection unlike several other cancers [10]. Despite the qualitative progress in the index of existence for other carcinomas, the index of existence for pancreatic carcinoma has been almost unchanged since the 1960s. The disease is often diagnosed at a late stage, with many treatment methods exhibiting limited efficacy, resulting in a discouraging prognosis, with influencing factors that are strongly correlated with genetics, obesity, alcohol and diabetes, among others [11]. Esophageal carcinoma has an increased lethality ratio. The main factors include smoking, alcohol consumption and pancreatic dysphagia, with simple dysphagia or unexpected weight loss being the most common first symptoms [12].
Indigenous cultures around the world have long used traditional herbal medicine to treat a myriad of ailments, with plants gaining recognition for their therapeutic properties, and this proliferation has produced highly effective medicines for widespread clinical use, including many plant natural products and derived analogues, among others [13]. The discovery and development of natural complexes, valid constituents and formulations in herbal medicines with the option for a cure that can prioritize the destruction of tumor cells and suppress carcinoma proliferation, as well as toxicity reduction, is a critical field of carcinoma remedy. Cancer is a multifactorial and multistep disease, so the therapeutic effect of combined herbal formulas with multiple targets and signaling pathways on tumors may be superior to that of drugs acting on a single molecular target [14]. Studies have indicated that edible plants can decrease the probability of illness, especially cancer. China has explored a variety of traditional Chinese medicines through long-term practice in history and summarized their application to different diseases [15]. Some traditional Chinese medicines can be used for both medicine and food, that is, medicine and food are homologous [16]. Its therapeutic effect is due to the effective chemical constituents of plants [17]. Modern pharmacological studies have shown that the broad pharmacological effects of P. vulgaris mainly contain anti-inflammatory, antioxidation, anti-hypertension, anti-hyperlipidemia, immune regulation, anti-virus and anti-carcinoma functions [18]. It also has a crucial impact on the fight against digestive system tumors. It can be used to prevent and treat gastric carcinoma [19], liver carcinoma [20], colorectal carcinoma [21], pancreatic carcinoma [22], gallbladder carcinoma, esophageal carcinoma and other malignant tumors [23].
P. vulgaris, commonly known as “Xiakucao” in China, is a perennial herb. P. vulgaris belongs to the Lamiaceae family [24]. P. vulgaris is a highly valuable medicinal plant that is widely distributed and adapts to different geographic environments, such as China, Korea, Japan and Europe [25]. It has been shown to be used to treat sore throats and promote wound healing [26]. P. vulgaris has been widely recognized for its various active ingredients and remarkable pharmacological effects, showing great potential for clinical applications [27]. This paper provides a brief overview of the history and pharmacological effects of P. vulgaris. It also systematically reviews the active ingredients in relation to digestive system tumors and discusses mechanisms for reducing toxicity. As a plant with both medicinal and culinary uses, P. vulgaris shows promise for further development.

2. Methodology

Firstly, the keywords “P. vulgaris”, “digestive system tumors” and “detoxification effect” were used in PubMed, Google Scholar, ScienceDirect, Scopus and Web of Science databases, and the search time was set to within 5 years. It was found that there was no review on the treatment of digestive system tumors with P. vulgaris in the past five years, and only a few articles were on the direction of tumors. The whole article only reviewed breast neoplasm, thyroid neoplasm, liver neoplasm, lung neoplasm or colon neoplasm. Secondly, the keywords “P. vulgaris”, “active ingredient”, “carcinoma”, “neoplasm”, “cancer” and “toxicity” were used to search the literature within 20 years. Finally, “P. vulgaris”, “oleanolic Acid”, “stigmasterol”, “luteolin”, “rosmarinic Acid”, “gastric cancer”, “liver cancer”, “esophageal cancer”, “pancreatic cancer”, “gallbladder cancer” and “colorectal cancer” were used as keywords, and we set the search time to be within 20 years. We systematically reviewed the intervention of active components of P. vulgaris in digestive system tumors, including not only different types of cancers in the digestive system but also the corresponding targets, signal axes and tumor phenotypes of each cancer type. The literature retrieval process is shown in Figure 1.

3. History of Medicine and Food

As the earliest record of medicinal use, P. vulgaris was found in the “Shen Nong’s Herbal Classic” of the Han Dynasty. P. vulgaris is bitter, pungent and cold in nature. It has many effects [28], such as “main heat scrofula, rat fistula, head trauma, broken disease, scattered galls, knot gas, swollen feet, wet arthralgia, clearing liver and gallbladder”. P. vulgaris was first recorded to be edible in the Song Dynasty “Bencao Yanyi”: “P. vulgaris... When the young leaves are used as vegetables, they must be washed to remove bitter water”. It is worth mentioning that Yao Kecheng compiled the “Food Materia Medica” in the Ming Dynasty. This collection is a monograph on diet conditioning and food treatment before the Ming Dynasty in China. P. vulgaris is also included: “P. vulgaris, bitter, cold, non-toxic... The tender seedlings are reduced, the bitterness is soaked, and the oil and salt are mixed to make the crispy and beautiful”. Li Shizhen made a detailed record of the medicinal value of P. vulgaris in the “Compendium of Materia Medica”. At the same time, he also explained the eating suggestion of P. vulgaris: “the young seedlings are reduced, the bitter taste is soaked, and the oil and salt are edible”. It can be seen that P. vulgaris has been widely consumed as food and vegetables as early as thousands of years ago [29]. P. vulgaris has the effects of detumescence and relieving heat. As a drug, it is also gradually integrated into the upsurge in traditional Chinese medicine health preservation. It has been developed into P. vulgaris tea, P. vulgaris porridge, cool P. vulgaris and so on [30]. P. vulgaris has a variety of dosage forms for clinical application, such as tablets, ointments, capsules, granules, etc. However, due to its different extraction methods and preparation processes, its clinical efficacy is also different. It has been proven to have good application prospects and research value in breast, thyroid, tumor and other diseases [31].

4. Active Ingredient Related Target Pathway Network Diagram

The effective ingredients of P. vulgaris were searched using TCMSP (https://old.tcmsp-e.com/tcmsp.php), accessed on 15 May 2023. With oral bioavailability (OB) ≥ 30% and drug-like properties (DL) ≥ 0.18 set as the select range, the effective ingredients and their relevant target proteins with high activity were screened, the objective protein designations were entered into UniProt (https://www.uniprot.org/), accessed on 15 May 2023 to gain the corresponding target gene designations, and all the obtained gene names were merged and deduplicated. The predicted targets of P. vulgaris were embed into David (https://david.ncifcrf.gov/tools.jsp), accessed on 15 May 2023 for Kyoto Encyclopedia of Genes and Genomes (KEGG) channel enrichment analysis. The top 30 signaling channels were screened according to the p-value. According to the screening conditions of the p-value and Count value, the enrichment analysis results were visualized in the form of a histogram by using the micro-bioinformatics online platform. The results showed that the number of genes enriched to pathways in cancer was the largest, and the p-value was the most significant. The screened active components of P. vulgaris and their corresponding target genes were used to establish an effective component–drug–target network analysis diagram through Cytoscape 3.9.1 software. Target acquisition and related pathway screening of P. vulgaris is shown in Figure 2.

5. Active Ingredient

Various bioactive components were identified in the extract of P. vulgaris [32]. The main active components were triterpenes [33], saponins [34], sterols, flavonoids [35], phenylpropanoids [18], polysaccharides [36] and volatile oils.

5.1. Triterpenes and Their Saponins

A total of 28 triterpenoids were isolated from P. vulgaris, including 20 triterpenoid sapogenins and 8 saponins [37]. The highest content was oleanolic acid, which was significantly correlated with the pharmacological effects of P. vulgaris. In nature, the compound exists in the structure of a free acid or a triterpenoid saponin glycoside ligand precursor, where it can be combined with sugar chains [38]. The existence of α-l-rhamnose residues at the C-3\28 ends of oleanolic acid double-stranded glycosides is important for enhancing cytotoxicity, and the introduction of further sugar at C3-OH and C-28 carboxylic acid is an advantageous decoration to improve defense against tumor function [39].

5.2. Sterols

The sterol compounds in P. vulgaris mainly include β-sitosterol, stigmasterol and ∆7-stigmasterol. The structure of plant sterols is similar to that of cholesterol. A double bond consisting of three cyclohexane rings and one cyclopentane ring was found at the C5-6 position. It has been indicated that phytosterols can induce apoptosis in some carcinoma cell lines and exert anticancer activity [40].

5.3. Flavonoids

P. vulgaris also includes luteolin, iso-orientin and luteoloside. Luteolin is a tetrahydroxy flavonoid compound composed of a carbon structure and two benzene rings connected by heterocycles. It mainly exists in the form of glycoside ligands or glycosides. Luteolin has been found to have the external and internal mechanisms of apoptotic cell mortality in carcinoma. The inhibition of neoplasm cell metastasis and angiogenesis is a potential influence of luteolin and has the possibility of reversing the drug resistance of neoplasm cells [41].

5.4. Phenylpropanoids

Phenylpropanoids in P. vulgaris are cis- and trans-caffeic acids and rosmarinic acid [42]. Among them, rosmarinic acid has been studied more. In industry, rosmarinic acid can be manufactured via esterification concerning 3,4-dihydroxyphenyllactic acid and caffeic acid [43], which has been proven to have a strong anti-tumor role [44]. The structural formula of the active components of P. vulgaris is shown in Figure 3.

6. Pharmacological Actions

P. vulgaris is a plant source of a variety of medicinal active ingredients [45], and its wide range of uses can treat various diseases, including cancer. It has anti-inflammatory effect. The wound-healing effect of the chemical compound of P. vulgaris in rats and mice was studied via wound models. The experimentation studies have indicated its remarkable ability to heal a wound and its anti-inflammatory efficacy [46]; antibacterial effect studies have shown that P. vulgaris extract has antibacterial activity against Escherichia coli in patients with urinary tract infection [47], as P. vulgaris extract is a natural antioxidant that restores the level of the oxidation index and has significant antioxidant and hepatoprotective activities [48]; and studies of anti-hypertensive effects have shown that these effects can significantly reduce systolic blood pressure and blood pressure in rats, with the anti-hypertensive effect having a dose–effect relationship. The acute and chronic hypoglycemic effects of P. vulgaris in type I diabetes were evaluated in a mouse model, and studies showed that serum insulin in mice increased and α-amylase and α-glucosidase decreased under the intervention of P. vulgaris; therefore, the active ingredients in P. vulgaris may be potential drugs to improve type I diabetes and related complications [49]. The effect of anti-hyperlipidemia was evaluated via metabolomics in high-fat rats, and the results showed that P. vulgaris significantly reduced the specific gravity of the body, Total Cholesterol, low-density lipoprotein, high-density lipoprotein, malondialdehyde and abdominal fat mass in rats; increased glutathione peroxidase activity; and improved lipid metabolism disorder in rats to interfere with hyperlipidemia [50]. For the immunomodulatory effect, the heteropolysaccharide isolated from the fruit cluster of P. vulgaris showed stable immune activity in further research and could be used as an effective immunomodulator in functional food and pharmacology [51]. Regarding antiviral effects, P. vulgaris extract can enhance the activity of monoclonal antibodies against Ebola virus and provide new ideas for the development of anti-Ebola virus infection [52]. Regarding anti-tumor effects, P. vulgaris inhibits the metastasis of various tumor cells and promotes apoptosis through different pathways [53]. Studies have shown that the P. vulgaris polysaccharide complex effectively inhibits the progression of liver carcinoma cells by promoting apoptosis, manifested as cell cycle arrest, chromatin condensation and morphological changes [54]. P. vulgaris can regulate various signaling pathways and, thus, improve the microenvironment of liver carcinoma metastasis, so it has the therapeutic potential to suppress the migration and invasion of hepatocellular carcinoma cells [55]. Colon carcinoma cells treated with extracts from the spicate plants of P. vulgaris showed a remarkable decrease in cell growth activity [21]. The extracts from P. vulgaris inhibited the proliferation of gastric cancer cells by increasing the levels of BAX and decreasing Bcl-2 levels [19] and suppressed the proliferation of gastric carcinoma by downregulating vascular endothelial growth factor (VEGF) expression [56].

7. Anti-Tumor Effect of Active Ingredients

The anti-tumor mechanisms of P. vulgaris include inhibiting cell proliferation, promoting carcinoma cell apoptosis, promoting autophagy, adjusting cell cyclin, suppressing vascular endothelial growth against angiogenesis and restraining carcinoma cell migration and invasion [23]. Types and Phenotypes of P. vulgaris against digestive system tumors is shown in Figure 4.

7.1. Oleanolic Acid

As one of the pentacyclic triterpenoids, oleanolic acid has positive anti-tumor activity. The mechanism comprises the following aspects: the suppression of cell proliferation, promotion of apoptosis, facilitation of autophagy, regulation of cell cyclin, suppression of vascular endothelial growth, suppression of angiogenesis, restriction of tumor cell migration and infiltration, etc. [57]. Oleanolic acid has been shown to inhibit a variety of molecular targets that play a crucial function in the progression of carcinoma. In recent years, several synthetic oleanolic acid derivatives have been synthesized, which showed strong anti-tumor properties [58]. The suppression of cell dynamism by OA is caused though the apoptosis of gastric carcinoma cells and the S-stage block of SGC7901, thereby restraining gastric carcinoma cells by suppressing the Protein kinase-B (Akt)/Nuclear factor kappa-B (NF-κB) channel [59]. Oleanolic acid promotes G2-/M-stage stagnation and the apoptosis of hepatocellular neoplasm cells. By diminishing the level of cyclin B1 and regulating the phosphorylation of protein kinase Chk1 and p21, the level of Bcl-2 was significantly reduced, and the BAX, cytoplasmic Cytochrome-C (Cyt c), Caspase-9 and Caspase-3 levels were increased to produce an anti-tumor effect [60]. The combination of oleanolic acid and 5-FU can collaboratively enhance the cell death of pancreatic neoplasm cells and increase the pro-apoptotic effect. Further studies have shown that combination therapy can strengthen mitochondrial depolarization, lysosomal membrane permeability and cathepsin D percolation and reduce the levels of Bcl-2 and survivin [61].

7.2. Stigmasterol

Stigmasterol has anti-neoplasmic role by boosting neoplasmic cell apoptosis; restraining growth, metastasis and invasion; and facilitating tumor cell autophagy against carcinoma effects on a variety of malignant tumors. According to mechanism studies, stigmasterol causes neoplasm cell apoptosis by inhibiting the Phosphatidylinositol 3-kinase (PI3K)/AKT pathway and producing mitochondrial reactive oxygen species. Its active role principally depends on its regulation of cyclin, and there are many potential influences on the regulation of cellular activities [62]. Stigmasterol inhibited the growth of gastric cancer cells. Its anti-proliferative effect was due to the induction of mitochondrial-mediated apoptosis, a diminution of BAX and elevation of Bcl-2 levels. It was further confirmed that stigmasterol also suppressed neoplasm cell migration and facilitated G2/M cell cycle stagnation. In addition, stigmasterol can regulate the JAK/STAT channel [63] to exert anticancer activity. Research has shown that stigmasterol inhibits lung neoplasm cell reproduction and promotes apoptosis. By directly targeting stigmasterol in lung cancer by RAR-related orphan receptor C (RORC), RORC overexpression reverses the inhibitory effect of stigmasterol on lung cancer [64].

7.3. Luteolin

Luteolin is a naturally occurring polyphenolic compound known to have antioxidant and anti-tumor activities in vitro. It is regarded as an anti-neoplasmic active ingredient to cure various forms of malignant tumors, such as lung carcinoma, gastric carcinoma, colon carcinoma and pancreatic carcinoma. It also prevents the progression of cancer by inhibiting the proliferation of carcinoma cells, protecting against neoplastic irritation, blocking the cell cycle and boosting apoptosis via diverse channels. In addition, it increases the levels of intracellular reactive oxygen species (ROS), PKR-like endoplasmic reticulum kinase (PERK), C/Ebp-Homologous Protein (CHOP), recombinant activating transcription factor 4 (ATF4) and Caspase 12 in glioblastoma cells via the activation of the lethal endoplasmic reticulum stress response and mitochondrial dysfunction and through the activation of endoplasmic reticulum stress-related protein reflection [65]. Luteolin led to the inactivation of cyclin B, Cell-division-cycle kinase 2 (CDC2), Caspase-9 and other proteins in the mouse colon HCT-15 cell line model; upregulated apoptotic protease activating factor-1 (APAF-1), Cyt.C, caspase-9/3 and cyclin-dependent kinase (CDK); arrested the G2/M stage of the cell cycle; and ultimately led to apoptosis and exerted anticancer activity [66].

7.4. Rosmarinic Acid

Rosmarinic acid is a common labiatae plant, and its rich nutritional properties are used to improve health. Studies have found that rosmarinic acid can prevent tumorigenesis, inhibit tumor growth and act as an adjuvant therapy to treat sensitization caused by radiotherapy and chemotherapy drugs [67]. In vivo experiments on mouse colon cancer indicated that rosmarinic acid significantly restrained the progression of cancer cells by inhibiting NF-κB and STAT3 [68]. Studies have shown that rosmarinic acid can inhibit NF-κB p65 in the xenotransplantation microenvironment and effectively inhibit the progression of liver carcinoma cells by regulating the secretion of inflammation- and angiogenesis-related factors [69]. The anti-gastric cancer activity of anti-MUC1 and rosmarinic acid is reflected in the inhibition of mRNA expression of enzymes that produce cancer-associated antigens and specific antigens; the promotion of BAX, Bad and Caspase-3 and -9; and inhibition of Bcl-2 expression [70]. Studies have shown that rosmarinic acid blocks the G2/S cycle in pancreatic carcinoma cells by adjusting the communication of cyclin E, BAX and Bcl-2; inhibits the migration and invasion of pancreatic cancer cells through E-cadherin and Matrix metallopeptidase 9 (MMP-9); and inhibits tumor growth [71]. Possible mechanism of P. vulgaris against digestive system tumors is shown in Figure 5 and Table 1.

8. The Attenuated Effect of Active Ingredients

Other active ingredients in P. vulgaris also have protective effects on liver, kidney and lung injury.
Oleanolic acid can reduce liver ischemia-reperfusion injury. The protective mechanism is correlated to the Heme oxygenase-1 (HO-1)/Recombinant Sestrin 2 (Sesn2) channel induced by oleanolic acid, which upregulates Sesn2, PI3K, Akt and HO-1 in the liver and protects the liver [90]. Stigmasterol ameliorates neuroinflammation in mice and inhibits microglial inflammation by relying on AMP-activated protein kinase (AMPK)/NF-κB and AMPK/NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) oligomers. Its function also inhibits neuroinflammation by diminishing proinflammatory element levels and microglial sensitization [91]. Luteolin is a strong anti-inflammatory agent that inhibits NF-κB, Mitogen-activated protein kinase (MAPK), Interleukin 6 (IL-6), Interleukin 8 (IL-8) and TNF-α and reduces ROS content. Luteolin can chelate transition metal ions responsible for ROS production, thereby inhibiting lipoxygenase. It has been proven that the suppression of NF-κB channel by luteolin stimulates the expression of inducible proinflammatory enzymes such as inducible nitric oxide synthase (iNOS), cyclo-oxygenase-2 (COX-2) and IL-1β, IL-6 and TNF-α [92]. Rosmarinic acid can remove active superoxide and peroxynitrite; reduce hepatotoxicity; inhibit the proliferation of hepatic stellate cells; downregulate the activity of transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF) and α-Smooth muscle actin (α-SMA); and improve liver tissue indexes [93]. Possible mechanisms of P. vulgaris that reduce toxicity is shown in Figure 6 and Table 2.

9. Discussion

P. vulgaris is a traditional Chinese medicine that is rich in nutrients and active ingredients as a medicinal and edible plant [105]. P. vulgaris not only acts as a cure for diseases but can also be consumed as a nutrient [106]. The active components in P. vulgaris plants have extensive biological functions, including anti-inflammatory, antioxidation, anti-hypertension, anti-hyperlipidemia, immunomodulation, anti-virus and anti-tumor. Cancer is a malignant disease that causes human death. The extremely high morbidity and fatality rate consumes a lot of medical resources and is a huge social and economic burden [107]. The active ingredients of P. vulgaris especially play an active role in the treatment of digestive system tumors, including stomach, liver, colorectal, gallbladder, pancreatic, esophageal and other malignant tumors. Oleanolic acid, stigmasterol, luteolin and rosmarinic acid in P. vulgaris can effectively inhibit tumor progression, and the possible mechanism is to inhibit tumor progression by regulating the expression of target genes, including inducing the apoptosis of cancer cells, inhibiting angiogenesis, inhibiting the migration and invasion of tumor cells, and inhibiting cellular autophagy.
The active ingredients of P. vulgaris Cao not only have significant efficacy as anti-tumor agents but also reduce liver and kidney damage and play a protective role. The possible mechanisms of its attenuating action are regulating the expression of target genes in the mitochondrial apoptotic pathway to inhibit apoptosis, inhibiting the release of inflammatory factors and macrophage activation by inhibiting the NF-κB signaling pathway and Nrf2 signaling pathway, and, on the contrary, increasing the levels of oxidative stress indicators such as GSH, SOD, CAT, TAC, GST and so on. Surgery, radiotherapy and chemotherapy are effective ways to treat tumors, but they are often accompanied by large toxic side effects that reduce patient compliance. In this study, we reviewed the anti-tumor effects and toxicity reduction mechanisms of P. vulgaris, which exerts its efficacy while minimizing hepatic and renal injuries as much as possible. Its high efficiency and low toxicity may make it a promising clinical treatment modality for tumors.
P. vulgaris is widely used in medicine and food as a promising economic medicinal material and functional food. The secondary metabolites of medicinal and edible homologous plants are terpenoids, flavonoids, coumarins, volatile oils, saponins, sterols, etc., which have been confirmed to have a diversified pharmacological activity. They can be combined for multi-target and multi-channel, with good medicinal effects and small side effects [108]. At present, the related research on P. vulgaris is not comprehensive and specific enough, and its quality control, plant identification, toxicology and pharmacokinetics are relatively lacking. Therefore, we should further improve the weak part, based on the superiority of the multi-component and multi-target properties of traditional Chinese medicine to clarify the pharmacological effects and mechanisms of traditional Chinese medicine plants and conduct in-depth research on the intervention of traditional Chinese medicine plants in cancer research. To explore its potential nutritional and medicinal value, we have provided a theoretical and scientific basis for subsequent research and supplied a substantial evaluation of the value of P. vulgaris in the field of medicine and food.

Author Contributions

All authors have contributed to the study of concepts and designs. N.N., Y.N., G.C., S.H. and D.L. contributed to the structure of and ideas in the paper. N.N., Y.Y., F.M. and G.C. carried out document retrieval and sorting analysis. The manuscript was written by N.N. L.Y., Y.N. and D.L. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the “Young Scholars of Western China” (Class A) West Light Foundation of the Chinese Academy of Sciences (XAB2019AW13) and Ningxia Natural Science Foundation (No. 2022AAC02039).

Acknowledgments

The authors acknowledge any support given which is not covered by the authors’ contribution or funding sections.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

OBOral Availability
DLDrug-like properties
KEGGKyoto Encyclopedia of Traditional Chinese Medicine
TCTotal Cholestero
LDL-CLow-density lipoprotein cholesterol
HDL-CHigh-density lipoprotein cholesterol
BAXBcl-2-associated X
Bcl-2B-cell lymphoma-2
VEGFVascular endothelial growth factor
NF-κBNuclear factor kappa-B
PI3KPhosphatidylinositol 3-kinase
AKTProtein kinase-B
JAKJanus Kinase
STATSignal transducer and activator of transcription
ROSReactive oxygen species
PERKPKR-like endoplasmic reticulum kinase
ATF4Recombinant activating transcription factor 4
APAF-1Apoptotic protease activating factor-1
NF-κBNuclear factor kappa-beta
MEKMethyl ethyl ketone
ERKExtracellular regulated protein kinases
JNKC-Jun N-terminal kinase
VEGFR2Vascular endothelial growth factor receptor 2
PCNAProliferating cell nuclear antigen
PARPPoly-ADP-ribose polymerase
mTORMechanistic target of rapamycin
LC3-IIIight chain 3-II
XIAPX-linked inhibitor of apoptosis protein
Jab1Jun activation domain-binding protein 1
cMetC-mesenchymal-epithelial transition
PARP-1Poly-ADP-ribose polymerase 1
FOXO3aForkhead box O3a
Nrf2Nuclear factor erythroid-2 related factor 2
HO-1Heme oxygenase-1
MMPMatrix metalloproteinase
CYT-cCytochrome C
MAPKMitogen-activated protein kinase
ERKExtracellular signal-regulated kinase
MMP2Matrix metallopeptidase 2
MMP16Matrix metallopeptidase 16
MMP9Matrix metallopeptidase 9
AMPKAMP-activated protein kinase
IL-6Interleukin 6
IL-8Interleukin 8
IL-10Interleukin 10
TNF-αTumor necrosis factorα
COX-2Cyclo-oxygenase-2
TGF-β1Transforming growth factor-β1
α-SMAα-Smooth muscle actin
NLRP3NOD-like receptor thermal protein domain associated protein 3
SIRT1Recombinant Sirtuin 1
GSHGlutathione
SODSuperoxide dismutase
MDAMalondialdehyde
ALTAlanine transaminase
ASTAspartate transaminase
CSF-1Macrophage colony-stimulating factor 1
CATCatalase
IL-1βInterleukin—1β
ALPAlkaline phosphatase
TGTriglyceride

References

  1. Nagaraju, G.P.; Kasa, P.; Dariya, B.; Surepalli, N.; Peela, S.; Ahmad, S. Epigenetics and therapeutic targets in gastrointestinal malignancies. Drug Discov. Today 2021, 26, 2303–2314. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Laversanne, M.; Weiderpass, E.; Soerjomataram, I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer 2021, 127, 3029–3030. [Google Scholar] [CrossRef] [PubMed]
  3. Ying, J.; Zhang, M.; Qiu, X.; Lu, Y. The potential of herb medicines in the treatment of esophageal cancer. Biomed. Pharmacother. 2018, 103, 381–390. [Google Scholar] [CrossRef] [PubMed]
  4. Li, J.; Sun, J.; Zeng, Z.; Liu, Z.; Ma, M.; Zheng, Z.; He, Y.; Kang, W. Tumour-associated macrophages in gastric cancer: From function and mechanism to application. Clin. Transl. Med. 2023, 13, e1386. [Google Scholar] [CrossRef] [PubMed]
  5. Johnston, F.M.; Beckman, M. Updates on Management of Gastric Cancer. Curr. Oncol. Rep. 2019, 21, 67. [Google Scholar] [CrossRef] [PubMed]
  6. Gravitz, L. Liver cancer. Nature 2014, 516, S1. [Google Scholar] [CrossRef] [PubMed]
  7. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed]
  8. Anwanwan, D.; Singh, S.K.; Singh, S.; Saikam, V.; Singh, R. Challenges in liver cancer and possible treatment approaches. Biochim. Biophys. Acta Rev. Cancer 2020, 1873, 188314. [Google Scholar] [CrossRef]
  9. Haraldsdottir, S.; Einarsdottir, H.M.; Smaradottir, A.; Gunnlaugsson, A.; Halfdanarson, T.R. Colorectal cancer—Review. Laeknabladid 2014, 100, 75–82. [Google Scholar] [CrossRef]
  10. Baiu, I.; Visser, B. Gallbladder Cancer. JAMA 2018, 320, 1294. [Google Scholar] [CrossRef]
  11. Ansari, D.; Tingstedt, B.; Andersson, B.; Holmquist, F.; Sturesson, C.; Williamsson, C.; Sasor, A.; Borg, D.; Bauden, M.; Andersson, R. Pancreatic cancer: Yesterday, today and tomorrow. Future Oncol. 2016, 12, 1929–1946. [Google Scholar] [CrossRef] [PubMed]
  12. Kawakubo, H.; Ozawa, S.; Ando, N.; Kitagawa, Y.; Mukai, M.; Ueda, M.; Kitajima, M. Alterations of p53, cyclin D1 and pRB expression in the carcinogenesis of esophageal squamous cell carcinoma. Oncol. Rep. 2005, 14, 1453–1459. [Google Scholar] [CrossRef]
  13. Li, F.S.; Weng, J.K. Demystifying traditional herbal medicine with modern approach. Nat. Plants 2017, 3, 17109. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, C.Y.; Bai, X.Y.; Wang, C.H. Traditional Chinese medicine: A treasured natural resource of anticancer drug research and development. Am. J. Chin. Med. 2014, 42, 543–559. [Google Scholar] [CrossRef]
  15. Guo, P.; Zhang, B.; Zhao, J.; Wang, C.; Wang, Z.; Liu, A.; Du, G. Medicine-Food Herbs against Alzheimer’s Disease: A Review of Their Traditional Functional Features, Substance Basis, Clinical Practices and Mechanisms of Action. Molecules 2022, 27, 901. [Google Scholar] [CrossRef] [PubMed]
  16. Hou, Y.; Jiang, J.G. Origin and concept of medicine food homology and its application in modern functional foods. Food Funct. 2013, 4, 1727–1741. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, J.; Lv, G.P.; Chen, Y.W.; Li, S.P. Advanced development in analysis of phytochemicals from medicine and food dual purposes plants used in China. J. Chromatogr. A 2011, 1218, 7453–7475. [Google Scholar] [CrossRef] [PubMed]
  18. Psotová, J.; Kolár, M.; Sousek, J.; Svagera, Z.; Vicar, J.; Ulrichová, J. Biological activities of Prunella vulgaris extract. Phytother. Res. 2003, 17, 1082–1087. [Google Scholar] [CrossRef] [PubMed]
  19. Tan, J.; Qi, H.; Ni, J. Extracts of endophytic fungus xkc-s03 from Prunella vulgaris L. spica inhibit gastric cancer in vitro and in vivo. Oncol. Lett. 2015, 9, 945–949. [Google Scholar] [CrossRef]
  20. Kim, S.H.; Huang, C.Y.; Tsai, C.Y.; Lu, S.Y.; Chiu, C.C.; Fang, K. The aqueous extract of Prunella vulgaris suppresses cell invasion and migration in human liver cancer cells by attenuating matrix metalloproteinases. Am. J. Chin. Med. 2012, 40, 643–656. [Google Scholar] [CrossRef]
  21. Fang, Y.; Zhang, L.; Feng, J.; Lin, W.; Cai, Q.; Peng, J. Spica Prunellae extract suppresses the growth of human colon carcinoma cells by targeting multiple oncogenes via activating miR-34a. Oncol. Rep. 2017, 38, 1895–1901. [Google Scholar] [CrossRef]
  22. Pan, J.; Wang, H.; Chen, Y. Prunella vulgaris L.—A Review of its Ethnopharmacology, Phytochemistry, Quality Control and Pharmacological Effects. Front. Pharmacol. 2022, 13, 903171. [Google Scholar] [CrossRef]
  23. Luo, H.; Zhao, L.; Li, Y.; Xia, B.; Lin, Y.; Xie, J.; Wu, P.; Liao, D.; Zhang, Z.; Lin, L. An in vivo and in vitro assessment of the anti-breast cancer activity of crude extract and fractions from Prunella vulgaris L. Heliyon 2022, 8, e11183. [Google Scholar] [CrossRef]
  24. Guo, Q.; Qu, H.; Zhang, H.; Zhong, X. Prunella vulgaris L. Attenuates Experimental Autoimmune Thyroiditis by Inhibiting HMGB1/TLR9 Signaling. Drug Des. Devel Ther. 2021, 15, 4559–4574. [Google Scholar] [CrossRef]
  25. Han, E.H.; Choi, J.H.; Hwang, Y.P.; Park, H.J.; Choi, C.Y.; Chung, Y.C.; Seo, J.K.; Jeong, H.G. Immunostimulatory activity of aqueous extract isolated from Prunella vulgaris. Food Chem. Toxicol. 2009, 47, 62–69. [Google Scholar] [CrossRef]
  26. Hwang, Y.J.; Lee, E.J.; Kim, H.R.; Hwang, K.A. NF-κB-targeted anti-inflammatory activity of Prunella vulgaris var. lilacina in macrophages RAW 264.7. Int. J. Mol. Sci. 2013, 14, 21489–21503. [Google Scholar] [CrossRef]
  27. Li, Z.; He, Q.; Xu, F.; Yin, X.; Guan, Z.; Song, J.; He, Z.; Yang, X.; Situ, C. Exploring the Antibacterial Potential and Underlying Mechanisms of Prunella vulgaris L. on Methicillin-Resistant Staphylococcus aureus. Foods 2024, 13, 660. [Google Scholar] [CrossRef]
  28. Chen, Y.; Guo, Q.; Wang, C. Research on the Changes of Prunella vulgaris and Its Medicinal Parts. Chin. J. Chin. Mater. Med. 2010, 35, 242–246. [Google Scholar]
  29. Guo, Q.; Chen, Y. Research on the Original Plants of Prunella vulgaris and Their Dietary Therapy History. Chin. J. Chin. Mater. Med. 2011, 36, 3057–3062. [Google Scholar]
  30. Wang, X.; Ma, M.; Zhang, J.; Dai, Y.; Ji, Q.; Gu, X.; Nian, H. Overview of Traditional Chinese Medicine Prunella vulgaris for Medicinal Use. Chin. J. Mod. Appl. Pharm. 2019, 36, 625–632. [Google Scholar] [CrossRef]
  31. Jin, Z.; He, J.; Hu, Y.; Bu, Z.; Ma, N. Progress in Clinical Application of Prunella vulgaris Preparations. China Pharm. 2016, 27, 5034–5037. [Google Scholar]
  32. Brindley, M.A.; Widrlechner, M.P.; McCoy, J.A.; Murphy, P.; Hauck, C.; Rizshsky, L.; Nikolau, B.; Maury, W. Inhibition of lentivirus replication by aqueous extracts of Prunella vulgaris. Virol. J. 2009, 6, 8. [Google Scholar] [CrossRef]
  33. Ryu, S.Y.; Oak, M.H.; Yoon, S.K.; Cho, D.I.; Yoo, G.S.; Kim, T.S.; Kim, K.M. Anti-allergic and anti-inflammatory triterpenes from the herb of Prunella vulgaris. Planta Med. 2000, 66, 358–360. [Google Scholar] [CrossRef]
  34. Rasool, R.; Ganai, B.A.; Akbar, S.; Kamili, A.N.; Masood, A. Phytochemical screening of Prunella vulgaris l. —An important medicinal plant of Kashmir. Pak. J. Pharm. Sci. 2010, 23, 399–402. [Google Scholar]
  35. Xia, B.H.; Xiong, S.H.; Tang, J.; Zhang, Z.M.; Li, Y.M.; Li, M.J.; Lin, L.M. Extraction of flavonoids in Prunella vulgaris based on deep eutectic solvent method: Application of new green solvent. Zhongguo Zhong Yao Za Zhi 2018, 43, 3484–3492. [Google Scholar] [CrossRef]
  36. Fang, X.; Yu, M.M.; Yuen, W.H.; Zee, S.Y.; Chang, R.C. Immune modulatory effects of Prunella vulgaris L. on monocytes/macrophages. Int. J. Mol. Med. 2005, 16, 1109–1116. [Google Scholar]
  37. Wang, Z.J.; Zhao, Y.Y.; Chen, Y.Y.; Ma, B.N. Triterpenoid compounds of Prunella genus and their features of 13C NMR spectroscopy. Zhongguo Zhong Yao Za Zhi 2000, 25, 583–588. [Google Scholar]
  38. Pollier, J.; Goossens, A. Oleanolic acid. Phytochemistry 2012, 77, 10–15. [Google Scholar] [CrossRef]
  39. Liu, Q.; Liu, H.; Zhang, L.; Guo, T.; Wang, P.; Geng, M.; Li, Y. Synthesis and antitumor activities of naturally occurring oleanolic acid triterpenoid saponins and their derivatives. Eur. J. Med. Chem. 2013, 64, 1–15. [Google Scholar] [CrossRef]
  40. Kim, Y.S.; Li, X.F.; Kang, K.H.; Ryu, B.; Kim, S.K. Stigmasterol isolated from marine microalgae Navicula incerta induces apoptosis in human hepatoma HepG2 cells. BMB Rep. 2014, 47, 433–438. [Google Scholar] [CrossRef]
  41. Singh Tuli, H.; Rath, P.; Chauhan, A.; Sak, K.; Aggarwal, D.; Choudhary, R.; Sharma, U.; Vashishth, K.; Sharma, S.; Kumar, M.; et al. Luteolin, a Potent Anticancer Compound: From Chemistry to Cellular Interactions and Synergetic Perspectives. Cancers 2022, 14, 5373. [Google Scholar] [CrossRef]
  42. Psotova, J.; Svobodova, A.; Kolarova, H.; Walterova, D. Photoprotective properties of Prunella vulgaris and rosmarinic acid on human keratinocytes. J. Photochem. Photobiol. B 2006, 84, 167–174. [Google Scholar] [CrossRef]
  43. 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] [PubMed]
  44. González-Vallinas, M.; Reglero, G.; Ramírez de Molina, A. Rosemary (Rosmarinus officinalis L.) Extract as a Potential Complementary Agent in Anticancer Therapy. Nutr. Cancer 2015, 67, 1221–1229. [Google Scholar] [CrossRef]
  45. Gu, X.; Li, Y.; Mu, J.; Zhang, Y. Chemical constituents of Prunella vulgaris. J. Environ. Sci. 2013, 25 (Suppl. S1), S161–S163. [Google Scholar] [CrossRef]
  46. Küpeli Akkol, E.; Renda, G.; İlhan, M.; Bektaş, N.Y. Wound healing acceleration and anti-inflammatory potential of Prunella vulgaris L.: From conventional use to preclinical scientific verification. J. Ethnopharmacol. 2022, 295, 115411. [Google Scholar] [CrossRef]
  47. Komal, S.; Kazmi, S.A.J.; Khan, J.A.; Gilani, M.M. Antimicrobial activity of Prunella vulgaris extracts against multi-drug resistant Escherichia Coli from patients of urinary tract infection. Pak. J. Med. Sci. 2018, 34, 616–620. [Google Scholar] [CrossRef] [PubMed]
  48. Li, L.; Lin, L.M.; Deng, J.; Lin, X.L.; Li, Y.M.; Xia, B.H. The therapeutic effects of Prunella vulgaris against fluoride-induced oxidative damage by using the metabolomics method. Environ. Toxicol. 2021, 36, 1802–1816. [Google Scholar] [CrossRef] [PubMed]
  49. Raafat, K.; Wurglics, M.; Schubert-Zsilavecz, M. Prunella vulgaris L. active components and their hypoglycemic and antinociceptive effects in alloxan-induced diabetic mice. Biomed. Pharmacother. 2016, 84, 1008–1018. [Google Scholar] [CrossRef]
  50. Zhang, Z.; Zhou, Y.; Lin, Y.; Li, Y.; Xia, B.; Lin, L.; Liao, D. GC-MS-based metabolomics research on the anti-hyperlipidaemic activity of Prunella vulgaris L. polysaccharides. Int. J. Biol. Macromol. 2020, 159, 461–473. [Google Scholar] [CrossRef]
  51. Li, C.; You, L.; Fu, X.; Huang, Q.; Yu, S.; Liu, R.H. Structural characterization and immunomodulatory activity of a new heteropolysaccharide from Prunella vulgaris. Food Funct. 2015, 6, 1557–1567. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, X.; Ao, Z.; Bello, A.; Ran, X.; Liu, S.; Wigle, J.; Kobinger, G.; Yao, X. Characterization of the inhibitory effect of an extract of Prunella vulgaris on Ebola virus glycoprotein (GP)-mediated virus entry and infection. Antiviral Res. 2016, 127, 20–31. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, S.J.; Wang, X.H.; Dai, Y.Y.; Ma, M.H.; Rahman, K.; Nian, H.; Zhang, H. Prunella vulgaris: A Comprehensive Review of Chemical Constituents, Pharmacological Effects and Clinical Applications. Curr. Pharm. Des. 2019, 25, 359–369. [Google Scholar] [CrossRef] [PubMed]
  54. Li, C.; Huang, Q.; Xiao, J.; Fu, X.; You, L.; Liu, R.H. Preparation of Prunella vulgaris polysaccharide-zinc complex and its antiproliferative activity in HepG2 cells. Int. J. Biol. Macromol. 2016, 91, 671–679. [Google Scholar] [CrossRef] [PubMed]
  55. Su, Y.C.; Lin, I.H.; Siao, Y.M.; Liu, C.J.; Yeh, C.C. Modulation of the Tumor Metastatic Microenvironment and Multiple Signal Pathways by Prunella vulgaris in Human Hepatocellular Carcinoma. Am. J. Chin. Med. 2016, 44, 835–849. [Google Scholar] [CrossRef]
  56. Zhang, X.; Shen, T.; Zhou, X.; Tang, X.; Gao, R.; Xu, L.; Wang, L.; Zhou, Z.; Lin, J.; Hu, Y. Network pharmacology based virtual screening of active constituents of Prunella vulgaris L. and the molecular mechanism against breast cancer. Sci. Rep. 2020, 10, 15730. [Google Scholar] [CrossRef]
  57. Baer-Dubowska, W.; Narożna, M.; Krajka-Kuźniak, V. Anti-Cancer Potential of Synthetic Oleanolic Acid Derivatives and Their Conjugates with NSAIDs. Molecules 2021, 26, 4957. [Google Scholar] [CrossRef]
  58. Shanmugam, M.K.; Dai, X.; Kumar, A.P.; Tan, B.K.; Sethi, G.; Bishayee, A. Oleanolic acid and its synthetic derivatives for the prevention and therapy of cancer: Preclinical and clinical evidence. Cancer Lett. 2014, 346, 206–216. [Google Scholar] [CrossRef]
  59. Gao, L.; Xu, Z.; Wang, Y.; Sun, B.; Song, Z.; Yang, B.; Liu, X.; Lin, Y.; Peng, J.; Han, G.; et al. Anticancer effect of SZC017, a novel derivative of oleanolic acid, on human gastric cancer cells. Oncol. Rep. 2016, 35, 1101–1108. [Google Scholar] [CrossRef]
  60. Liu, L.; Zhao, J.L.; Wang, J.G. [Oleanolic acid induces G₂/M phase arrest and apoptosis in human hepatocellular carcinoma Bel-7402 cells]. Zhongguo Zhong Yao Za Zhi 2015, 40, 4897–4902. [Google Scholar]
  61. Wei, J.; Liu, H.; Liu, M.; Wu, N.; Zhao, J.; Xiao, L.; Han, L.; Chu, E.; Lin, X. Oleanolic acid potentiates the antitumor activity of 5-fluorouracil in pancreatic cancer cells. Oncol. Rep. 2012, 28, 1339–1345. [Google Scholar] [CrossRef]
  62. Zhang, X.; Wang, J.; Zhu, L.; Wang, X.; Meng, F.; Xia, L.; Zhang, H. Advances in Stigmasterol on its anti-tumor effect and mechanism of action. Front. Oncol. 2022, 12, 1101289. [Google Scholar] [CrossRef] [PubMed]
  63. Li, K.; Yuan, D.; Yan, R.; Meng, L.; Zhang, Y.; Zhu, K. Stigmasterol exhibits potent antitumor effects in human gastric cancer cells mediated via inhibition of cell migration, cell cycle arrest, mitochondrial mediated apoptosis and inhibition of JAK/STAT signalling pathway. JBUON 2018, 23, 1420–1425. [Google Scholar] [PubMed]
  64. Dong, Y.; Chen, C.; Chen, C.; Zhang, C.; Zhang, L.; Zhang, Y.; Li, Y.; Dong, Z. Stigmasterol inhibits the progression of lung cancer by regulating retinoic acid-related orphan receptor C. Histol. Histopathol. 2021, 36, 1285–1299. [Google Scholar] [CrossRef]
  65. Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef]
  66. Chen, Z.; Zhang, B.; Gao, F.; Shi, R. Modulation of G(2)/M cell cycle arrest and apoptosis by luteolin in human colon cancer cells and xenografts. Oncol. Lett. 2018, 15, 1559–1565. [Google Scholar] [CrossRef]
  67. Liu, Y.; Xu, X.; Tang, H.; Pan, Y.; Hu, B.; Huang, G. Rosmarinic acid inhibits cell proliferation, migration, and invasion and induces apoptosis in human glioma cells. Int. J. Mol. Med. 2021, 47, 67. [Google Scholar] [CrossRef]
  68. Jin, B.R.; Chung, K.S.; Hwang, S.; Hwang, S.N.; Rhee, K.J.; Lee, M.; An, H.J. Rosmarinic acid represses colitis-associated colon cancer: A pivotal involvement of the TLR4-mediated NF-κB-STAT3 axis. Neoplasia 2021, 23, 561–573. [Google Scholar] [CrossRef] [PubMed]
  69. Cao, W.; Hu, C.; Wu, L.; Xu, L.; Jiang, W. 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]
  70. Radziejewska, I.; Supruniuk, K.; Bielawska, A. Anti-cancer effect of combined action of anti-MUC1 and rosmarinic acid in AGS gastric cancer cells. Eur. J. Pharmacol. 2021, 902, 174119. [Google Scholar] [CrossRef]
  71. Zhou, X.; Wang, W.; Li, Z.; Chen, L.; Wen, C.; Ruan, Q.; Xu, Z.; Liu, R.; Xu, J.; Bai, Y.; et al. Rosmarinic Acid Decreases the Malignancy of Pancreatic Cancer Through Inhibiting Gli1 Signaling. Phytomedicine 2022, 95, 153861. [Google Scholar] [CrossRef] [PubMed]
  72. Rui, L.X.; Shu, S.Y.; Jun, W.J.; Mo, C.Z.; Wu, S.Z.; Min, L.S.; Yuan, L.; Yong, P.J.; Cheng, S.Z.; Sheng, W.S.; et al. The dual induction of apoptosis and autophagy by SZC014, a synthetic oleanolic acid derivative, in gastric cancer cells via NF-κB pathway. Tumour Biol. 2016, 37, 5133–5144. [Google Scholar] [CrossRef]
  73. Niu, G.; Sun, L.; Pei, Y.; Wang, D. Oleanolic Acid Inhibits Colorectal Cancer Angiogenesis by Blocking the VEGFR2 Signaling Pathway. Anticancer. Agents Med. Chem. 2018, 18, 583–590. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, X.; Bai, H.; Zhang, X.; Liu, J.; Cao, P.; Liao, N.; Zhang, W.; Wang, Z.; Hai, C. Inhibitory effect of oleanolic acid on hepatocellular carcinoma via ERK-p53-mediated cell cycle arrest and mitochondrial-dependent apoptosis. Carcinogenesis 2013, 34, 1323–1330. [Google Scholar] [CrossRef] [PubMed]
  75. Sharma, P.; Caldwell, T.S.; Rivera, M.N.; Gullapalli, R.R. Cadmium exposure activates Akt/ERK Signaling and pro-inflammatory COX-2 expression in human gallbladder epithelial cells via a ROS dependent mechanism. Toxicol. In Vitro 2020, 67, 104912. [Google Scholar] [CrossRef] [PubMed]
  76. Wei, J.; Liu, M.; Liu, H.; Wang, H.; Wang, F.; Zhang, Y.; Han, L.; Lin, X. Oleanolic acid arrests cell cycle and induces apoptosis via ROS-mediated mitochondrial depolarization and lysosomal membrane permeabilization in human pancreatic cancer cells. J. Appl. Toxicol. 2013, 33, 756–765. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, H.; Zhang, X.; Wang, M.; Lin, Y.; Zhou, S. Stigmasterol Simultaneously Induces Apoptosis and Protective Autophagy by Inhibiting Akt/mTOR Pathway in Gastric Cancer Cells. Front. Oncol. 2021, 11, 629008. [Google Scholar] [CrossRef] [PubMed]
  78. Choi, Y.H.; Kong, K.R.; Kim, Y.A.; Jung, K.O.; Kil, J.H.; Rhee, S.H.; Park, K.Y. Induction of BAX and activation of caspases during beta-sitosterol-mediated apoptosis in human colon cancer cells. Int. J. Oncol. 2003, 23, 1657–1662. [Google Scholar]
  79. Pandey, P.; Bajpai, P.; Siddiqui, M.H.; Sayyed, U.; Tiwari, R.; Shekh, R.; Mishra, K.; Kapoor, V.K. Elucidation of the Chemopreventive Role of Stigmasterol Against Jab1 in Gall Bladder Carcinoma. Endocr. Metab. Immune Disord. Drug Targets 2019, 19, 826–837. [Google Scholar] [CrossRef]
  80. Lu, J.; Li, G.; He, K.; Jiang, W.; Xu, C.; Li, Z.; Wang, H.; Wang, W.; Wang, H.; Teng, X.; et al. Luteolin exerts a marked antitumor effect in cMet-overexpressing patient-derived tumor xenograft models of gastric cancer. J. Transl. Med. 2015, 13, 42. [Google Scholar] [CrossRef]
  81. Potočnjak, I.; Šimić, L.; Gobin, I.; Vukelić, I.; Domitrović, R. Antitumor activity of luteolin in human colon cancer SW620 cells is mediated by the ERK/FOXO3a signaling pathway. Toxicol. In Vitro 2020, 66, 104852. [Google Scholar] [CrossRef] [PubMed]
  82. Jang, C.H.; Moon, N.; Lee, J.; Kwon, M.J.; Oh, J.; Kim, J.S. Luteolin Synergistically Enhances Antitumor Activity of Oxaliplatin in Colorectal Carcinoma via AMPK Inhibition. Antioxidants 2022, 11, 626. [Google Scholar] [CrossRef] [PubMed]
  83. Xu, H.; Yang, T.; Liu, X.; Tian, Y.; Chen, X.; Yuan, R.; Su, S.; Lin, X.; Du, G. Luteolin synergizes the antitumor effects of 5-fluorouracil against human hepatocellular carcinoma cells through apoptosis induction and metabolism. Life Sci. 2016, 144, 138–147. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, H.; Huang, M.; Liu, Y.; Shu, Y.; Liu, P. Luteolin Induces Apoptosis by Up-regulating miR-34a in Human Gastric Cancer Cells. Technol. Cancer Res. Treat. 2015, 14, 747–755. [Google Scholar] [CrossRef] [PubMed]
  85. Xavier, C.P.; Lima, C.F.; Fernandes-Ferreira, M.; Pereira-Wilson, C. Salvia fruticosa, Salvia officinalis, and rosmarinic acid induce apoptosis and inhibit proliferation of human colorectal cell lines: The role in MAPK/ERK pathway. Nutr. Cancer 2009, 61, 564–571. [Google Scholar] [CrossRef] [PubMed]
  86. Yehya, A.H.S.; Asif, M.; Abdul Majid, A.M.S.; Oon, C.E. Complementary effects of Orthosiphon stamineus standardized ethanolic extract and rosmarinic acid in combination with gemcitabine on pancreatic cancer. Biomed. J. 2021, 44, 694–708. [Google Scholar] [CrossRef] [PubMed]
  87. Han, Y.; Ma, L.; Zhao, L.; Feng, W.; Zheng, X. Rosmarinic inhibits cell proliferation, invasion and migration via up-regulating miR-506 and suppressing MMP2/16 expression in pancreatic cancer. Biomed. Pharmacother. 2019, 115, 108878. [Google Scholar] [CrossRef]
  88. An, Y.; Zhao, J.; Zhang, Y.; Wu, W.; Hu, J.; Hao, H.; Qiao, Y.; Tao, Y.; An, L. Rosmarinic Acid Induces Proliferation Suppression of Hepatoma Cells Associated with NF-κB Signaling Pathway. Asian Pac. J. Cancer Prev. 2021, 22, 1623–1632. [Google Scholar] [CrossRef] [PubMed]
  89. Li, W.; Li, Q.; Wei, L.; Pan, X.; Huang, D.; Gan, J.; Tang, S. Rosmarinic Acid Analogue-11 Induces Apoptosis of Human Gastric Cancer SGC-7901 Cells via the Epidermal Growth Factor Receptor (EGFR)/Akt/Nuclear Factor kappa B (NF-κB) Pathway. Med. Sci. Monit. Basic. Res. 2019, 25, 63–75. [Google Scholar] [CrossRef]
  90. Hao, B.B.; Pan, X.X.; Fan, Y.; Lu, L.; Qian, X.F.; Wang, X.H.; Zhang, F.; Rao, J.H. Oleanolic acid attenuates liver ischemia reperfusion injury by HO-1/Sesn2 signaling pathway. Hepatobiliary Pancreat. Dis. Int. 2016, 15, 519–524. [Google Scholar] [CrossRef]
  91. Jie, F.; Yang, X.; Yang, B.; Liu, Y.; Wu, L.; Lu, B. Stigmasterol attenuates inflammatory response of microglia via NF-κB and NLRP3 signaling by AMPK activation. Biomed. Pharmacother. 2022, 153, 113317. [Google Scholar] [CrossRef]
  92. Mahdiani, S.; Omidkhoda, N.; Heidari, S.; Hayes, A.W.; Karimi, G. Protective effect of luteolin against chemical and natural toxicants by targeting NF-κB pathway. Biofactors 2022, 48, 744–762. [Google Scholar] [CrossRef] [PubMed]
  93. Wu, S.; Luo, H.; Zhong, Z.; Ai, Y.; Zhao, Y.; Liang, Q.; Wang, Y. Phytochemistry, Pharmacology and Quality Control of Xiasangju: A Traditional Chinese Medicine Formula. Front. Pharmacol. 2022, 13, 930813. [Google Scholar] [CrossRef]
  94. Peng, X.P.; Li, X.H.; Li, Y.; Huang, X.T.; Luo, Z.Q. The protective effect of oleanolic acid on NMDA-induced MLE-12 cells apoptosis and lung injury in mice by activating SIRT1 and reducing NF-κB acetylation. Int. Immunopharmacol. 2019, 70, 520–529. [Google Scholar] [CrossRef] [PubMed]
  95. Kim, N.Y.; Lee, M.K.; Park, M.J.; Kim, S.J.; Park, H.J.; Choi, J.W.; Kim, S.H.; Cho, S.Y.; Lee, J.S. Momordin Ic and oleanolic acid from Kochiae Fructus reduce carbon tetrachloride-induced hepatotoxicity in rats. J. Med. Food 2005, 8, 177–183. [Google Scholar] [CrossRef] [PubMed]
  96. Bai, X.; Qiu, A.; Guan, J.; Shi, Z. Antioxidant and protective effect of an oleanolic acid-enriched extract of A. deliciosa root on carbon tetrachloride induced rat liver injury. Asia Pac. J. Clin. Nutr. 2007, 16 (Suppl. 1), 169–173. [Google Scholar]
  97. Feng, S.; Sui, M.; Wang, D.; Ritzoulis, C.; Farag, M.A.; Shao, P. Pectin-zein based stigmasterol nanodispersions ameliorate dextran sulfate sodium-induced colitis in mice. Food Funct. 2021, 12, 11656–11670. [Google Scholar] [CrossRef] [PubMed]
  98. Owumi, S.E.; Lewu, D.O.; Arunsi, U.O.; Oyelere, A.K. Luteolin attenuates doxorubicin-induced derangements of liver and kidney by reducing oxidative and inflammatory stress to suppress apoptosis. Hum. Exp. Toxicol. 2021, 40, 1656–1672. [Google Scholar] [CrossRef]
  99. Dar, A.A.; Fehaid, A.; Alkhatani, S.; Alarifi, S.; Alqahtani, W.S.; Albasher, G.; Almeer, R.; Alfarraj, S.; Moneim, A.A. The protective role of luteolin against the methotrexate-induced hepato-renal toxicity via its antioxidative, anti-inflammatory, and anti-apoptotic effects in rats. Hum. Exp. Toxicol. 2021, 40, 1194–1207. [Google Scholar] [CrossRef]
  100. Yan, Y.; Jun, C.; Lu, Y.; Jiangmei, S. Combination of metformin and luteolin synergistically protects carbon tetrachloride-induced hepatotoxicity: Mechanism involves antioxidant, anti-inflammatory, antiapoptotic, and Nrf2/HO-1 signaling pathway. Biofactors 2019, 45, 598–606. [Google Scholar] [CrossRef]
  101. Jafaripour, L.; Naserzadeh, R.; Alizamani, E.; Javad Mashhadi, S.M.; Moghadam, E.R.; Nouryazdan, N.; Ahmadvand, H. Effects of Rosmarinic Acid on Methotrexate-induced Nephrotoxicity and Hepatotoxicity in Wistar Rats. Indian. J. Nephrol. 2021, 31, 218–224. [Google Scholar] [CrossRef] [PubMed]
  102. Xiang, Y.; Ji, M.; Wu, L.; Lv, L.; Liang, Q.; Deng, R.; Deng, Z.; Liu, X.; Ren, L.; Feng, X.; et al. Rosmarinic Acid Prevents Cisplatin-Induced Liver and Kidney Injury by Inhibiting Inflammatory Responses and Enhancing Total Antioxidant Capacity, Thereby Activating the Nrf2 Signaling Pathway. Molecules 2022, 27, 7815. [Google Scholar] [CrossRef]
  103. Lu, Y.H.; Hong, Y.; Zhang, T.Y.; Chen, Y.X.; Wei, Z.J.; Gao, C.Y. Rosmarinic acid exerts anti-inflammatory effect and relieves oxidative stress via Nrf2 activation in carbon tetrachloride-induced liver damage. Food Nutr. Res. 2022, 66, 8359. [Google Scholar] [CrossRef] [PubMed]
  104. Hasanein, P.; Sharifi, M. Effects of rosmarinic acid on acetaminophen-induced hepatotoxicity in male Wistar rats. Pharm. Biol. 2017, 55, 1809–1816. [Google Scholar] [CrossRef] [PubMed]
  105. Ji, M.Y.; Bo, A.; Yang, M.; Xu, J.F.; Jiang, L.L.; Zhou, B.C.; Li, M.H. The Pharmacological Effects and Health Benefits of Platycodon grandiflorus-A Medicine Food Homology Species. Foods 2020, 9, 142. [Google Scholar] [CrossRef] [PubMed]
  106. Taoerdahong, H.; Kadeer, G.; Chang, J.; Kang, J.; Ma, X.; Yang, F. A Review Concerning the Polysaccharides Found in Edible and Medicinal Plants in Xinjiang. Molecules 2023, 28, 2054. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, Y.; Yan, Q.; Fan, C.; Mo, Y.; Wang, Y.; Li, X.; Liao, Q.; Guo, C.; Li, G.; Zeng, Z.; et al. Overview and countermeasures of cancer burden in China. Sci. China Life Sci. 2023, 66, 2515–2526. [Google Scholar] [CrossRef]
  108. Lu, Q.; Li, R.; Yang, Y.; Zhang, Y.; Zhao, Q.; Li, J. Ingredients with anti-inflammatory effect from medicine food homology plants. Food Chem. 2022, 368, 130610. [Google Scholar] [CrossRef]
Figure 1. Literature retrieval process.
Figure 1. Literature retrieval process.
Molecules 29 01843 g001
Figure 2. Target acquisition and related pathway screening of P. vulgaris.
Figure 2. Target acquisition and related pathway screening of P. vulgaris.
Molecules 29 01843 g002
Figure 3. Structural formula of active components in P. vulgaris: the compounds highlighted in red are the four active ingredients that were the focus of the review in this study. (Created via Figdraw).
Figure 3. Structural formula of active components in P. vulgaris: the compounds highlighted in red are the four active ingredients that were the focus of the review in this study. (Created via Figdraw).
Molecules 29 01843 g003
Figure 4. Types and Phenotypes of P. vulgaris against digestive system tumors (Created via Figdraw).
Figure 4. Types and Phenotypes of P. vulgaris against digestive system tumors (Created via Figdraw).
Molecules 29 01843 g004
Figure 5. Possible mechanism of P. vulgaris against digestive system tumors (Created via Figdraw).
Figure 5. Possible mechanism of P. vulgaris against digestive system tumors (Created via Figdraw).
Molecules 29 01843 g005
Figure 6. Possible mechanisms of P. vulgaris that reduce toxicity (By Figdraw).
Figure 6. Possible mechanisms of P. vulgaris that reduce toxicity (By Figdraw).
Molecules 29 01843 g006
Table 1. The anticancer active constituents and their mechanisms of action of P. vulgaris are based on the suggestion that compounds previously identified in P. vulgaris.
Table 1. The anticancer active constituents and their mechanisms of action of P. vulgaris are based on the suggestion that compounds previously identified in P. vulgaris.
ComponentTumorExperimental ModelMechanismAction TargetPhenotypeReferences
Oleanolic AcidGastric cancerGastric cancer cell lines SGC-7901 and MGC803NF-κB axisBlocked G2/M phase, upregulated Bcl-2 and Beclin 1 and downregulated the reflection of BAX and ATG 5, inducing apoptosis and autophagy.Apoptosis
Autophagy
[72]
Colorectal cancerHuman Colorectal carcinoma cell line HCT-116MEK/ERK/JNK axisInhibited VEGFR2 phosphorylation and tumor angiogenesis.Angiogenesis[73]
Liver cancerHuman Liver cancer cell HepG2Mitochondrial apoptosis pathwayBlocked the G2/M phase; upregulated Caspase3, BAX; and Cyt. C; and downregulated Bcl-2 expression, inducing tumor cell apoptosis.Apoptosis[74]
Gallbladder cancerHuman Gallbladder cancer cell lines GBC-SD and NOZAKT/ERK axisBlocked G0 phase and downregulated PCNA, ICAM-1 and RhoA to inhibit tumor development.Proliferation[75]
Pancreatic cancerHuman Pancreatic cancer cell line PANC-28Mitochondrial apoptosis pathwayBlocked G1 phase and G2/M phases, inducing ROS production and Cyt. C release, and activated the lysis of caspases-3/9 and PARP, inducing cell apoptosis.Apoptosis[76]
Stigmasterol Gastric cancerGastric cancer cell lines SGC-7901 and MGC803AKT/mTOR axisThe expressions of BAX, Caspase-3, PARP and LC3-II were upregulated, while the expression of Bcl-2 was downregulated, inducing apoptosis and autophagy.Apoptosis
Autophagy
[77]
Liver cancerHuman Liver cancer cell HepG2Mitochondrial apoptosis pathwayBlocked G2/M phase, BAX and p53 were upregulated, Bcl-2 and XIAP were downregulated and aspase-8/9 was activated, inducing apoptosis.Apoptosis[40]
Colorectal cancerHuman Colorectal cancer cell line HCT-116-Decline in the expressions of Bcl-2, cIAP-1 and mRNA; elevation in BAX and Mrn;, and promotion of the release of Cyt.C, inducing cell apoptosis.Apoptosis[78]
Gallbladder cancer Mitochondrial apoptosis pathwayUpregulation of p27 expression, downregulation of Jab1 gene and activation of Caspase-3, inducing cell apoptosis.Apoptosis[79]
LuteolinGastric cancerGastric cancer cell line SGC-7901cMet/Akt/ERK axisReduction in the reflection and phosphorylation of MMP9 and cMet and increase in the reflection of Caspase-3 and PARP-1, inducing apoptosis and reduce invasion.Apoptosis
Invasion
[80]
Colon cancerHuman Colon cancer SW620 cellsERK/FOXO3a axisDecrease BAX, Caspase-3, PARP, and FOXO3a, increase Beclin-1, Atg5, and LC3B-I/II; Induce apoptosis and inhibit autophagy.Apoptosis
Autophagy
[81]
Rosmarinic AcidColorectal cancerHuman Colorectal cancer cell line HCT-116 Nrf2/ARE/HO-1 axisUpregulation of Nrf2 transcription activity, enhancement of HO-1 expression and suppression of proliferation.Proliferation[82]
Liver cancerHuman Liver carcinoma cell HepG2-Upregulation of BAX and p53, downregulation of Bcl-2 expression and induction of PARP lysis, inducing apoptosis.Apoptosis[83]
Esophageal cancerEsophageal carcinoma cell line EC1-Downregulation of MMP; upregulation of p21, p53, CYT-c, Bim, and cPARP levels; and activation of Caspase-3 expression, suppressing tumor growth.Apoptosis[84]
Colon cancerHuman Colon carcinoma cell line HCT15MAPK/ERK axisThe reflection of BAX and Caspase-3 was decreased, while the reflection of Bcl-2 was increased, inducing apoptosis of cancer cells.Apoptosis
Proliferation
[85]
Pancreatic cancerHuman Pancreatic carcinoma cell line Panc-1-Blocked S stage stagnation, activated the cleavage of Caspase-3/9 and PARP and facilitated cell apoptosis.Apoptosis[86]
Pancreatic cancerHuman Pancreatic carcinoma cell line Panc-1MMP-2/16 axis Downregulated MMP2 and MMP16 and inhibited cell invasion and migration.Invasion
Migration
[87]
Liver cancerHuman Liver carcinoma cell HepG2NF-κB axisDownregulated Bcl-2 and upregulated BAX, Caspase-3, MMP2 and MMP9.Migration
Invasion
Proliferation
[88]
Gastric cancerGastric carcinoma cell line SGC-7901-Downregulated Bcl-2, EGFR, Akt, p-Akt and NF-κ, and upregulated BAX and Caspase-3, inducing apoptosis of cancer cells.Apoptosis[89]
Table 2. Possible mechanism of the detoxification of active ingredients in P. vulgaris.
Table 2. Possible mechanism of the detoxification of active ingredients in P. vulgaris.
Active IngredientDiseaseExperimental ModelMechanismEffectReferences
Oleanolic AcidLung injuryNMDA-induced acute lung injury in mice.Lower NF-κB, NLRP3 and BAX upregulate the levels of SIRT1, Nrf2 and Bcl-2 proteins and reduce lung injury.Inflammation
Oxidative stress
Apoptosis
[94]
Liver injuryCCl4-induced liver injury in ratsThe levels of GSH and SOD in mice with liver injury were upregulated, and the antioxidant defense system of liver was enhanced to reduce liver injury.-[95]
Liver injuryCCl4-induced liver injury in ratsDownregulate MDA, ALT and AST and upregulate GSH to alleviate liver toxicity.Inflammation[96]
StigmasterolColitisDSS-induced colitis mouse modelDownregulated levels of TNF-α, IL-6, IL-1β, CSF-1 and COX-2; inhibited NF-κB pathway; and alleviated colitis.Inflammation[97]
LuteolinLiver and kidney injuryDoxorubicin-induced hepatic and renal disorders in ratsThe antioxidant capacity and IL-10 level were improved, the activity of caspase-3/9 was reduced and hepatorenal toxicity was alleviated.Inflammation
Apoptosis
[98]
Liver and kidney injuryMethotrexate-induced hepatorenal toxicity in ratsThe expressions of Nrf2, GSH, CAT and Bcl-2 were upregulated, and the expressions of ROS, NF-κB and BAX were downregulated to reduce hepatorenal toxicity.Inflammation
Apoptosis
[99]
Liver injuryCCl4-induced liver injury in miceTNF-α, IL-6, IL-1β, Caspase-3 and BAX were downregulated, and Bcl-2 was upregulated, reduce toxicity and protect ing against liver damage.Inflammation
Apoptosis
[100]
Rosmarinic AcidLiver and kidney injuryMethotrexate-induced hepatorenal toxicity in ratsUpregulation of GSH and CAT, downregulation of MDA, reduction in degeneration and cell vacuolization in liver tissue and reduction in hepatorenal toxicity.Inflammation[101]
Liver and kidney injuryCisplatin-induced liver and kidney injury in mouse modelDownregulation of ALT, AST, BUN and CRE levels and inflammatory factor IL-1 β, IL-6 and TNF-α, activating the Nrf2 signaling pathway to prevent cisplatin induced liver and kidney damage.Inflammation[102]
Liver and kidney injuryCCl4-induced liver injury in miceDownregulated ALT, ALP, Caspase-3, TG, TC, MDA, TNF-α, IL-6 and IL-8 and upregulated GSH, SOD, CAT and Nrf2 levels to alleviate liver and kidney damage.Inflammation
Apoptosis
[103]
Liver injuryParacetamol-induced hepatotoxicity in ratsMDA, ALT and AST were decreased, while TAC, GSH and GST were increased, reducing liver damage.Inflammation[104]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ning, N.; Nan, Y.; Chen, G.; Huang, S.; Lu, D.; Yang, Y.; Meng, F.; Yuan, L. Anti-Tumor Effects and Toxicity Reduction Mechanisms of Prunella vulgaris: A Comprehensive Review. Molecules 2024, 29, 1843. https://doi.org/10.3390/molecules29081843

AMA Style

Ning N, Nan Y, Chen G, Huang S, Lu D, Yang Y, Meng F, Yuan L. Anti-Tumor Effects and Toxicity Reduction Mechanisms of Prunella vulgaris: A Comprehensive Review. Molecules. 2024; 29(8):1843. https://doi.org/10.3390/molecules29081843

Chicago/Turabian Style

Ning, Na, Yi Nan, Guoqing Chen, Shicong Huang, Doudou Lu, Yating Yang, Fandi Meng, and Ling Yuan. 2024. "Anti-Tumor Effects and Toxicity Reduction Mechanisms of Prunella vulgaris: A Comprehensive Review" Molecules 29, no. 8: 1843. https://doi.org/10.3390/molecules29081843

Article Metrics

Back to TopTop