Next Article in Journal
Complexity of Damage-Associated Molecular Pattern Molecule Expression Profile in Porcine Brain Affected by Ischemic Stroke
Previous Article in Journal
Bioinformatics Analysis Reveals the Evolutionary Characteristics of the Phoebe bournei ARF Gene Family and Its Expression Patterns in Stress Adaptation
Previous Article in Special Issue
Galangin’s Neuroprotective Role: Targeting Oxidative Stress, Inflammation, and Apoptosis in Ischemic Stroke in a Rat Model of Permanent Middle Cerebral Artery Occlusion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Flavonoids from Scutellaria baicalensis: Promising Alternatives for Enhancing Swine Production and Health

1
Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
2
State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
3
Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
4
Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(8), 3703; https://doi.org/10.3390/ijms26083703
Submission received: 5 March 2025 / Revised: 9 April 2025 / Accepted: 10 April 2025 / Published: 14 April 2025
(This article belongs to the Special Issue Natural Bioactives and Inflammation, 2nd Edition)

Abstract

Concerns over vaccine safety, bacterial resistance, and drug residues have led to increased interest in plant extracts for improving swine nutrition and health. Scutellaria baicalensis Georgi, rich in four primary flavonoids—baicalin, baicalein, wogonoside, and wogonin—demonstrates significant pharmacological properties, including anti-inflammatory, antioxidant, antibacterial, and antiviral activities in swine. These flavonoids have been shown to enhance growth performance, improve immunity, modulate gut microbiota, and aid in the prevention and treatment of various diseases. This review highlights the pharmacological effects of these flavonoids in swine, with a focus on network pharmacology to reveal the underlying molecular mechanisms. By constructing drug-target networks and identifying key signaling pathways, the review reveals how these flavonoids interact with biological systems to promote health. Furthermore, it discusses the practical applications of Scutellaria baicalensis flavonoids in swine production and outlines potential future research directions. This work provides a theoretical framework for understanding the therapeutic targets of these flavonoids, offering valuable insights for advancing sustainable and healthy pig farming practices.

Graphical Abstract

1. Introduction

Pork is a critical source of protein in human diets; however, concerns regarding the safety of its production have increased, particularly due to the misuse of antibiotics and other medications. The overuse of antibiotics in livestock is a significant public health issue, as antibiotic residues can accumulate in the food chain and be ingested by humans. This leads to the development of antibiotic resistance in pathogenic bacteria, posing a substantial threat to global health [1]. As a result, there is an urgent need to identify safe and effective alternatives to antibiotics in animal husbandry.
Scutellaria baicalensis Georgi, a perennial herb native to China, is characterized by its robust rhizome, lanceolate leaves, and vibrant purple flowers [2]. In Traditional Chinese Medicine (TCM), the roots of Scutellaria baicalensis, known as Huang-Qin, have been used for centuries to treat conditions such as diarrhea, hypertension, and inflammation [3]. These roots are a rich source of bioactive flavonoids, including baicalin, baicalein, wogonoside, and wogonin, which exhibit a range of pharmacological properties, including anti-inflammatory, anticancer, and antioxidant effects [3]. Recent studies have demonstrated that flavonoids extracted from Scutellaria baicalensis (SFs) can significantly enhance growth performance and improve disease resistance in pigs [4,5]. However, the underlying mechanisms of SFs remain poorly understood, limiting their broader application in the swine industry.
Based on previous research, and by incorporating network pharmacology analyses, this review aims to summarize the biological activities, mechanisms, and roles of SFs in promoting health in pigs. Additionally, it highlights future research directions and explores the potential applications of SFs in animal production.

2. Methods

2.1. Predicted Potential Targets of SFs

The 3D structures, PubChem-CID, and SMILES files of flavonoids from SFs were retrieved from the PubChem database (https://PubChem.NCBI.nlm.nih.gov/, accessed on 2 October 2024). To investigate the underlying mechanisms of SFs, a comprehensive analysis was performed using multiple bioinformatics tools. Target genes were identified using PharmMapper (http://www.lilab-ecust.cn/pharmmapper/, accessed on 2 October 2024), Swiss Target Prediction (http://www.swisstargetprediction.ch/), SEA Search Server (http://sea.bkslab.org/, accessed on 2 October 2024), and the STITCH online database (http://stitch.embl.de/). The identified genes were then merged, deduplicated, and integrated into a drug–ingredient–target interaction network using the String database (string-db.org). The network was visualized using Cytoscape 3.10 (https://github.com/cytoscape/cytoscape, accessed on 2 October 2024), a robust tool for the visualization of complex biological data.

2.2. Predicted Potential Targets of SFs in Inflammation and Oxidative Stress

Inflammation- and oxidative stress-related target genes were obtained from the GeneCards database (https://www.genecards.org/, accessed on 2 October 2024) and the Comparative Toxicogenomics Database (https://ctdbase.org/, accessed on 2 October 2024). These target genes were merged with those of SFs, deduplicated, and uploaded into Venny (https://www.bioinformatics.com.cn/, accessed on 2 October 2024) to identify the potential targets. The resulting potential targets were then downloaded for further analysis

2.3. Construction of Drug–Ingredients–Disease–Targets–KEGG Pathway Network

To perform KEGG pathway enrichment analysis, the potential targets of SFs related to inflammation and oxidative stress were imported into the Database for Annotation, Visualization, and Integrated Discovery (DAVID) (https://david.ncifcrf.gov/) with a threshold of p < 0.05. These potential targets were then loaded into a protein–protein interaction (PPI) network diagram using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database (https://www.string-db.org/). Sus scrofa was selected as the organism, and a minimum interaction score of 0.9 was set as the threshold. For core target screening and visualization, the TSV-format file was loaded into Cytoscape 3.7.2 (https://github.com/cytoscape/cytoscape/, accessed on 2 October 2024) to construct the drug–ingredients–disease–targets–KEGG pathways network.

3. Results and Discussion

3.1. Active Ingredients and Target Genes of SFs

A total of 29 active ingredients from Scutellaria baicalensis Georgi (Huangqin), as documented in the TCMSP database, were evaluated according to Lipinski’s Rule of Five [6], as summarized in Table 1. The bioactive components identified in Scutellaria baicalensis Georgi include flavonoids, flavonoid glycosides, polysaccharides, trace elements, and volatile oils. Among these, flavonoids and their glycosides are particularly notable for their high content and diverse biological activities [7,8]. Notably, the flavonoids baicalin, wogonoside, baicalein, and wogonin (Figure 1) are widely used in clinical practice due to their pharmacological properties, which include heat-clearing, dampness-drying, fire-resolving, detoxifying, and hemostatic effects [9]. Furthermore, these compounds have garnered significant attention in the livestock industry for their potential applications [3].
Network analysis of the typical active ingredients and targets of SFs (Figure 1) reveals potential molecular targets, providing a comprehensive understanding of the intricate interactions between the bioactive components of SFs and their targets. This analysis offers valuable insights into the therapeutic potential of SFs.

3.2. Pharmacological Effects and Mechanisms of SFs

SFs exhibit a broad spectrum of significant medicinal properties and have shown efficacy in various therapeutic areas, including anti-inflammatory [10], antibacterial [11], antiviral [12], antioxidant [13], anticancer [14], and hepatoprotective effects [15]. In this section, we explore the primary pharmacological effects of SFs. To gain insight into the underlying mechanisms, we constructed a network of related pathways using Cytoscape 3.10. This network map clarifies the complex interactions among SFs, as well as their pharmacological effects, molecular targets, and KEGG pathways, providing a solid foundation for further research into the mechanisms of action of SFs.

3.2.1. Anti-Inflammatory Effects

Inflammation is a crucial innate response to tissue injury, primarily aimed at facilitating tissue repair and promoting cell proliferation [16]. SFs, known for their significant anti-inflammatory properties, have been shown to regulate multiple inflammatory cytokines, thereby modulating the inflammatory response [17]. Specifically, baicalin has been demonstrated to reduce inflammation by down-regulating the production of pro-inflammatory cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), interleukin-17 (IL-17), interleukin-1beta (IL-1β), and matrix metalloproteinase-9 (MMP-9) [18]. Additionally, both baicalein and baicalin mitigate inflammation through key signaling pathways, including the NLRP3 inflammasome, toll-like receptor 4 (TLR4), NF-κB, PI3K/Akt/Nrf2, and heme oxygenase 1 (HO-1) [19,20,21]. Wogonin, baicalin, and wogonoside have also been found to significantly reduce lipopolysaccharide (LPS)-induced inflammation [22,23], potentially through mechanisms involving the inhibition of NF-κB and the phosphorylation of p38 mitogen-activated protein kinase (MAPK), while up-regulating heat shock protein 72 (HSP72) [24,25].
To further explore the anti-inflammatory mechanisms of SFs, we utilized bioinformatics tools such as VENNY (bioinformatics.com.cn) and the STRING database (string-db.org) to analyze the ingredient–target–gene–disease–pathway network. We identified 15,580 inflammation-related target genes from the GeneCards database (https://www.genecards.org/). The analysis revealed 332 potential targets, including six core targets (MAPK3, MAPK1, AKT1, PIK3R1, EGFR, and NF-κB), with a degree greater than 15. These targets were significantly enriched in pathways such as MAPK, HIF-1, C-type lectin receptor, IL-17, and PI3K-Akt signaling (p < 3.06 × 10−12) (Figure 2). In conclusion, SFs, including baicalin, wogonin, baicalein, and wogonoside, exert anti-inflammatory effects through multiple pathways, suggesting that they have potential as natural therapeutics for inflammatory diseases.

3.2.2. Antioxidant Effects

Oxidative stress is a major factor contributing to cellular damage, and antioxidants play a critical role in mitigating this harmful process. The antioxidant activity of SFs has been extensively documented, providing protection against oxidative stress-induced damage. SFs exhibit their antioxidant effects through various mechanisms, including inhibition of lipid peroxidation, activation of antioxidant enzymes, free radical scavenging, metal ion complex formation, and elimination of reactive oxygen species (ROS) [26,27]. Notably, the flavonoid compounds baicalin and baicalein demonstrate pronounced antioxidant activity, attributed to their phenolic hydroxyl groups [28,29]. Research has shown that these compounds protect neuronal cells from peroxynitrite anion-induced apoptosis by modulating the expression of 12/15-lipoxygenase (12/15-LOX), reducing mitochondrial ROS production, regulating the Bcl-2–Bax ratio, and inhibiting cytochrome c release [30,31,32,33]. Among these compounds, baicalein is reported to possess superior antioxidant potency compared to baicalin [34]. Furthermore, baicalein’s protective effect against oxidative stress-induced apoptosis is enhanced through activation of the PI3K/Akt pathway [35,36,37].
The antioxidant effects of SFs extend beyond the central nervous system to include skin protection, where they mitigate ultraviolet-induced oxidative damage by scavenging free radicals and resisting lipid peroxidation. This highlights their potential value in dermatological applications [38,39]. The correlation between the antioxidant activity of SFs and their total phenolic content underscores their potential as natural antioxidants in the food industry [26,40]. Additionally, SFs show promise for treating diseases induced by oxidative stress, such as diabetes and steatohepatitis [41]. Baicalin and baicalein are particularly effective in reducing oxidative stress, promoting the expression of antioxidant-related genes, and protecting cells by modulating the Nrf2 pathway [42,43].
Although extensive research has been conducted on the antioxidant activity of SFs, a comprehensive understanding of the molecular mechanisms requires further investigation. To address this, we performed a network pharmacology analysis to systematically elucidate the targets and pathways associated with the antioxidant effects of SFs. Our analysis identified 337 potential targets from a pool of 13,633 oxidative stress-related genes, including MAPK1, MAPK3, AKT1, PIK3R1, RELA, EGFR, and NF-κB (degree ≥ 15), which were significantly enriched in various pathways, such as lipid metabolism, atherosclerosis, MAPK, HIF-1, C-type lectin receptor, EGFR tyrosine kinase inhibitor resistance, and apoptosis (p < 3.40 × 10−12) (Figure 3). These findings highlight the multifaceted antioxidant mechanisms of SFs and their potential therapeutic applications.

3.2.3. Antibacterial Effects

SFs are widely recognized for their antibacterial activity. Baicalein, wogonin, and baicalin have shown significant inhibitory effects on a range of common pathogenic bacteria, including Aeromonas hydrophila, Edwardsiella tarda, Vibrio alginolyticus, Vibrio harveyi, Cariogenic bacteria, Bacillus subtilis, Enterococcus faecalis, Klebsiella pneumoniae, Salmonella enterica, and Escherichia coli [44,45,46,47]. These flavonoids also exhibit antimycotic activity against fungi such as Rhodotorula rubra, Candida albicans, Aspergillus fumigatus, and Geotrichum candidum [48]. Direct bactericidal effects of SFs have been documented against Helicobacter pylori and Staphylococcus aureus [49,50]. Furthermore, SFs demonstrate synergistic effects with certain antibiotics against resistant strains, such as enhancing activity against methicillin-resistant Staphylococcus aureus (MRSA) when combined with ciprofloxacin, ceftriaxone, gentamicin, and penicillin [51,52].
The antibacterial mechanisms of SFs are multifaceted. For example, baicalein inhibits Staphylococcus aureus by disrupting bacterial biofilm formation and reducing the secretion of staphylococcal enterotoxin A and α-hemolysin [53]. It also diminishes virulence by binding directly to the major virulence factor of S. aureus, the von Willebrand factor-binding protein (vWbp), at Asp-75 and Lys-80 residues [54]. Both baicalin and baicalein disrupt nucleic acid formation and alter bacterial energy metabolism, inhibiting bacterial proliferation [55]. Baicalin’s interaction with cell wall synthesis proteins, such as 1UAG and 2X5O, demonstrates its ability to inhibit cell wall synthesis, disrupt cell wall integrity, reduce bacterial enzyme activity, and inhibit nucleotide synthesis [56].
In addition to the direct effects on bacterial proliferation and virulence factors, SFs also demonstrate antibacterial efficacy by targeting various molecular pathways to regulate the host immune response (Table 2). For example, SFs inhibit the activity of extra-intestinal pathogenic Escherichia coli (ExPEC), enterotoxigenic E. coli (ETEC), E. coli K88, Haemophilus parasuis, and Lactobacillus by inhibiting NF-κB/MAPK signaling pathways and reducing activation of the NLRP3 inflammasome [57,58,59,60,61,62]. Furthermore, SFs influence antibacterial activities by inhibiting the PANX-1/P2Y6 signaling pathway, disrupting the LuxS/AI-2 quorum-sensing system, and attenuating biofilm formation, which modulates virulence factor expression. SFs also protect vascular tight junctions, reduce ROS production, and modulate apoptosis [63,64,65,66]. Moreover, SFs alter the expression profiles of microRNAs, long non-coding RNAs, and mRNAs, suggesting their potential role in gene regulation [67,68,69].
Collectively, these mechanisms underscore the potential of SFs as broad-spectrum antibacterial agents, capable of modulating both bacterial and host immune responses, to effectively combat infections. Further research is needed to fully elucidate these mechanisms and explore the therapeutic potential of SFs in promoting healthy pig husbandry.

3.2.4. Antiviral Effects

SFs have demonstrated therapeutic potential against a wide range of viral pathogens, including the influenza A virus [72], severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [73], Zika virus [74], dengue virus [75], and various arboviruses [76]. Furthermore, the immunomodulatory effects of SFs on porcine reproductive and respiratory syndrome virus (PRRSV) and pseudorabies in piglets have been documented [77,78]. However, research on the antiviral effects of SFs in swine husbandry remains in its early stages.
The antiviral mechanisms of SFs involve direct interference with the viral life cycle, enhancement of host immunity, and direct binding to viral proteins [79] (Figure 4). These compounds alleviate pathological manifestations induced by viral infections by modulating the host immune response, which includes regulation of interferon and pro-inflammatory cytokine production, such as TNF-α, IFN-α, IFN-γ, and various interleukins [80]. Baicalin, recognized as a non-nucleoside reverse transcriptase inhibitor with anti-HIV-1 properties, obstructs HIV-1 entry into host cells by disrupting the interaction between the virus and the cell surface receptors [81,82]. Baicalein, a derivative of baicalin [83], induces a conformational change by binding to the hydrophobic region within the catalytic core of integrase, acting as an inhibitor of HIV-1 integrase [84]. Notably, baicalein’s inhibitory effect on HIV-1 reverse transcriptase is four times greater than that of baicalin [83].
In summary, this section emphasizes the substantial research potential of SFs in antiviral therapy. However, further research is needed to fully elucidate the specific antiviral mechanisms and expand their applications, especially concerning porcine viral diseases.

3.3. Application of SFs in Swine Health Management

In addition to their pharmaceutical applications, SFs play a crucial role in enhancing pig health as feed additives, as summarized in Table 3.

3.3.1. Improvement in Growth Performance

The inclusion of SFs in feed additives has shown promising results in enhancing the growth performance of swine (Table 3). For example, a study involving weaned piglets aged 28 to 56 days reported that the administration of fermented S. baicalensis, a derivative of SFs, at a concentration of 1.5 mg/kg of diet, improved appetite, average daily feed intake, and feed conversion efficiency. It also reduced the feed-to-weight ratio and diarrhea rate [85]. Additionally, the inclusion of mixed fermented plants containing SFs in the diets of growing pigs has been shown to improve growth performance, significantly enhance nutrient digestibility, and reduce the emission of noxious gases from excreta [86,87]. The addition of SFs to the diets of pregnant and lactating sows has also been observed to reduce weight loss post-delivery and improve litter performance [88]. In reproductive health, baicalin has been found to enhance the development of both parthenogenetically stimulated and in vitro fertilized pig embryos by modulating mitochondrial activity, stimulating the sonic hedgehog signaling pathway, preventing reactive oxygen species generation, and inhibiting apoptosis [97]. These findings collectively demonstrate the potential of SFs to optimize nutritional management and health in pigs, leading to improved overall growth performance. The application of SFs in feed additives represents a significant advancement in swine nutrition and health that warrants further research and development.

3.3.2. Enhancement of Immunity

SFs have shown significant immunomodulatory effects in swine health management, enhancing the immune systems of weaned piglets, thereby reducing infection rates and improving overall health (Table 3). For example, baicalein has demonstrated considerable benefits in preventing infections, boosting immunity, and modifying the intestinal microbial composition in 28-day-old weaned piglets [89]. In another study, the administration of baicalin at a concentration of 1 g/kg of feed for the first week, followed by 500 mg/kg for the next two weeks, was found to alleviate intestinal injury in 21-day-old weaned piglets, suggesting a protective role against intestinal inflammation [90]. Additionally, feeds containing SFs have been shown to offer preventive benefits against Salmonella infections in piglets due to their anti-inflammatory properties [89]. These studies collectively highlight SFs’ potential as immunomodulators in the dietary management of pigs, especially during the critical early weaning stage when their immune systems are not yet fully developed. The strategic use of SFs at this stage can provide essential support for the health and development of these young animals, emphasizing the importance of further research and optimization of SFs in swine nutrition and health management.

3.3.3. Regulation of the Intestinal Microbiome

The modulation of the gut microbiome is a key aspect of swine health management, and SFs have emerged as effective feed additives in this process (Table 3). Research has shown that SFs significantly influence the composition of the intestinal microbiota. For instance, administering a mixed fermented plant containing SFs to weaned piglets at 25 days of age has been demonstrated to alter gut microbial composition, which may contribute to improved ether extract digestibility, gut health, and overall health status [5]. Furthermore, SFs have shown protective effects against intestinal injury caused by E. coli K88 in weaned piglets, exerting their effects through the NF-κB/P38 signaling pathways [58]. Additionally, administering baicalin–aluminum complexes at a dosage of 1.36 g/day for three days has been found to modulate the gut microbiomes of 10-day-old piglets, suggesting a role in maintaining gut homeostasis [91].
These findings underscore the potential of SFs to foster a healthy gut environment, which is essential for preventing gastrointestinal disorders and enhancing overall health in swine. The capacity of SFs to modulate the gut microbiome highlights their significance as a dietary supplement in swine nutrition, promoting the development of robust and diverse microbial communities that are essential for animal health and well-being.

3.3.4. Disease Prevention and Treatment

SFs have been extensively studied for their therapeutic potential in treating various diseases in pigs, owing to their anti-inflammatory, antioxidant, antibacterial, and antiviral properties. As detailed in Table 2, SFs have demonstrated significant efficacy in the swine industry, particularly against bacterial infections. Baicalin, a key component of SFs, has proven effective in preventing and treating infection with Haemophilus parasuis, a common cause of Glässer’s disease in pigs, which is characterized by vascular inflammation and damage [68,69,70]. Fermented plants and traditional Chinese medicine formulations containing SFs also play a crucial role in reducing the incidence of diarrhea in pigs, due to their regulatory effects on intestinal health and potent antimicrobial activity [60,92,93,98]. In the treatment of edema disease in weaned piglets aged 28 to 35 days, baicalein injections at doses of 0.2 and 0.4 mL/kg have achieved therapeutic efficacy rates as high as 90% [94], indicating that they are effective in treating edema in weaned piglets. In vitro experiments have shown that baicalin, at concentrations of 5–20 μg/mL, significantly inhibits the porcine reproductive and respiratory syndrome virus (PRRSV) in a dose-dependent manner, through the regulation of antiviral cytokine expression and direct interaction with virions, thereby affecting various stages of the virus life cycle [77]. Additionally, polysaccharides from Scutellaria baicalensis (SGP) have exhibited therapeutic effects against pseudorabies virus (PRV) infection in piglets by reducing clinical symptoms and pathological damage while enhancing T lymphocyte conversion [78].
In summary, the broad-spectrum pharmacological effects of SFs underscore their potential as valuable therapeutic feed additives for pig diseases. Their ability to regulate immune responses, improve intestinal health, and directly target pathogens positions SFs as promising candidates for advancement in veterinary medicine, potentially serving as alternatives to antibiotics in feed additives.

4. Conclusions and Future Directions

SFs demonstrate a broad spectrum of pharmacological properties that effectively enhance porcine growth performance and overall health status, consequently improving production efficiency and economic viability in swine operations. Experimental evidence reveals that the administration of SFs significantly improves intestinal barrier integrity and modulates immune responses, thereby reducing disease susceptibility and associated veterinary expenditures. Their demonstrated efficacy as natural antimicrobial and anti-inflammatory compounds enables partial substitution of conventional antibiotics in feed formulations, addressing critical concerns regarding antimicrobial resistance development while meeting increasing market demand for antibiotic-free animal products. From an environmental perspective, supplementation with SFs contributes to ecological sustainability through reduced antibiotic residues in effluents, thereby mitigating soil and water contamination risks. These multifaceted advantages position SFs as promising phytogenic additives for sustainable swine production systems, demonstrating considerable potential for practical applications in modern intensive farming systems.
Despite their demonstrated anti-inflammatory, antibacterial, antiviral, and antioxidant properties, the clinical application of SFs is currently limited due to their low bioavailability. To overcome this challenge, advanced technologies such as high-throughput screening should be employed to rapidly identify the bioactive constituents of SFs with therapeutic potential. Additionally, bioinformatics and proteomics can provide deeper insights into the efficacy and safety of SFs. Optimizing the extraction process, innovating SF formulations in conjunction with other TCM components, and exploring modern production methods such as ultra-micro grinding technology and TCM fermentation, are crucial for developing more effective products. Furthermore, enhancing the solubility, stability, and bioavailability of SFs’ secondary metabolites, as well as improving their palatability as feed additives, is essential for their broader application in animal husbandry.
SFs have emerged as important regulators of lipid metabolism and fat synthesis in animals. While data regarding their effects on fat metabolism in pigs are limited, studies indicate that baicalin and baicalein possess anti-hyperlipidemic properties by modulating pathways such as PPAR signaling, glycerolipid metabolism, and cholesterol biosynthesis [99,100]. Additionally, SFs have been shown to reduce triglyceride levels, prevent hepatic fat accumulation, and improve insulin resistance through the AMPK-mediated SREBP pathway [101,102]. SFs have also demonstrated potential in mitigating metabolic dysregulation caused by high-fat and high-sugar diets [103]. These findings suggest that SFs may have significant effects on lipid metabolism and fat synthesis in pigs, warranting further investigation.
As a natural antiviral agent, SFs hold promise for mitigating the emergence of drug resistance and modulating the host immune system. Future research should focus on exploring the application of SFs in the prevention and control of swine diseases, integrating the concepts of “prevention before disease onset” and “medicine–food homology.” Incorporating SFs into modern husbandry practices, such as feed additives, could reduce the stress associated with vaccination and drug treatment while enhancing pig immunity and disease resistance. Moreover, understanding the mechanisms underlying the antibacterial and antiviral actions of SFs is critical for evaluating their potential as antibiotic-free feed additives and disinfectants. This would integrate the traditional properties of SFs with modern scientific advancements, offering innovative solutions for sustainable and healthy pig husbandry.
In conclusion, this review has emphasized the significant role of SFs in promoting healthy pig husbandry. We have highlighted their active ingredients, target genes, pharmacological effects, signaling pathways, and practical applications. By elucidating the underlying mechanisms of SFs, we aim to provide guidelines for their integration into the development of healthier and more sustainable pig husbandry practices.

Author Contributions

J.W. conceptualized the project, collected data, drafted the manuscript, and revised the manuscript; Y.Q. and M.T. corrected the language in the manuscript; K.G., L.W., and Z.J. revised the manuscript; X.Y. administered the project and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Guangdong Basic and Applied Basic Research Foundation (2025A1515012362), Special Fund for Scientific Innovation Strategy–construction of High-Level Academy of Agriculture Science (R2023PY-JG013, R2020PY-JX007), Guangzhou Science and Technology Project (No. 202206010193), Guangdong Feed Industry Technology System (Grant No. 2024CXTD14), earmarked fund for CARS (CARS-35).

Acknowledgments

Graphical abstract and Figure 4 in this review were created using FigDraw (https://www.figdraw.com/). We kindly provide thanks for the authorization from the platform.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

SFsScutellaria baicalensis’s Flavonoids
ADGAverage Daily Gain
ADFIAverage Daily Feed Intake
KEGGKyoto Encyclopedia of Genes and Genomes
MAPKMitogen-Activated Protein Kinase
AKTProtein Kinase B
PI3KPhosphoinositide 3-Kinase
EGFREpidermal Growth Factor Receptor
NF-kBNuclear Factor kappa-light-chain-enhancer of Activated B cells
NLRP3NOD-like Receptor Family Pyrin Domain Containing 3
ROSReactive Oxygen Species
HIVHuman Immunodeficiency Virus
TCMTraditional Chinese Medicine

References

  1. Cabello, F.C. Heavy Use of Prophylactic Antibiotics in Aquaculture: A Growing Problem for Human and Animal Health and for the Environment. Environ. Microbiol. 2010, 8, 1137–1144. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, T.; Tang, H.; Xie, L.; Zheng, Y.; Ma, Z.; Sun, Q.; Li, X. Scutellaria baicalensis Georgi. (Lamiaceae): A Review of Its Traditional Uses, Botany, Phytochemistry, Pharmacology and Toxicology. J. Pharm. Pharmacol. 2019, 71, 1353–1369. [Google Scholar] [CrossRef] [PubMed]
  3. Shen, J.; Li, P.; Liu, S.; Liu, Q.; Li, Y.; Sun, Y.; He, C.; Xiao, P. Traditional Uses, Ten-Years Research Progress on Phytochemistry and Pharmacology, and Clinical Studies of the Genus Scutellaria. J. Ethnopharmacol. 2021, 265, 113198. [Google Scholar] [CrossRef] [PubMed]
  4. He, Z.; Li, C.; Xiao, X.; Liu, H.; Wang, M.; Zhou, X.; He, J. Effects of Extracts of Scutellaria baicalensis and Lonicerae flos on Reproductive Performance, Milk Quality and Serum Indexes in Sows and Growth Performance in Suckling Piglets. Livest. Sci. 2023, 275, 105295. [Google Scholar] [CrossRef]
  5. Zhang, F.; Jin, E.; Liu, X.; Ji, X.; Hu, H. Effect of Dietary Fructus Mume and Scutellaria baicalensis Georgi on the Fecal Microbiota and Its Correlation with Apparent Nutrient Digestibility in Weaned Piglets. Animals 2022, 12, 2418. [Google Scholar] [CrossRef]
  6. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings. Adv. Drug Deliv. Rev. 2012, 64, 4–17. [Google Scholar] [CrossRef]
  7. Bochořáková, H.; Paulová, H.; Slanina, J.; Musil, P.; Táborská, E. Main Flavonoids in the Root of Scutellaria baicalensis Cultivated in Europe and Their Comparative Antiradical Properties. Phytotherapy Res. 2003, 17, 640–644. [Google Scholar] [CrossRef]
  8. Li, C.; Lin, G.; Zuo, Z. Pharmacological Effects and Pharmacokinetics Properties of Radix scutellariae and Its Bioactive Flavones. Biopharm. Drug Disp. 2011, 32, 427–445. [Google Scholar] [CrossRef]
  9. Li, H.; Jiang, Y.; Chen, F. Separation Methods Used for Scutellaria baicalensis Active Components. J. Chromatogr. B 2004, 812, 277–290. [Google Scholar] [CrossRef]
  10. Chen, Q.; Wang, C.; Yang, Z.; Tang, Q.; Tan, H.; Wang, X.; Cai, S. Differences in Anti-Inflammatory Effects between Two Specifications of Scutellariae Radix in LPS-Induced Macrophages in Vitro. Chin. J. Nat. Med. 2017, 15, 515–524. [Google Scholar] [CrossRef]
  11. Shan, B.; Cai, Y.-Z.; Brooks, J.D.; Corke, H. The In Vitro Antibacterial Activity of Dietary Spice and Medicinal Herb Extracts. Int. J. Food Microbiol. 2007, 117, 112–119. [Google Scholar] [CrossRef] [PubMed]
  12. Eiden, M.; Leidel, F.; Strohmeier, B.; Fast, C.; Groschup, M.H. A Medicinal Herb Scutellaria Lateriflora Inhibits PrP Replication in Vitro and Delays the Onset of Prion Disease in Mice. Front. Psychiatry 2012, 3, 20692. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, D.; Cao, J.; Zhu, B.; Wang, Z.; Wang, T.; Tang, J. Baicalin Attenuates Joint Pain and Muscle Dysfunction by Inhibiting Muscular Oxidative Stress in an Experimental Osteoarthritis Rat Model. Arch. Immunol. Ther. Exp. 2018, 66, 453–461. [Google Scholar] [CrossRef] [PubMed]
  14. Cheng, C.; Chen, J.; Tan, H.; Wang, N.; Chen, Z.; Feng, Y. Scutellaria baicalensis and Cancer Treatment: Recent Progress and Perspectives in Biomedical and Clinical Studies. Am. J. Chin. Med. 2018, 46, 25–54. [Google Scholar] [CrossRef]
  15. Dong, Q.; Chu, F.; Wu, C.; Huo, Q.; Gan, H.; Li, X.; Liu, H. Scutellaria baicalensis Georgi Extract Protects against Alcohol-Induced Acute Liver Injury in Mice and Affects the Mechanism of ER Stress. Mol. Med. Rep. 2016, 13, 3052–3062. [Google Scholar] [CrossRef]
  16. Medzhitov, R. Origin and Physiological Roles of Inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef]
  17. Smith, J.F.; Starr, E.G.; Goodman, M.A.; Hanson, R.B.; Palmer, T.A.; Woolstenhulme, J.B.; Weyand, J.A.; Marchant, A.D.; Bueckers, S.L.; Nelson, T.K.; et al. Topical Application of Wogonin Provides a Novel Treatment of Knee Osteoarthritis. Front. Physiol. 2020, 11, 80. [Google Scholar] [CrossRef]
  18. Dinda, B.; Dinda, S.; DasSharma, S.; Banik, R.; Chakraborty, A.; Dinda, M. Therapeutic Potentials of Baicalin and Its Aglycone, Baicalein against Inflammatory Disorders. Eur. J. Med. Chem. 2017, 131, 68–80. [Google Scholar] [CrossRef]
  19. Jin, X.; Liu, M.; Zhang, D.; Zhong, X.; Du, K.; Qian, P.; Yao, W.; Gao, H.; Wei, M. Baicalin Mitigates Cognitive Impairment and Protects Neurons from Microglia-mediated Neuroinflammation via Suppressing NLRP 3 Inflammasomes and TLR4/NF-κB Signaling Pathway. CNS Neurosci. Ther. 2019, 25, 575–590. [Google Scholar] [CrossRef]
  20. Rui, W.; Li, S.; Xiao, H.; Xiao, M.; Shi, J. Baicalein Attenuates Neuroinflammation by Inhibiting NLRP3/Caspase-1/GSDMD Pathway in MPTP-Induced Mice Model of Parkinson’s Disease. Int. J. Neuropsychopharmacol. 2020, 23, 762–773. [Google Scholar] [CrossRef]
  21. Wen, Y.; Wang, Y.; Zhao, C.; Zhao, B.; Wang, J. The Pharmacological Efficacy of Baicalin in Inflammatory Diseases. Int. J. Mol. Sci. 2023, 24, 9317. [Google Scholar] [CrossRef] [PubMed]
  22. Han, Y.K.; Kim, H.; Shin, H.; Song, J.; Lee, M.K.; Park, B.; Lee, K.Y. Characterization of Anti-Inflammatory and Antioxidant Constituents from Scutellaria baicalensis Using LC-MS Coupled with a Bioassay Method. Molecules 2020, 25, 3617. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, S.; Sun, A.; Liu, R. Separation and Purification of Baicalin and Wogonoside from the Chinese Medicinal Plant Scutellaria baicalensis Georgi by High-Speed Counter-Current Chromatography. J. Chromatogr. A 2005, 1066, 243–247. [Google Scholar] [CrossRef]
  24. Guo, M.; Zhang, N.; Li, D.; Liang, D.; Liu, Z.; Li, F.; Fu, Y.; Cao, Y.; Deng, X.; Yang, Z. Baicalin Plays an Anti-Inflammatory Role through Reducing Nuclear Factor-κB and P38 Phosphorylation in S. aureus-Induced Mastitis. Int. Immunopharmacol. 2013, 16, 125–130. [Google Scholar] [CrossRef]
  25. Yang, W.; Li, H.; Cong, X.; Wang, X.; Jiang, Z.; Zhang, Q.; Qi, X.; Gao, S.; Cao, R.; Tian, W. Baicalin Attenuates Lipopolysaccharide Induced Inflammation and Apoptosis of Cow Mammary Epithelial Cells by Regulating NF-κB and HSP72. Int. Immunopharmacol. 2016, 40, 139–145. [Google Scholar] [CrossRef]
  26. Chen, Z.; Su, Y.; Bi, Y.; Tsang, S.; Huang, Y. Effect of Baicalein and Acetone Extract of Scutellaria baicalensis on Canola Oil Oxidation. J. Am. Oil Chem. Soc. 2000, 77, 73–78. [Google Scholar] [CrossRef]
  27. Cheng, F.; Lu, Y.; Zhong, X.; Song, W.; Wang, X.; Sun, X.; Qin, J.; Guo, S.; Wang, Q. Baicalin’s Therapeutic Time Window of Neuroprotection during Transient Focal Cerebral Ischemia and Its Antioxidative Effects in Vitro and in Vivo. Evidence-Based Complement. Altern. Med. 2013, 2013, 120261. [Google Scholar] [CrossRef]
  28. Tong, R.; Mehendale, S.R.; Wang, C.-Z.; Shao, Z.; Yuan, C.-S. Letter to the Editor—Comparison of Antioxidant Effects of Various Scutellaria baicalensis Fractions and the Potential Role of Catalase Upregulation. Am. J. Chin. Med. 2009, 37, 621–623. [Google Scholar] [CrossRef]
  29. Xue, Y.; Zheng, Y.; An, L.; Dou, Y.; Liu, Y. Density Functional Theory Study of the Structure–Antioxidant Activity of Polyphenolic Deoxybenzoins. Food Chem. 2014, 151, 198–206. [Google Scholar] [CrossRef]
  30. Cui, L.; Zhang, X.; Yang, R.; Liu, L.; Wang, L.; Li, M.; Du, W. Baicalein Is Neuroprotective in Rat MCAO Model: Role of 12/15-Lipoxygenase, Mitogen-Activated Protein Kinase and Cytosolic Phospholipase A2. Pharmacol. Biochem. Behav. 2010, 96, 469–475. [Google Scholar] [CrossRef]
  31. He, X.; Wang, Y.; Gao, M.; Li, X.; Zhang, T.; Du, G. Baicalein Protects Rat Brain Mitochondria against Chronic Cerebral Hypoperfusion-Induced Oxidative Damage. Brain Res. 2009, 1249, 212–221. [Google Scholar] [CrossRef] [PubMed]
  32. Szabó, C.; Ischiropoulos, H.; Radi, R. Peroxynitrite: Biochemistry, Pathophysiology and Development of Therapeutics. Nat. Rev. Drug Discov. 2007, 6, 662–680. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, M.; Chen, X.; Gu, Y.; Peng, T.; Yang, D.; Chang, R.C.-C.; So, K.-F.; Liu, K.; Shen, J. Baicalin Can Scavenge Peroxynitrite and Ameliorate Endogenous Peroxynitrite-Mediated Neurotoxicity in Cerebral Ischemia-Reperfusion Injury. J. Ethnopharmacol. 2013, 150, 116–124. [Google Scholar] [CrossRef] [PubMed]
  34. Gao, Z.; Huang, K.; Xu, H. Protective Effects of Flavonoids in the Roots of Scutellaria baicalensis Georgi against Hydrogen Peroxide-Induced Oxidative Stress in Hs-Sy5y Cells. Pharmacol. Res. 2001, 43, 173–178. [Google Scholar] [CrossRef]
  35. Liu, C.; Wu, J.; Xu, K.; Cai, F.; Gu, J.; Ma, L.; Chen, J. Neuroprotection by Baicalein in Ischemic Brain Injury Involves PTEN/AKT Pathway. J. Neurochem. 2010, 112, 1500–1512. [Google Scholar] [CrossRef]
  36. Manning, B.D.; Cantley, L.C. AKT/PKB Signaling: Navigating Downstream. Cell 2007, 129, 1261–1274. [Google Scholar] [CrossRef]
  37. Wang, J.; Chen, T.; Jiang, J.; Shi, H.; Gui, C.; Luo, R.; Xie, X.; Xiang, M.; Zhang, X. Hypoxic Preconditioning Attenuates Hypoxia/Reoxygenation-Induced Apoptosis in Mesenchymal Stem Cells. Acta Pharmacol. Sin. 2008, 29, 74–82. [Google Scholar] [CrossRef]
  38. Gabrielska, J.; Oszmiańsk, J.; Żyłka, R.; Komorowska, M. Antioxidant Activity of Flavones from Scutellaria baicalensis in Lecithin Liposomes. Z. Naturforsch. C J. Biosci. 1997, 52, 817–823. [Google Scholar] [CrossRef]
  39. Gao, B.; Zhu, H.; Liu, Z.; He, X.; Sun, J.; Li, Y.; Wu, X.; Pehrsson, P.; Zhang, Y.; Yu, L. Chemical Compositions of Scutellaria baicalensis Georgi. (Huangqin) Extracts and Their Effects on ACE2 Binding of SARS-CoV-2 Spike Protein, ACE2 Activity, and Free Radicals. Int. J. Mol. Sci. 2024, 25, 2045. [Google Scholar] [CrossRef]
  40. Chan, E.; Wong, C.Y.-K.; Wan, C.-W.; Kwok, C.-Y.; Wu, J.-H.; Ng, K.-M.; So, C.-H.; Au, A.L.-S.; Poon, C.C.-W.; Seto, S.-W.; et al. Evaluation of Anti-Oxidant Capacity of Root of Scutellaria baicalensis Georgi, in Comparison with Roots of Polygonum Multiflorum Thunb and Panax Ginseng CA Meyer. Am. J. Chin. Med. 2010, 38, 815–827. [Google Scholar] [CrossRef]
  41. Xue, Y.; Wei, Y.; Cao, L.; Shi, M.; Sheng, J.; Xiao, Q.; Cheng, Z.; Luo, T.; Jiao, Q.; Wu, A.; et al. Protective Effects of Scutellaria-Coptis Herb Couple against Non-Alcoholic Steatohepatitis via Activating NRF2 and FXR Pathways in Vivo and in Vitro. J. Ethnopharmacol. 2024, 318, 116933. [Google Scholar] [CrossRef] [PubMed]
  42. Yin, B.; Li, W.; Qin, H.; Yun, J.; Sun, X. The Use of Chinese Skullcap (Scutellaria baicalensis) and Its Extracts for Sustainable Animal Production. Animals 2021, 11, 1039. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, Q.; Liu, J.; Duan, H.; Li, R.; Peng, W.; Wu, C. Activation of Nrf2/HO-1 Signaling: An Important Molecular Mechanism of Herbal Medicine in the Treatment of Atherosclerosis via the Protection of Vascular Endothelial Cells from Oxidative Stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef] [PubMed]
  44. Duan, C.; Matsumura, S.; Kariya, N.; Nishimura, M.; Shimono, T. In Vitro Antibacterial Activities of Scutellaria baicalensis Georgi against Cariogenic Bacterial. Pediatr. Dent. J. 2007, 17, 58–64. [Google Scholar] [CrossRef]
  45. Jiang, H.; Bai, Z.; Xu, Z.; Sun, J.; Françoise, H.; Luan, Z.; Wang, H. Antimicrobial Mechanism of Semi-Bionic Extracts of Three Traditional Medicinal Plants—Rheum Palmatum L., Scutellaria baicalensis Georgi, and Houttuynia cordata Thunb—That Can Be Used as Antibiotic Alternatives. Front. Vet. Sci. 2023, 9, 1083223. [Google Scholar] [CrossRef]
  46. Pant, C.C.; Melkani, A.B.; Mohan, L.; Dev, V. Composition and Antibacterial Activity of Essential Oil from Scutellaria Grossa Wall Ex Benth. Nat. Prod. Res. 2012, 26, 190–192. [Google Scholar] [CrossRef]
  47. Xia, Y.; Chan, G.K.; Wang, H.; Dong, T.T.; Duan, R.; Hu, W.; Qin, Q.; Wang, W.; Tsim, K.W. The Anti-Bacterial Effects of Aerial Parts of Scutellaria baicalensis: Potential Application as an Additive in Aquaculture Feedings. Aquaculture 2020, 526, 735418. [Google Scholar] [CrossRef]
  48. Blaszczyk, T.; Krzyzanowska, J.; Lamer-Zarawska, E. Screening for Antimycotic Properties of 56 Traditional Chinese Drugs. Phytother. Res. 2000, 14, 210–212. [Google Scholar] [CrossRef]
  49. Qiu, F.; Meng, L.; Chen, J.; Jin, H.; Jiang, L. In Vitro Activity of Five Flavones from Scutellaria baicalensis in Combination with Cefazolin against Methicillin Resistant Staphylococcus Aureus (MRSA). Med. Chem. Res. 2016, 25, 2214–2219. [Google Scholar] [CrossRef]
  50. Wu, J.; Hu, D.; Wang, K.-X. Study of Scutellaria baicalensis and Baicalin against antimicrobial susceptibility of Helicobacter pylori strains in vitro. Zhong Yao Cai 2008, 31, 707–710. [Google Scholar]
  51. Chan, B.C.; Ip, M.; Lau, C.B.; Lui, S.; Jolivalt, C.; Ganem-Elbaz, C.; Litaudon, M.; Reiner, N.E.; Gong, H.; See, R.H.; et al. Synergistic Effects of Baicalein with Ciprofloxacin against NorA Over-Expressed Methicillin-Resistant Staphylococcus aureus (MRSA) and Inhibition of MRSA Pyruvate Kinase. J. Ethnopharmacol. 2011, 137, 767–773. [Google Scholar] [CrossRef] [PubMed]
  52. Qian, M.; Tang, S.; Wu, C.; Wang, Y.; He, T.; Chen, T.; Xiao, X. Synergy between Baicalein and Penicillins against Penicillinase-Producing Staphylococcus aureus. Int. J. Med. Microbiol. 2015, 305, 501–504. [Google Scholar] [CrossRef] [PubMed]
  53. Mao, Y.; Liu, P.; Chen, H.; Wang, Y.; Li, C.; Wang, Q. Baicalein Inhibits the Staphylococcus aureus Biofilm and the LuxS/AI-2 System in Vitro. Infect. Drug Res. 2023, 16, 2861–2882. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, H.; Luan, Y.; Jing, S.; Wang, Y.; Gao, Z.; Yang, P.; Ding, Y.; Wang, L.; Wang, D.; Wang, T. Baicalein Mediates Protection against Staphylococcus aureus-Induced Pneumonia by Inhibiting the Coagulase Activity of vWbp. Biochem. Pharmacol. 2020, 178, 114024. [Google Scholar] [CrossRef]
  55. Chen, W.; Li, B.; Li, S.; Ou, Y.; Ou, Q. Effects of Scutellaria baicalensis on Activity and Biofilm Formation of Klebsiella pneumoniae. Chin. Med. Sci. J. 2016, 31, 180–184. [Google Scholar] [CrossRef]
  56. Dharani, R.S.; Ranjitha, R.; Sripathi, R.; Ali Muhammad, K.S.; Ravi, S. Docking Studies in Target Proteins Involved in Antibacterial Action Mechanisms: Alkaoids Isolated from Scutellaria Genus. Asian J. Pharm. Clin. Res. 2016, 9, 121. [Google Scholar] [CrossRef]
  57. Fu, S.; Liu, H.; Xu, L.; Qiu, Y.; Liu, Y.; Wu, Z.; Ye, C.; Hou, Y.; Hu, C.-A.A. Baicalin Modulates NF-κB and NLRP3 Inflammasome Signaling in Porcine Aortic Vascular Endothelial Cells Infected by Haemophilus parasuis Causing Glässer’s Disease. Sci. Rep. 2018, 8, 807. [Google Scholar] [CrossRef]
  58. Huang, C.; Wang, Y.; He, X.; Jiao, N.; Zhang, X.; Qiu, K.; Piao, X.; Yin, J. The Involvement of NF-κB/P38 Pathways in Scutellaria baicalensis Extracts Attenuating of Escherichia coli K88-Induced Acute Intestinal Injury in Weaned Piglets. Br. J. Nutr. 2019, 122, 152–161. [Google Scholar] [CrossRef]
  59. Liu, X.; Liu, F.; Ma, Y.; Li, H.; Ju, X.; Xu, J. Effect of Puerarin, Baicalin and Berberine Hydrochloride on the Regulation of IPEC-J2 Cells Infected with Enterotoxigenic Escherichia coli. Evid. Based Complement. Alternat. Med. 2019, 2019, 7438593. [Google Scholar] [CrossRef]
  60. Luo, Z.; Liu, C.; Hu, Y.; Xia, T.; Zhang, B.; Chen, F.; Tan, X.; Zheng, Z. Gegen Qinlian Decoction Restores the Intestinal Barrier in Bacterial Diarrhea Piglets by Promoting Lactobacillus Growth and Inhibiting the TLR2/MyD88/NF-κB Pathway. Biomed. Pharmacother. 2022, 155, 113719. [Google Scholar] [CrossRef]
  61. Zong, B.; Xiao, Y.; Wang, P.; Liu, W.; Ren, M.; Li, C.; Fu, S.; Zhang, Y.; Qiu, Y. Baicalin Weakens the Virulence of Porcine Extraintestinal Pathogenic Escherichia coli by Inhibiting the LuxS/AI-2 Quorum-Sensing System. Biomolecules 2024, 14, 452. [Google Scholar] [CrossRef] [PubMed]
  62. Zong, B.; Xiao, Y.; Ren, M.; Wang, P.; Fu, S.; Qiu, Y. Baicalin Weakens the Porcine ExPEC-Induced Inflammatory Response in 3D4/21 Cells by Inhibiting the Expression of NF-κB/MAPK Signaling Pathways and Reducing NLRP3 Inflammasome Activation. Microorganisms 2023, 11, 2126. [Google Scholar] [CrossRef] [PubMed]
  63. Fu, S.; Tian, X.; Peng, C.; Zhang, D.; Zhou, L.; Yuan, Y.; He, J.; Guo, L.; Qiu, Y.; Ye, C.; et al. Baicalin Inhibited PANX-1/P2Y6 Signaling Pathway Activation in Porcine Aortic Vascular Endothelial Cells Infected by Glaesserella parasuis. Heliyon 2024, 10, e23632. [Google Scholar] [CrossRef] [PubMed]
  64. Fu, S.; Zhuang, F.; Guo, L.; Qiu, Y.; Xiong, J.; Ye, C.; Liu, Y.; Wu, Z.; Hou, Y.; Hu, C.-A.A. Effect of Baicalin-Aluminum Complexes on Fecal Microbiome in Piglets. Int. J. Mol. Sci. 2019, 20, 2390. [Google Scholar] [CrossRef]
  65. Liu, Y.; Li, X.; Zhang, Z.; Zhang, J.; Xu, J.; Qiu, Y.; Ye, C.; Fu, S.; Wu, Z.; Hu, C.-A.A. Baicalin Protects Vascular Tight Junctions in Piglets during Glaesserella parasuis Infection. Front. Vet. Sci. 2021, 8, 671936. [Google Scholar] [CrossRef]
  66. Zhang, S.; Hu, B.; Xu, J.; Ren, Q.; Wang, Z.; Wang, S.; Dong, Y.; Yang, G. Baicalin Suppress Growth and Virulence-Related Factors of Methicillin-Resistant Staphylococcus aureus in Vitro and Vivo. Microb. Pathog. 2020, 139, 103899. [Google Scholar] [CrossRef]
  67. Fu, S.; Meng, Q.; Zhang, D.; Zuo, S.; He, J.; Guo, L.; Qiu, Y.; Ye, C.; Liu, Y.; Hu, C.-A.A. Effect of Baicalin on Transcriptome Changes in Piglet Vascular Endothelial Cells Induced by a Combination of Glaesserella parasuis and Lipopolysaccharide. DNA Cell Biol. 2021, 40, 776–790. [Google Scholar] [CrossRef]
  68. Fu, S.; Liu, J.; Xu, J.; Zuo, S.; Zhang, Y.; Guo, L.; Qiu, Y.; Ye, C.; Liu, Y.; Wu, Z.; et al. The Effect of Baicalin on microRNA Expression Profiles in Porcine Aortic Vascular Endothelial Cells Infected by Haemophilus parasuis. Mol. Cell. Biochem. 2020, 472, 45–56. [Google Scholar] [CrossRef]
  69. Guo, L.; Liu, J.; Zhang, Y.; Fu, S.; Qiu, Y.; Ye, C.; Liu, Y.; Wu, Z.; Hou, Y.; Hu, C.-A.A. The Effect of Baicalin on the Expression Profiles of Long Non-Coding RNAs and mRNAs in Porcine Aortic Vascular Endothelial Cells Infected with Haemophilus parasuis. DNA Cell Biol. 2020, 39, 801–815. [Google Scholar] [CrossRef]
  70. Ye, C.; Li, R.; Xu, L.; Qiu, Y.; Fu, S.; Liu, Y.; Wu, Z.; Hou, Y.; Hu, C.-A.A. Effects of Baicalin on Piglet Monocytes Involving PKC-MAPK Signaling Pathways Induced by Haemophilus parasuis. BMC Vet. Res. 2019, 15, 98. [Google Scholar] [CrossRef]
  71. Fu, S.; Liu, H.; Chen, X.; Qiu, Y.; Ye, C.; Liu, Y.; Wu, Z.; Guo, L.; Hou, Y.; Hu, C.-A.A. Baicalin Inhibits Haemophilus parasuis-Induced High-Mobility Group Box 1 Release during Inflammation. Int. J. Mol. Sci. 2018, 19, 1307. [Google Scholar] [CrossRef] [PubMed]
  72. Jin, J.; Chen, Y.; Wang, D.; Ma, L.; Guo, M.; Zhou, C.; Dou, J. The Inhibitory Effect of Sodium Baicalin on Oseltamivir-Resistant Influenza A Virus via Reduction of Neuraminidase Activity. Arch. Pharm. Res. 2018, 41, 664–676. [Google Scholar] [CrossRef] [PubMed]
  73. Zhao, P.; Huang, L.; Wang, H.; Wang, C.; Chen, J.; Yang, P.; Ni, M.; Chen, C.; Li, C.; Xie, Y.; et al. An Ultrasensitive High-Performance Baicalin Sensor Based on C3N4-SWCNTs/Reduced Graphene Oxide/Cyclodextrin Metal-Organic Framework Nanocomposite. Sens. Actuators B Chem. 2022, 350, 130853. [Google Scholar] [CrossRef]
  74. Sawadpongpan, S.; Jaratsittisin, J.; Hitakarun, A.; Roytrakul, S.; Wikan, N.; Smith, D.R. Investigation of the Activity of Baicalein towards Zika Virus. BMC Complement. Med. Ther. 2023, 23, 143. [Google Scholar] [CrossRef]
  75. Low, Z.X.; OuYong, B.M.; Hassandarvish, P.; Poh, C.L.; Ramanathan, B. Antiviral Activity of Silymarin and Baicalein against Dengue Virus. Sci. Rep. 2021, 11, 21221. [Google Scholar] [CrossRef]
  76. Qian, X.; Zhou, H.; Liu, Y.; Dong, J.; Tang, W.; Zhao, P.; Tang, H.; Jin, Y. Synthesis of Baicalein Derivatives and Evaluation of Their Antiviral Activity against Arboviruses. Bioorg. Med. Chem. Lett. 2022, 72, 128863. [Google Scholar] [CrossRef]
  77. Chang, W.; Wang, J.; Wu, F.; Zhang, H.; Yang, M. Antiviral Activity and Underlying Mechanisms of Baicalin against Porcine Reproductive and Respiratory Syndrome Virus in Vitro. Microb. Pathog. 2024, 193, 106712. [Google Scholar] [CrossRef]
  78. Shu, X.; Zhang, Y.; Zhang, X.; Zhang, Y.; Shu, Y.; Wang, Y.; Zhang, Z.; Song, C. Therapeutic and Immune-Regulation Effects of Scutellaria baicalensis Georgi Polysaccharide on Pseudorabies in Piglets. Front. Vet. Sci. 2024, 11, 1356819. [Google Scholar] [CrossRef]
  79. Leonova, G.; Shutikova, A.; Lubova, V.; Maistrovskaya, O. Inhibitory Activity of Scutellaria baicalensis Flavonoids against Tick-Borne Encephalitis Virus. Bull. Exp. Biol. Med. 2020, 168, 665–668. [Google Scholar] [CrossRef]
  80. Błach-Olszewska, Z.; Jatczak, B.; Rak, A.; Lorenc, M.; Gulanowski, B.; Drobna, A.; Lamer-Zarawska, E. Production of Cytokines and Stimulation Ofresistance to Viral Infection in Human Leukocytes by Scutellaria baicalensis Flavones. J. Interf. Cytokine Res. 2008, 28, 571–582. [Google Scholar] [CrossRef]
  81. Kitamura, K.; Honda, M.; Yoshizaki, H.; Yamamoto, S.; Nakane, H.; Fukushima, M.; Ono, K.; Tokunaga, T. Baicalin, an Inhibitor of HIV-1 Production in Vitro. Antivir. Res. 1998, 37, 131–140. [Google Scholar] [CrossRef] [PubMed]
  82. Li, B.; Fu, T.; Dongyan, Y.; Mikovits, J.A.; Ruscetti, F.W.; Wang, J.M. Flavonoid Baicalin Inhibits HIV-1 Infection at the Level of Viral Entry. Biochem. Biophys. Res. Commun. 2000, 276, 534–538. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, J.; Zhang, Z.; Chen, H.; Zhang, X.; Chen, X. Synthesis of baicalin derivatives and evaluation of their anti-human immunodeficiency virus (HIV-1) activity. Yao Xue Xue Bao 1998, 33, 22–27. [Google Scholar]
  84. Jiang, Y.; Ng, T.B.; Wang, C.R.; Zhang, D.; Cheng, Z.H.; Liu, Z.K.; Qiao, W.T.; Geng, Y.Q.; Li, N.; Liu, F. Inhibitors from Natural Products to HIV-1 Reverse Transcriptase, Protease and Integrase. Mini-Rev. Med. Chem. 2010, 10, 1331–1344. [Google Scholar] [CrossRef]
  85. Li, G.; Diao, X. Effect of Adding Fermented Scutellaria in Feed on Production Performance of Weaned Piglets. Chin. Feed 2012, 12, 27–28. [Google Scholar] [CrossRef]
  86. Jeong, J.S.; Kim, I.H. Effect of Probiotic Bacteria-fermented Medicinal Plants Ynura procumbens, Ehmannia glutinosa, Cutellaria baicalensis as Performance Enhancers in Growing Pigs. Anim. Sci. J. 2015, 86, 603–609. [Google Scholar] [CrossRef]
  87. Liu, W.; Pi, S.; Kim, I. Effects of Scutellaria baicalensis and Lonicera Japonicaextract Mixture Supplementation on Growth Performance, Nutrient Digestibility, Blood Profiles and Meat Quality in Finishing Pigs. Itali. J. Anim. Sci. 2016, 15, 446–452. [Google Scholar] [CrossRef]
  88. Gheisar, M.M.; Cheong, J.Y.; Zhao, P.; Kim, I.H. Evaluating the Influence of Dietary Phytogenic Blends on Gestating and Lactating Sows and Suckling Piglets. Anim. Prod. Sci. 2018, 58, 2071. [Google Scholar] [CrossRef]
  89. Chang, C.; Chen, Y.; Chiou, M.; Su, C.; Chen, D.S.; Tsai, C.-E.; Yu, B.; Hsu, Y.-M. Application of Scutellariae Radix, Gardeniae Fructus, and Probiotics to Prevent Salmonella enterica Serovar Choleraesuis Infection in Swine. Evidence-Based Complement. Altern. Med. 2013, 2013, 568528. [Google Scholar] [CrossRef]
  90. Zhang, S.; Tang, S.; Zhong, R.; Chen, L.; Zhang, H. PSVIII-26 Baicalin Alleviates Lincomycin-Induced Intestinal Injury by Regulating Gut Microbiota in Weaned Pigs. J. Anim. Sci. 2023, 101, 662–663. [Google Scholar] [CrossRef]
  91. Lu, Q.; Wang, N.; Wen, D.; Guo, P.; Liu, Y.; Fu, S.; Ye, C.; Wu, Z.; Qiu, Y. Baicalin Attenuates Lipopolysaccharide-Induced Intestinal Inflammatory Injury via Suppressing PARP1-Mediated NF-κB and NLRP3 Signalling Pathway. Toxicon 2024, 239, 107612. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, W.; Yun, H.; Pi, S.; Kim, I. Supplementing Lactation Diets with Herbal Extract Mixture during Summer Improves the Performance of Sows and Nursing Piglets. Ann. Anim. Sci. 2017, 17, 835–847. [Google Scholar] [CrossRef]
  93. Zhao, P.; Li, H.; Lei, Y.; Li, T.; Kim, S.; Kim, I. Effect of Fermented Medicinal Plants on Growth Performance, Nutrient Digestibility, Fecal Noxious Gas Emissions, and Diarrhea Score in Weanling Pigs. J. Sci. Food Agric. 2016, 96, 1269–1274. [Google Scholar] [CrossRef] [PubMed]
  94. Liao, Y.; Cui, Y.; Hu, X.; Zhen, Y.; Lv, H.; Huang, D.; Deng, Y.; Deng, X. Treatment Test of Skullcap Injection on the Artificial Infection of Swine Edema Disease. Chin. J. Vet. Sci. 2016, 5, 4. [Google Scholar] [CrossRef]
  95. Zhang, S.; Tang, S.; Liu, Z.; Lv, H.; Cai, X.; Zhong, R.; Chen, L.; Zhang, H. Baicalin Restore Intestinal Damage after Early-Life Antibiotic Therapy: The Role of the MAPK Signaling Pathway. Pharmacol. Res. 2024, 204, 107194. [Google Scholar] [CrossRef]
  96. Wang, L.; Huo, B.; Huang, L.; Che, L.; Feng, B.; Lin, Y.; Xu, S.; Wu, D.; Fang, Z. Dietary Supplementation with a Mixture of Herbal Extracts during Late Gestation and Lactation Improves Performance of Sows and Nursing Piglets through Regulation of Maternal Metabolism and Transmission of Antibodies. Front. Vet. Sci. 2022, 9, 1026088. [Google Scholar] [CrossRef]
  97. Guo, Q.; Xuan, M.; Luo, Z.; Wang, J.; Han, S.; Ri, M.; Choe, Y.; Hwang, K.; Yin, X.; Kang, J. Baicalin Improves the in Vitro Developmental Capacity of Pig Embryos by Inhibiting Apoptosis, Regulating Mitochondrial Activity and Activating Sonic Hedgehog Signaling. Mol. Hum. Reprod. 2019, 25, 538–549. [Google Scholar] [CrossRef]
  98. Ling, X.; Xiang, Y.; Chen, F.; Tang, Q.; Zhang, W.; Tan, X. Intestinal Absorption Differences of Major Bioactive Compounds of Gegenqinlian Decoction between Normal and Bacterial Diarrheal Mini-Pigs in Vitro and in Situ. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2018, 1083, 93–101. [Google Scholar] [CrossRef]
  99. Baradaran Rahimi, V.; Askari, V.R.; Hosseinzadeh, H. Promising Influences of Scutellaria baicalensis and Its Two Active Constituents, Baicalin, and Baicalein, against Metabolic Syndrome: A Review. Phytother. Res. 2021, 35, 3558–3574. [Google Scholar] [CrossRef]
  100. Ge, P.; Qi, Y.; Qu, S.; Zhao, X.; Ni, S.; Yao, Z.-Y.; Guo, R.; Yang, N.-Y.; Zhang, Q.-C.; Zhu, H.-X. Potential Mechanism of S. baicalensis on Lipid Metabolism Explored via Network Pharmacology and Untargeted Lipidomics. Drug Des. Dev. Ther. 2021, 15, 1915–1930. [Google Scholar] [CrossRef]
  101. Chen, Q.; Liu, M.; Yu, H.; Li, J.; Wang, S.; Zhang, Y.; Qiu, F.; Wang, T. Scutellaria baicalensis Regulates FFA Metabolism to Ameliorate NAFLD through the AMPK-Mediated SREBP Signaling Pathway. J. Nat. Med. 2018, 72, 655–666. [Google Scholar] [CrossRef] [PubMed]
  102. Na, H.-Y.; Lee, B.-C. Scutellaria baicalensis Alleviates Insulin Resistance in Diet-Induced Obese Mice by Modulating Inflammation. Int. J. Mol. Sci. 2019, 20, 727. [Google Scholar] [CrossRef] [PubMed]
  103. Ansari, A.; Bose, S.; Lim, S.-K.; Wang, J.-H.; Choi, Y.-H.; Kim, H. Combination of Scutellaria baicalensis and Metformin Ameliorates Diet-Induced Metabolic Dysregulation in Mice via the Gut-Liver-Brain Axis. Am. J. Chin. Med. 2020, 48, 1409–1433. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Active ingredients and target genes of SFs. (A) SFs’ 3D structure; (B) SFs–typical active ingredients–targets network. Green represents the SFs, orange represents the typical active ingredients of SFs, and blue represents the target genes of the active ingredients.
Figure 1. Active ingredients and target genes of SFs. (A) SFs’ 3D structure; (B) SFs–typical active ingredients–targets network. Green represents the SFs, orange represents the typical active ingredients of SFs, and blue represents the target genes of the active ingredients.
Ijms 26 03703 g001
Figure 2. SFs–targets–inflammation–KEGG pathway network. Dark green represents the SFs, yellow represents the typical active ingredients of SFs, purple represents the disease, light green represents the target genes of SFs’ anti-inflammatory effects, and blue represents the KEGG pathway of SFs’ anti-inflammatory effects.
Figure 2. SFs–targets–inflammation–KEGG pathway network. Dark green represents the SFs, yellow represents the typical active ingredients of SFs, purple represents the disease, light green represents the target genes of SFs’ anti-inflammatory effects, and blue represents the KEGG pathway of SFs’ anti-inflammatory effects.
Ijms 26 03703 g002
Figure 3. SFs–targets–oxidative stress–KEGG pathway network. Yellow represents the typical active ingredients of SFs, pink represents the disease, light green represents the target genes of SFs’ antioxidant effects, and blue represents the KEGG pathway of SFs’ antioxidant effects.
Figure 3. SFs–targets–oxidative stress–KEGG pathway network. Yellow represents the typical active ingredients of SFs, pink represents the disease, light green represents the target genes of SFs’ antioxidant effects, and blue represents the KEGG pathway of SFs’ antioxidant effects.
Ijms 26 03703 g003
Figure 4. The underlying antiviral mechanisms of SFs include direct interference with the viral life cycle, improvement in host immunity, and direct binding to viral proteins.
Figure 4. The underlying antiviral mechanisms of SFs include direct interference with the viral life cycle, improvement in host immunity, and direct binding to viral proteins.
Ijms 26 03703 g004
Table 1. Active ingredients of Scutellaria baicalensis Georgi.
Table 1. Active ingredients of Scutellaria baicalensis Georgi.
Mol IDMolecule NameStructureMWAlogpHdonHaccOB (%)DL
MOL001689AcacetinIjms 26 03703 i001284.282.592534.970.24
MOL000173WogoninIjms 26 03703 i002284.282.592530.680.23
MOL000228(2R)-7-hydroxy-5-methoxy-2-phenylchroman-4-oneIjms 26 03703 i003270.32.821455.230.2
MOL002714BaicaleinIjms 26 03703 i004270.252.333533.520.21
MOL0029085,8,2′-Trihydroxy-7-methoxyflavoneIjms 26 03703 i005300.282.323637.010.27
MOL0029095,7,2,5-tetrahydroxy-8,6-dimethoxyflavoneIjms 26 03703 i006376.342.024933.820.45
MOL002910CarthamidinIjms 26 03703 i007288.272.034641.150.24
MOL0029112,6,2′,4′-tetrahydroxy-6′-methoxychaleoneIjms 26 03703 i008302.32.624669.040.22
MOL002913Dihydrobaicalin_qtIjms 26 03703 i009272.272.33540.040.21
MOL002914EriodyctiolIjms 26 03703 i010288.272.034641.350.24
MOL002915SalvigeninIjms 26 03703 i011328.342.821649.070.33
MOL0029175,2′,6′-Trihydroxy-7,8-dimethoxyflavoneIjms 26 03703 i012330.312.33745.050.33
MOL0029255,7,2′,6′-TetrahydroxyflavoneIjms 26 03703 i013286.252.074637.010.24
MOL002926Dihydrooroxylin AIjms 26 03703 i014286.32.552538.720.23
MOL002927Skullcapflavone IIIjms 26 03703 i015374.372.542869.510.44
MOL002928Oroxylin aIjms 26 03703 i016284.282.592541.370.23
MOL002932PanicolinIjms 26 03703 i017314.312.572676.260.29
MOL0029335,7,4′-Trihydroxy-8-methoxyflavoneIjms 26 03703 i018300.282.323636.560.27
MOL002934NeobaicaleinIjms 26 03703 i019374.372.5428104.340.44
MOL002937DihydrooroxylinIjms 26 03703 i020286.32.552566.060.23
MOL000525NorwogoninIjms 26 03703 i021270.252.333539.40.21
MOL0005525,2′-Dihydroxy-6,7,8-trimethoxyflavoneIjms 26 03703 i022344.342.552731.710.35
MOL000073EpicatechinIjms 26 03703 i023290.291.925648.960.24
MOL001458CoptisineIjms 26 03703 i024320.343.250430.670.86
MOL002897EpiberberineIjms 26 03703 i025336.393.450443.090.78
MOL008206MoslosooflavoneIjms 26 03703 i026298.312.841544.090.25
MOL0122455,7,4′-trihydroxy-6-methoxyflavanoneIjms 26 03703 i027302.32.283636.630.27
MOL0122465,7,4′-trihydroxy-8-methoxyflavanoneIjms 26 03703 i028302.32.283674.240.26
MOL012266RivularinIjms 26 03703 i029344.342.552737.940.37
Note: Mol ID, molecule ID; MW, molecular weight; Alogp, lipophilicity; Hdon, hydrogen bond donor; Hacc, hydrogen bond acceptor; OB, oral bioavailability; DL, drug-likeness.
Table 2. Antibacterial activities of SFs in the swine husbandry industry.
Table 2. Antibacterial activities of SFs in the swine husbandry industry.
CompoundAntibacterial ActivityEffective Concentration/MICRelated MechanismReference
BaicalinETEC1 µg/mLInhibits bacterial adhesion and activates NF-κB signaling pathway[19]
BaicalinH. parasuis12.5–100 μg/mLSuppresses NLRP3 inflammasome and NF-κB signaling[57]
SBE (≥ 12% baicalin)E. coli K881000 mg/kg BWInhibits NF-κB and P38 signaling pathways[58]
GQDLactobacillus1.22 g/kgInhibits TLR2/MyD88/
NF-κB pathway
[60]
BaicalinExPEC1600 µg/mLInhibits LuxS/AI-2
quorum-sensing system
[61]
BaicalinExPEC1600 µg/mLInhibits the expression of NF-κB/MAPK signaling pathways and reduces NLRP3
inflammasome activation
[62]
BaicalinG. parasuis25–100 µMInhibits PANX-1/P2Y6 signaling pathway[63]
BaicalinH. parasuis12.5–100 μg/mLModulates apoptosis via RAGE, MAPK, and AP-1[64]
BaicalinG. parasuis25–100 mg/kg BWProtects vascular tight junctions[65]
BaicalinS.aureus760 μg/mLAttenuates biofilm formation and downregulates expression of virulence-related factors[66]
BaicalinG. parasuis50 μg/mLChanges lncRNA expression[67]
BaicalinH. parasuis50 μg/mLChanges microRNA expression profiles[68]
BaicalinH. parasuis50 μg/mLChanges expression profiles of long non-coding RNAs and mRNAs[69]
BaicalinH. parasuis80 mg/kg·BWReverses apoptosis through regulating PKC-MAPK signaling pathway[70]
BaicalinG. parasuis25–100 mg/kg BWInhibits activation of HMGB1, cell apoptosis, and
MAPK signaling pathway
[71]
BW, body weight; ExPEC, extra-intestinal pathogenic Escherichia coli; ETEC, enterotoxigenic Escherichia coli; S. aureus, Staphylococcus aureus; SBE, Scutellaria baicalensis extracts; E. coli K88, Escherichia coli K88; G. parasuis, Glaesserella parasuis; H. parasuis, Haemophilus parasuis; GQD, Gegen Qinlian decoction.
Table 3. The application of SFs in healthy swine husbandry.
Table 3. The application of SFs in healthy swine husbandry.
CompoundEffective ConcentrationType of AnimalAgeMain EffectsReferences
Mixed herbs containing S. baicalensis1000 mg/kgSows and suckling piglets80–107 d of gestationImproves antioxidant capacity, liver function, colostrum quality, and immunity in sows; improves growth performance and immunity in piglets[4]
Mixed fermented plant containing S. baicalensis-Weaned piglets25 dModifies gut microbial composition[5]
Scutellaria baicalensis extracts1000 mg/kg dietWeaned piglets24 ± 2 dAttenuates diarrhea[58]
Baicalin–aluminum complexes1.36 g/d, 3 dPiglets10 dModulates gut microbiome composition of piglets with diarrhea[64]
Baicalin5–20 μg/mLMARC145 cells and PAMs-Inhibits PRRSV infection in vitro[77]
Fermented S. baicalensi1.5 mg/kg dietWeaned piglets28–56 dEnhances appetite, increases average daily intake, reduces feed-to-weight ratio and diarrhea rate, and improves feed reward[85]
Mixed fermented plant containing S. baicalensis-Growing pigs25.50 ± 2.50 kg BWImproves growth performance and nutrient digestibility; reduces noxious gas emissions of excret[86]
Mixed herbs containing S. baicalensis0.025% and 0.05%Finishing pigs44.2 ± 2.23 kg BWImproves growth performance and nutrient digestibility; decreases serum cortisol levels; benefits meat quality[87]
Mixed herbs containing S. baicalensis-Pregnant and lactating sows-Decreases weight loss and improves litter performance[88]
Mixed herbs containing Baicalin89.74 mg/gWeaned piglets28 dPrevents infection, enhances immunity, and adjusts intestinal microbial composition[89]
Baicalin1 g/kg for week and 500 mg/kg feed for 2 weeksWeaned piglets21 dAlleviates intestinal injury[90]
Baicalin100 μg/mLPig intestinal epithelial cell line J2-Attenuates intestinal inflammatory injury[91]
Mixed herbs containing S. baicalensis10 g/dLactating sows under heat stress and piglets-Improves feed intake, digestibility of dry matter, piglets’ weaning weight, and ADG; decreases backfat loss, serum cortisol levels, and diarrhea[92]
S. baicalensis extract-Weaned piglets-Improves growth performance and nutrient digestibility; decreases fecal noxious gas emissions and alleviates diarrhea[93]
Baicalin10 mg/kg BW, 1 time/d, 5 d; intramuscular injectionWeaned piglets28–35 dPrevents swine edema disease[94]
Baicalin500 mg/kgWeaned piglets-Restores intestinal health[95]
Mixed herbs containing S. baicalensis1000 mg/kgSows and suckling piglets-Improves growth performance, maternal metabolism, and transmission of antibodies[96]
BW, body weight; PAMs, porcine alveolar macrophages.
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

Wu, J.; Qiu, Y.; Tian, M.; Wang, L.; Gao, K.; Yang, X.; Jiang, Z. Flavonoids from Scutellaria baicalensis: Promising Alternatives for Enhancing Swine Production and Health. Int. J. Mol. Sci. 2025, 26, 3703. https://doi.org/10.3390/ijms26083703

AMA Style

Wu J, Qiu Y, Tian M, Wang L, Gao K, Yang X, Jiang Z. Flavonoids from Scutellaria baicalensis: Promising Alternatives for Enhancing Swine Production and Health. International Journal of Molecular Sciences. 2025; 26(8):3703. https://doi.org/10.3390/ijms26083703

Chicago/Turabian Style

Wu, Jing, Yueqin Qiu, Min Tian, Li Wang, Kaiguo Gao, Xuefen Yang, and Zongyong Jiang. 2025. "Flavonoids from Scutellaria baicalensis: Promising Alternatives for Enhancing Swine Production and Health" International Journal of Molecular Sciences 26, no. 8: 3703. https://doi.org/10.3390/ijms26083703

APA Style

Wu, J., Qiu, Y., Tian, M., Wang, L., Gao, K., Yang, X., & Jiang, Z. (2025). Flavonoids from Scutellaria baicalensis: Promising Alternatives for Enhancing Swine Production and Health. International Journal of Molecular Sciences, 26(8), 3703. https://doi.org/10.3390/ijms26083703

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

Article Metrics

Back to TopTop