Next Article in Journal
Effectiveness and Safety of Over-the-Counter Tooth-Whitening Agents Compared to Hydrogen Peroxide In Vitro
Next Article in Special Issue
Hydroxytyrosol and Its Potential Uses on Intestinal and Gastrointestinal Disease
Previous Article in Journal
Sly-miR398 Participates in Cadmium Stress Acclimation by Regulating Antioxidant System and Cadmium Transport in Tomato (Solanum lycopersicum)
Previous Article in Special Issue
Dietary Habits and Nutrient Deficiencies in a Cohort of European Crohn’s Disease Adult Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Bioactive Components from Scutellaria baicalensis Georgi in Inflammatory Bowel Disease and Colorectal Cancer: A Review

Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 1954; https://doi.org/10.3390/ijms24031954
Submission received: 27 December 2022 / Revised: 16 January 2023 / Accepted: 16 January 2023 / Published: 19 January 2023
(This article belongs to the Special Issue Dietary Bioactive Components in Inflammatory Bowel Disease)

Abstract

:
Scutellaria baicalensis Georgi (SBG), an herbal medicine with various biological activities, including anti-inflammatory, anticancer, antiviral, antibacterial, and antioxidant activities, is effective in treatment of colitis, hepatitis, pneumonia, respiratory infections, and allergic diseases. This herbal medicine consists of major active substances, such as baicalin, baicalein, wogonoside, and wogonin. Inflammatory bowel disease (IBD) comprises a group of inflammatory conditions of the colon and small intestine, with Crohn’s disease and ulcerative colitis being the main types. IBD can lead to serious complications, such as increased risk of colorectal cancer (CRC), one of the most common cancers worldwide. Currently, there is no cure for IBD, and its incidence has been increasing over the past few decades. This review comprehensively summarizes the efficacy of SBG in IBD and CRC and may serve as a reference for future research and development of drugs for IBD and cancer treatment.

1. Introduction

Inflammatory bowel disease (IBD), a type of chronic gastrointestinal inflammation, mainly includes Crohn’s disease (CD) and ulcerative colitis (UC) [1]. CD affects the small and large intestines, mouth, esophagus, stomach, and anus, whereas UC mainly affects the colon and rectum [2]. IBD is characterized by gastrointestinal discomfort, weight loss, bloody diarrhea, and periods of infiltration of neutrophils and macrophages, which release cytokines, proteolytic enzymes, and reactive species that cause ulceration and inflammation in the intestinal mucosa [3]. The prevalence and incidence of IBD increased remarkably during the second half of the 20th century, and, since the beginning of the 21st century, IBD is considered one of the most common gastrointestinal diseases as its incidence has increased in newly industrialized countries [4,5].
In the past, IBD was thought to occur primarily in white people in high-income countries, such as those of North America, Europe, and Australia. The incidence of IBD in these countries has leveled in recent years; however, its prevalence is still much higher than that in other countries. According to data from 2017, there were more than 6 million IBD cases worldwide, with nearly a quarter of them residing in the United States [6]. The highest prevalence of IBD was reported in Europe (UC 505 per 100,000 people in southeastern Norway; CD 322 per 100,000 people in Hessen, Germany) and North America (UC 286.3 per 100,000 people in Olmstead County, USA; CD 318.5 per 100,000 people in Nova Scotia, Canada) [4]. Patients with IBD have notably increased risk of colorectal cancer (CRC), primarily owing to the pro-neoplasmic effects of chronic intestinal inflammation [7,8,9]. According to a recent study, the risk of developing CRC in patients with IBD depends on the patient’s history of IBD and can increase by 2% after 10 years, 8% after 20 years, and 18% after 30 years [8,9]. However, owing to successful CRC surveillance programs and improved control of mucosal inflammation, its incidence has declined over the past 30 years. Thus, all major professional societies concur that IBD-associated CRC surveillance must occur regularly in the general population [9].
CRC, also known as colon or rectal cancer, is the third leading cause of cancer deaths in both men and women and has newly become the third most common cancer diagnosed in the United States [10]. According to South Korean projections for 2022, CRC ranks third in the expected mortality rate for men and second among women, and, in South Korea, it became the fourth most common cancer diagnosed in men and the third in women [11]. Currently, effective treatment methods for CRC include chemotherapy, surgery, radiation, or a combination of chemotherapy and radiotherapy. More than 50% of CRC patients who undergo surgical resection are cured, but 40–50% of these patients eventually relapse, and the likelihood of the patient returning to work is very low [12]. Drug resistance and chemotherapy-related toxic side effects are major causes of chemotherapy failure or discontinuation in CRC patients. As a result, there are problems in extending the survival period of CRC patients, reducing recurrence and metastasis rates, and improving quality of life, so alternative treatments are being sought [13].
Herbal plants have long been used to prevent and treat various diseases. Today, many people still rely on herbal nutraceuticals for primary healthcare. More than 50% of drugs currently in clinical use are of natural origin [14]. Scutellaria baicalensis Georgi (SBG) has been one of the most extensively used medicinal herbs in China for over 2000 years. The dried root of this plant has been used by the Chinese in traditional medicine called Scutellariae Radix or Huang-Qin and is officially listed in the Chinese Pharmacopoeia (2020) [15]. In addition, SBG is also listed in the European Pharmacopoeia (EP 9.0) and British Pharmacopoeia (BP 2018) [16]. Dried roots (Huang Qin) have a long history of medicinal use for treatment of inflammatory, gastrointestinal, and respiratory diseases [17]. A total of 56 flavonoids have been isolated from SBG, and the most representative components are baicalin, baicalein, wogonoside, and wogonin [18,19]. In this review, we present the effects of these components on IBD, IBD-associated CRC, and CRC.

2. IBD

2.1. CD and UC

IBD is caused by an interaction between genetic and environmental factors affecting the immune response [20]. Although the exact cause of IBD is unclear, the etiology is generally accepted as multifactorial and includes genetic predisposition, disorders of the gastrointestinal microbiota, dysfunction of the mucosal barrier, unregulated immune responses, and environmental and lifestyle factors [21,22].
CD can affect any part of the gastrointestinal tract, from the mouth to the anus, and usually affects individuals aged between 15 and 30 years. CD is discontinuous and can affect any layer of the intestinal wall. Patients with CD have symptoms that include abdominal fever, pain, bowel obstruction, or diarrhea with passage of mucus or blood [23].
UC is associated with persistent colonic inflammation, usually starting in the rectum and extending to the proximal portion of the colon, primarily affecting the inner epithelial layer of the colon [21]. It mainly affects adults aged 30–40 years and can weaken [24]. UC can be further classified into three types according to the degree of colonic involvement: proctitis, left-sided colitis, and diffuse colitis. Proctitis afflicts 30–60% of patients, and its symptoms include rectal bleeding, tension, and urgency. Left-side colitis affects 16–45% of patients with symptoms of proctitis, diarrhea, and abdominal cramps. The most severe form of UC is widespread colitis, affecting 15–35% of patients. This form can present with symptoms of left-sided colitis other than systemic symptoms, fatigue, and fever [23]. In addition to gastrointestinal symptoms, 25–40% of people with IBD may develop symptoms primarily associated with the skin, joints, eyes, and liver [25].

2.2. Onset of CRC in Patients with IBD

Although patients with IBD are at an increased risk of developing CRC, the overall incidence of IBD-associated CRC has declined in recent decades in Western countries. As demonstrated in previous studies, the risk of CRC in IBD increases with long-term, family history of CRC, coexisting primary sclerosing cholangitis, extent of colitis, and degree of inflammation [26]. However, the pathogenesis of IBD-associated CRC is not well understood. Similar to sporadic CRC, IBD-associated CRC results from sequential episodes of genomic changes [27]. Several interrelated pathways, including immune responses by mucosal inflammatory mediators, gut microbiota, and oxidative stress, are also associated with pathogenesis of IBD-associated CRC. Persistent colonic inflammation is a risk factor for CRC [26]. Sporadic CRC is defined by sequential histological and genetic changes, known as adenoma-carcinoma sequences. In contrast, IBD-associated CRC develops via the ‘inflammatory-dysplastic-carcinoma’ sequence [27,28,29]. Mutations contributing to pathogenesis of IBD-associated CRC are similar to those associated with sporadic CRC. However, multiple lines of evidence suggest that IBD-associated CRC may arise through different mechanisms of tumorigenesis compared with sporadic CRC. Loss of adenomatous polyposis coli (APC) occurs early in the development of sporadic CRC, whereas it is usually a late event in IBD-associated CRC. TP53 mutations appear early in IBD-associated CRC, even before onset of dysplasia, but late in sporadic CRC [26,28,29,30].

2.3. IBD Treatment

The goal of IBD treatment is to reduce inflammation, which causes signs and symptoms. In the best case, IBD treatment can lead to symptom relief and reduced risk of long-term remission and complications [1]. Conventional treatment includes administration of aminosalicylates (sulfasalazine and 5-aminosalicylic acid), corticosteroids, immunomodulators (thiopurines, methotrexate, calcineurin inhibitors, and Janus kinase inhibitors), biologics such as pro-inflammatory cytokines (tumor necrosis factor-α (TNF-α), interleukin (IL)-12 and IL-23 inhibitors, and integrin antagonists), and, if necessary, other common measures or surgical resection [31].
However, these drugs have several drawbacks and adverse effects. Nonbiological treatments, such as aminosalicylates, thiopurines, and steroids, provide symptomatic improvement but do not alter the overall disease course in IBD [32]. Among immunomodulators, tacrolimus, a calcineurin inhibitor, can cause diabetes [33]. Biological therapies, including monoclonal antibodies targeting TNF-α, such as adalimumab, infliximab, golimumab, and certolizumab pegol, were introduced in the late 1990s to induce and maintain remission [34,35,36,37,38,39]. However, roughly one-third of patients with IBD are first-line non-responders to TNF-α inhibitor induction therapy [38,39,40,41], and the others become second-line non-responders during TNF-α inhibitor maintenance therapy [42,43,44]. Although the safety profiles of TNF-α inhibitors are generally acceptable, these drugs have side effects associated with infections and malignancies [45,46,47]. In addition, these biological therapies have the disadvantage of being expensive. As research intensifies, new therapies for treatment of IBD are emerging, focusing on apheresis therapy, stem cell transplantation, improvement of intestinal microecology, and exosome therapy [31]. These unapproved new therapies are often applied in research protocols but are limited by their unclear effects on IBD. Therefore, more studies, including long-term data, are needed to minimize the risk and optimize treatment outcomes. Identifying new drugs that are effective, economical, and less toxic has become a new goal.

2.4. Experimental Models of IBD

For decades, researchers have attempted to determine the causes and pathology of IBD. Researchers have developed many in vitro, ex vivo, and in vivo model systems to understand its causes and properly identify therapies [2].

2.4.1. In Vitro and Ex Vivo IBD Models

In vitro cell line models included Caco-2, T84, and HT29 cell lines, organoids, Caco-2/HT29-MTX co-culture, and intestinal-immunological cultures [peripheral blood mononuclear cells (PBMCs), and macrophage cell lines from either human (THP-1) or murine origin (RAW 246.7)]. Caco-2 cells are an immortalized cell line of human colorectal adenocarcinoma [48]. Differentiated Caco-2 cells produce the same characteristics and specific digestive enzymes (e.g., peptidase and lactase) as small-bowel enterocytes, such as brush-border microvilli [49]. They can also produce various cytokines (IL-6, -8, and -15, TNF-α, and thymic stromal lymphopoietin) [50,51,52]. In addition, absorption-related studies are easy to perform [53,54,55]. HT29 cells are also human-colon-cancer-derived cells releasing cytokines similar to those released by Caco-2 cells [56]. HT29 cell lines produce similar cellular differentiation patterns and typical digestive enzymes (e.g., peptidase and lactase) to Caco-2 cells [57]. However, an important difference between Caco-2 and HT29 cell lines is that HT29 cells produce reasonably high levels of mucins [58].
Organoids are produced by pluripotent or adult stem cells. They are three-dimensional and can be differentiated into different cell types to perform related organ functions [59]. Organoids mimic the physiology of intestinal cells [60,61] and ecology of gut microbes [62,63]. They also enable investigation of the innate immune system [64]. Patient-derived organoids can also be used [65,66]. Researchers have found that exposure of fast-growing HT29 cells to high doses of methotrexate (MTX) results in development of mucus-secreting differentiated cells [67]. Therefore, the co-culture of Caco-2 and HT29-MTX can generate mucosa and comprise an excellent system to study microbial adhesion and colonic permeability [68]. This may also increase mucosal production [69]. Another cell line frequently used in intestinal research studies, the T84 cell line, contains microvilli on its surface and can develop into crypt-like cells when activated by modifying growth factors [70]. Similar to Caco-2 cells, T84 cells can rapidly develop into an absorptive layer of epithelial cells [71].
To simulate the interaction between intestinal and immune cells, researchers have used primary cells, such as PBMC, and THP-1 or RAW 246.7 macrophage cell lines, the main immune cells used to create various co-culture systems with intestinal cell lines. Human PBMCs are obtained from the peripheral blood of healthy donors or buffy coats and are used to study the effects of diet on immune cells [2]. The THP-1 cell line is a human leukemia cell line that was initially obtained from patients with acute monocytic leukemia. THP-1 cells are similar to macrophages and primary monocytes in terms of differentiation [72]. The RAW 246.7 cell line was obtained from mouse tumors using the Abelson leukemia virus. These cells can accurately simulate the intestinal environment and can express toll-like receptors that play an important role in pathogenesis of IBD [73].
Several ex vivo models have been used to overcome the aforementioned disadvantages of in vitro IBD models. The Ussing chamber evaluates ion transport in tissues, such as intestinal mucosa. It is used to study intestinal permeability and host–microbiome interactions using both animal and human tissues [74]. An inverted gut sac model was used to fully understand the kinetics and mechanisms of drug absorption; it has a mucous layer, intact intestinal tissue, and a large surface area [75]. Microfluidic gut-on-chip models have emerged as a new approach for studying gut function by integrating multiple cell types and the gut microbiome into one system [76]. This model enables co-culture of primary intestinal cells and macrophages; pluripotent human intestinal stem cells can also be used [77].

2.4.2. In Vivo IBD Models

Experimental murine models of human IBD display immune pathological signatures similar to those of CD or UC [78]. These models include chemically induced (dextran sulfate sodium (DSS), trinitrobenzene sulfonic acid (TNBS), and oxazolone), spontaneous mutant [C3H/HeJBir (C3Bir) and SAMP1/Yit], and genetically engineered models (IL-10−/− knockout mice).
The DSS-induced colitis model is the most widely used colitis experimental murine model, in which DSS (molecular weight 40–50 kDa) is administered via drinking water [78]. DSS interrupts the epithelial barrier, exposes the lamina propria to luminal contents, and causes vascular and mucosal damage, subsequently resulting in activation of the inflammatory pathways [79]. The acute inflammatory response in DSS-induced colitis is represented by increased expression of IL-1β, IL-6, and TNF-α in the colon [78]. DSS-induced colitis is frequently used because of the simplicity of the administration process, ease of dose control to determine the severity of colitis, and adjustable duration to study the inflammatory or recovery process [80]. TNBS, used in chemically induced experimental models, is an oxidative nitroaryl compound administered intrarectally to animals to induce IBD. It causes colon damage that leads to necrosis associated with inflammation. The immune response is T helper type 1 (Th1)-mediated, characterized by infiltration of neutrophils, CD4+ T cells, and macrophages. Inflammation that spreads transversely results in transmural colitis [81]. Oxazolone, used in another chemically induced experimental model, is a hapten compound that causes acute colitis in animals when administered intrarectally in combination with ethanol. Oxazolone causes acute superficial mucosal inflammation in the distal colon, leading to a Th2-cell-mediated immune response with increased production of interleukins; the immune response type distinguishes this model from the TNBS-induced model, characterized by a Th1-mediated response [82].
Spontaneous mutant IBD mouse models develop IBD because of spontaneously occurring mutations but not gene suppression or transgenic overexpression. C3H/HeJBir (C3Bir) mice with a missense mutation in the third exon of the toll-like receptor 4 (TLR4) gene develop spontaneous inflammation in the cecum and colon. The SAMP1/Yit mouse variant showed elevated interferon-gamma and TNF-α levels, resulting in CD-like ileitis at approximately 10 weeks of age. Overall extension of stem cells in the crypt epithelium with severe colitis was observed in these mice [83].
Genetically engineered mouse models spontaneously develop colitis or ileitis. The best-known model is the IL-10−/− knockout mouse, in which spontaneous colitis develops because of the inability of regulatory T (Treg) cells to produce IL-10. Specifically, inflammation of the colon is characterized by inflammatory infiltration of lymphocytes, macrophages, and neutrophils [82].

3. SBG

SBG, a flowering plant belonging to the Lamiaceae family, is widely distributed in China, Korea, Japan, Mongolia, and Russia [14]. Many scientific studies have demonstrated a wide range of pharmacological effects of SBG, including anti-inflammatory [84,85], antiviral [86], antibacterial [87], antioxidant [88], anticancer [89,90], and immunomodulatory activities [86]. The root of SBG, designated Scutellariae Radix or Huang Qin, is one of the most popular herbal medicines in China. SBG is particularly notable for its high flavonoid content. SBG contains four major flavones: baicalin (7-glucuronic acid, 5,6-dihydroxy-flavone), baicalein (5,6,7-trihydroxyfavone), wogonoside (wogonin-7-glucuronic acid), and wogonin (5,7-dihydroxy-8-methoxyflavone) at approximately 10.11%, 5.41%, 3.55%, and 1.3%, respectively, of the dry matter [91,92] (Figure 1A–D). Major aglycones baicalein and wogonin can be produced from glycosides baicalin and wogonoside, respectively [93]. Baicalin is hydrolyzed into baicalein by the action of the gut microbiota and reconverted back to baicalin through phase 2 metabolism in the liver [94,95]. Deglycosylation of wogonoside and its conversion to wogonin can be achieved via transformation of the enteric microbiome, enzyme catalysis, or herbal processing, such as soaking in water [96].
According to the Human Clinical Research Database, clinicaltrials.gov (accessed on 16 December 2022), a study on SBG was reported. NCT02828540 is a study of HT047, a complex herbal extract from the roots of Pueraria lobata and SBG. The main active compounds in HT047 are puerarin, daidzein, baicalin, baicalein, and wogonin. This study is the first human trial to explore the efficacy and safety of HT047 in patients with acute ischemic stroke. The results provided appropriate doses and evidence for the therapeutic effects of HT047 on stroke recovery [97]. Baicalin has been developed as a new drug (Huangqingan Tablets, manufactured by several companies, including Shanghai Hutchison Pharmaceuticals (Shanghai, China) and Jingfukang Pharmaceutical Group Co. Ltd., Chengde, China) to treat acute and chronic hepatitis. The total extract of the leaves and stems of SBG has also been developed as a new drug (Huangqin Jingye Jiedu Capsules) to treat sore throat [18].

4. Therapeutic Benefits of SBG

4.1. Effect of SBG on IBD

The anti-inflammatory effects of baicalin, baicalein, wogonoside, and wogonin have been reported. The effects of baicalin (Table 1), baicalein (Table 2), wogonoside (Table 3), and wogonin (Table 4) on IBD are summarized.

4.1.1. Effect of SBG on COX-2

Cyclooxygenases-1 (COX-1) and COX-2 are antioxidant enzymes catalyzing conversion of free arachidonic acid to prostaglandin (PG) H2, which plays a significant role in inflammatory and pain responses. COX-1, first purified in 1976, is expressed in most tissues, whereas COX-2 was recently identified as a major inducible isoform producing high amounts of PGH2 [144]. COX-2 plays a major role in regulating intestinal inflammation. COX-2 is primarily expressed at sites of inflammation. Patients with active IBD have increased COX-2 expression levels in colonic epithelial cells, the myenteric plexus, and the medial layer of arteries [145]. In addition, COX-2 levels in blood samples of patients with active UC were significantly higher than those in healthy people and patients with palliative UC [1].
According to a study by Cui et al. [104], COX-2 expression was markedly increased by lipopolysaccharide (LPS) treatment in RAW 264.7 cells and decreased by baicalin treatment. In addition, COX-2 levels decreased in TNBS-induced UC rats compared to those in the TNBS-treated group, and it can also down-regulate pro-inflammatory mediators in the colonic mucosa. A decrease in pro-inflammatory cytokine production correlated with a decrease in mucosal TLR4 protein expression. The expression of p-nuclear factor kappa B (NF-κB) p65 protein was notably decreased, which was related to a similar decrease in p-IκB protein.
Several IBD model studies have shown that baicalein exhibits anti-inflammatory activity via COX-2 inhibition [85,118,128]. According to a study by Zhong et al. [85], baicalein significantly reduced the severity of DSS-induced colitis. Baicalein inhibited expression of COX-2 and inducible nitric oxide synthase (iNOS) and attenuated activity and phosphorylation of IKKβ and subsequent degradation of IκBα. In addition, baicalein inhibited p65 phosphorylation and nuclear translocation, decreased the DNA-binding activity of NF-κB, and inhibited the signal transducer and activator of transcription 3 (STAT3) phosphorylation and cyclin D1 expression.
According to a study by Yang et al. [133], wogonoside treatment of LPS-induced RAW 264.7 cells not only reduced production of inflammatory mediators, including nitric oxide (NO) and prostaglandin E2 (PGE2), but also prevented release of pro-inflammatory cytokines, including TNF-α and IL-6. In addition, wogonoside significantly inhibited gene expression of COX-2, iNOS, TNF-α, and IL-6.
Similarly, wogonin has been found to have anti-inflammatory effects in IBD models by reducing COX-2 [134,140,141,143]. Chi et al. [140] demonstrated that wogonin inhibited NO and PGE2 production in LPS-induced RAW 264.7 cells. They also showed that wogonin inhibited iNOS and COX-2 induction. Thus, they demonstrated that wogonin is not only a direct COX-2 inhibitor but also an inhibitor of iNOS and COX-2 induction.

4.1.2. Effect of SBG on TLR4/NF-κB

TLR4 is a TLR receptor activated by bacterial LPS. Once activated, TLR4 up-regulates expression of genes that stimulate downstream mitogen-activated protein kinases and NF-κB signaling pathways and encode inflammatory cytokines [146]. Transcription factor NF-κB plays an important role in regulation of many genes encoding inflammatory mediators. NF-κB activation entails rapid phosphorylation of IκB in response to pro-inflammatory stimuli [147]. Free NF-κB produced during this process translocates to the nucleus and induces transcription of COX-2, iNOS, IL-1β, TNF-α, and PGE2 mRNAs. Thus, inhibition of NF-κB signaling has potential therapeutic benefits in treatment of inflammatory diseases [128,148]. Under normal physiological conditions, TLR4 expression is low in intestinal epithelial cells, resulting in mucosal integrity and protection against invading bacteria. However, its expression is up-regulated in IBD, leading to inflammation and tissue damage [149]. Additionally, patients with active UC have high TLR4 expression in intestinal epithelial cells [150]. Activation of the TLR4 signaling pathway induces intestinal myeloid cell infiltration and release of inflammatory mediators in the colonic tissue. Therefore, inhibition of this pathway is an effective strategy for treating IBD [151].
Numerous studies have demonstrated the anti-inflammatory effects of baicalin through TLR4/NF-κB inhibition in IBD models [84,98,99,103,104,107,108,114]. According to the study by Yan et al. [114], treatment of LPS-induced RAW 264.7 cells with baicalin resulted in effective suppression of the expression of inflammatory factors (TNF-α, IL-1β, IL-6, COX, and iNOS). Previous studies have reported that up-regulation of miR-181b is associated with endotoxin resistance of RAW 264.7 cells and restraint of the excessive immune response induced by LPS [152]. In addition, HMGB1 (high-mobility group box 1) affects the inflammatory response by binding to the TLR4 protein [153]. Thus, baicalin could prevent LPS-induced inflammation in RAW 264.7 cells through the miR-181b/HMGB1/TRL4/NF-κB pathway.
Several studies have shown that baicalein reduces TLR4/NF-κB, leading to anti-inflammatory effects [85,107,118]. According to a study by Luo et al. [118], expression of several pro-inflammatory mediator genes, including iNOS, intercellular adhesion molecule-1 (ICAM-1), monocyte chemotactic protein-1 (MCP-1), COX-2, TNF-α, and IL-1β, was significantly reduced in the colon tissues of TNBS-colitis mice. Baicalein attenuated TNBS-induced colitis, at least in part, by inhibiting the TLR4/MyD88 signaling cascade (NF-κB and mitogen-activated protein kinase (MAPK)) and inactivating nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3) inflammasome.
Wogonoside has anti-inflammatory effects via TLR4/NF-κB reduction [131,132]. Sun et al. [131] demonstrated that wogonoside can exert an anti-inflammatory effect through dual inhibition of NF-κB and NLRP3 inflammasome in DSS-induced murine colitis. In addition, wogonoside significantly reduced production of IL-1β, TNF-α, and IL-6 and inhibited activation of NF-KB and NLRP3, thus inhibiting the mRNA expression of proIL-1β and NLRP3 inflammasom in porbol-myristate-acetate-differentiated monocomposite THP-1 cells.
Several studies have reported that wogonin has anti-inflammatory effects through TLR4/NF-κB reduction [134,136,143]. According to a study by Zhou et al. [134], wogonin reduced oxidative damage and elevation of inflammatory mediators in DSS-induced UC mice. Wogonin also inhibits NF-κB by down-regulating COX-2 and iNOS, modulating the Nrf2 signaling pathway, and reducing TLR-4/NF-κB activation. In addition, wogonin induced apoptosis by suppressing B-cell lymphoma-2 (Bcl-2) and increasing Bcl-2-associated X protein (Bax) and caspase-3 and -9 expression.

4.2. Effect of SBG on CRC

Several studies have demonstrated that SBG inhibits CRC growth both in vitro and in vivo. We summarized the results of several studies on the effects of SBG, baicalin (Table 1), baicalein (Table 2), wogonoside (Table 3), and wogonin (Table 4) on CRC.

4.2.1. Effect of SBG on Apoptosis in CRC

One way to treat cancer is to control or terminate the uncontrolled growth of cancer cells. Use of a cell’s own death mechanism is a very effective method. Apoptosis is a natural mechanism of programmed cell death [154]. It is a highly regulated process that eliminates unnecessary or unwanted cells [155]. There are two different apoptotic pathways: intrinsic and extrinsic, involving different types of signals [156]. The intrinsic apoptotic pathway uses mitochondria and mitochondrial proteins. The intrinsic pathway is regulated by the Bcl-2 family of proteins [157]. Many apoptotic stimuli result in up-regulation of BH3-only proteins, thereby activating both Bax and Bak. Bax is controlled by tumor suppressor gene p53 [158]. Extrinsic or death-receptor-mediated pathways are initiated by binding of death-inducing ligands to cell surface death receptors. Membrane death receptors belong to the TNF receptor superfamily and include tumor necrosis factor-receptor 1 (TNF-R1/DR1), death receptor-4 (DR4) and -5 (DR5), and Fas (Apo-1/CD95/DR2). These receptors are activated by specific ligands, such as Fas ligand (FasL), TNF-α, and TNF-related apoptosis-inducing ligand (TRAIL) [157].
Baicalin inhibits growth of several types of human CRC cells and induces apoptosis [109,110,111,112]. According to a study by Yang et al. [110], baicalin significantly inhibited the viability of CRC (RKO and HCT116) cells. In CRC cells, baicalin treatment induced cell cycle arrest in the G1 phase, promoted p53-independent apoptosis, and prevented both exogenous and endogenous TGFβ1-induced epithelial–mesenchymal transition in CRC cells by inhibiting the TGF-β/Smad pathway. In vivo experiments confirmed the antitumor effects by down-regulating marker protein levels of the cell cycle, epithelial–mesenchymal transition (EMT), and stemness in orthotopically transplanted tumors of CRC cells in BALB/c nude mice.
Baicalein is cytotoxic and induces apoptosis in human CRC cells [89,119,120,123,124]. Sui et al. [123] have shown that baicalein inhibited proliferation of CRC (HCT116, A549, and Panc-1) cells in a dose-dependent manner. Decidual protein induced by progesterone (DEPP) was originally known as a progesterone-induced gene in a human endometrial stromal cell cDNA library [159]. Baicalein induced up-regulation of DEPP and GADD45A, resulting in a marked apoptotic response in human CRC cells by promoting activation of MAPKs, with a positive feedback loop between GADD45A and c-Jun N-terminal kinase (JNK)/p38.
Wogonoside inhibits human CRC cell growth and induces apoptosis [90,132]. According to a study by Han et al. [90], wogonoside prevented cell growth and induced mitochondria-mediated autophagy-related apoptosis in human LOVO CRC cells by controlling the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR)/p70S6K signaling pathway.
Wogonin induces apoptosis in in vitro and in vivo CRC models [96,124,135,137]. Feng et al. [135] have shown that wogonin exerts anti-colon cancer activity in AOM/DSS animal and HCT116 cell models. Wogonin induced apoptosis in HCT116 cells via increased endoplasmic reticulum (ER) stress. Excessive ER stress promoted cytoplasmic localization of p53 by increasing the phosphorylation of p53 at the S315 and S376 sites, induced caspase-dependent apoptosis, and inhibited autophagy.

4.2.2. Effect of SBG on PI3K/AKT/mTOR in CRC

PI3Ks are intracellular lipid kinases implicated in cell proliferation, differentiation, and survival. The PI3K/AKT/mTOR signaling cascade promotes CRC oncogenesis in multiple ways, such as induction of drug resistance, inhibition of apoptosis, angiogenesis, metastasis, and EMT [160]. Overexpression of PI3K/AKT/mTOR signaling has been reported in diverse forms of cancer, particularly CRC [161]. Therefore, targeting PI3K/AKT/mTOR may be an important approach for cancer prevention and treatment [162].
Baicalin inhibits growth of CRC cells through PI3K/AKT/mTOR signaling [102,110,113]. According to a study by Zhu et al. [113], baicalin not only significantly inhibited cell viability, proliferation, migration, invasion, and angiogenesis but also promoted apoptosis in SW620 CRC cells. However, following inhibition of the PI3K/AKT/GSK-3β pathway using PI3K inhibitor LY294002, baicalin showed no significant inhibitory effects on the biological behavior of CRC, except for angiogenesis. Baicalin significantly inhibited phosphorylation of PI3K, AKT, and GSK-3β proteins and reduced mRNA levels. LY294002 further inhibited protein phosphorylation but did not significantly affect the mRNA. These effects of baicalin may be related to its role in prevention of the PI3K/AKT/GSK-3β pathway.
Baicalein also exhibits anticancer activity via the AKT pathway in in vitro and in vivo models of CRC [124,125]. Kim et al. [124] revealed that baicalein caused apoptosis through increased cell cycle arrest in the G1 phase in HT29 CRC cells. In addition, baicalein inactivated PI3K/AKT in a dose-dependent manner. Furthermore, administration of baicalein to mice inhibited growth of HT29 xenografts. Thus, baicalein can induce apoptosis through AKT inactivation in a p53-dependent manner in HT29 CRC cells.
Several studies have suggested that wogonoside exerts anticancer activity through the PI3K/AKT/mTOR pathway in CRC models [90,132]. According to a study by Sun et al. [132], wogonoside notably decreased disease severity, lowered tumor incidence, and suppressed development of colorectal adenomas in an AOM/DSS mouse model. In addition, wogonoside prevented inflammatory cancer cell proliferation and cell infiltration at the tumor site. In conclusion, wogonoside attenuates colitis-related tumor development in mice by inhibiting NF-κB activation through the PI3K/AKT pathway and inhibits progression of human CRC in an inflammatory microenvironment. It is a promising therapeutic agent for CRC.
Wogonin exerts anticancer activity via the PI3K/AKT pathway in CRC models [124,136,139]. For example, Tan et al. [139] have shown that wogonin inhibited growth of SW48 CRC cells via autophagy and apoptotic cell death. Wogonin induced SW48 cell arrest at the G2/M checkpoint of the cell cycle and inhibited the PI3K/AKT and STAT3 signaling pathways.

5. Conclusions

In this review, we outlined the current understanding of the mechanisms of the anti-inflammatory effects of the major constituents of SBG—baicalin, baicalein, wogonoside, and wogonin—in IBD and their anticancer efficacy in CRC. Traditional Chinese medicine has attracted significant attention in recent years because of its effectiveness in treatment of human diseases. Natural drugs have many advantages over conventional drugs, including better accessibility, economic feasibility, and low toxicity. The main constituents of SBG reduced release of inflammatory factors in several in vitro and in vivo models of IBD and showed anticancer activity by regulating different pathways. Therefore, SBG may represent a promising drug for IBD, and its components may be important lead compounds for development of new chemopreventive agents against IBD-associated CRC and CRC. However, although the potential of these compounds has been proven in various in vitro and in vivo settings, more preclinical and clinical studies are needed to validate their potency and anticancer activity.

Author Contributions

Conceptualization, J.Y.J., E.I. and N.D.K.; Investigation, J.Y.J.; Writing—Original Draft Preparation, J.Y.J.; Writing—Review and Editing, E.I. and N.D.K.; Supervision, E.I. and N.D.K.; Project Administration, N.D.K.; Funding Acquisition, N.D.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2022R1A6A3A01085858) (J.Y.J.). This study was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2021R1F1A1051265) and the Basic Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2018R1D1A1B07044648) (N.D.K.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, X.; Xie, L.; Long, J.; Liu, K.; Lu, J.; Liang, Y.; Cao, Y.; Dai, X.; Li, X. Therapeutic effect of baicalin on inflammatory bowel disease: A review. J. Ethnopharmacol. 2022, 283, 114749. [Google Scholar] [CrossRef] [PubMed]
  2. Joshi, A.; Soni, A.; Acharya, S. In vitro models and ex vivo systems used in inflammatory bowel disease. Vitr. Model. 2022, 1, 213–227. [Google Scholar] [CrossRef]
  3. Guan, Q. A Comprehensive review and update on the pathogenesis of inflammatory bowel disease. J. Immunol. Res. 2019, 2019, 7247238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. Lancet 2017, 390, 2769–2778. [Google Scholar] [CrossRef] [PubMed]
  5. Hanauer, S.B. Inflammatory bowel disease: Epidemiology, pathogenesis, and therapeutic opportunities. Inflamm. Bowel. Dis. 2006, 12 (Suppl. 1), S3–S9. [Google Scholar] [CrossRef]
  6. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: A systematic analysis for the global burden of disease study 2017. Lancet Gastroenterol. Hepatol. 2020, 5, 17–30. [CrossRef] [Green Version]
  7. Rajamäki, K.; Taira, A.; Katainen, R.; Välimäki, N.; Kuosmanen, A.; Plaketti, R.M.; Seppälä, T.T.; Ahtiainen, M.; Wirta, E.V.; Vartiainen, E.; et al. Genetic and epigenetic characteristics of inflammatory bowel disease-associated colorectal cancer. Gastroenterology 2021, 161, 592–607. [Google Scholar] [CrossRef]
  8. Nadeem, M.S.; Kumar, V.; Al-Abbasi, F.A.; Kamal, M.A.; Anwar, F. Risk of colorectal cancer in inflammatory bowel diseases. Semin. Cancer Biol. 2020, 64, 51–60. [Google Scholar] [CrossRef]
  9. Stidham, R.W.; Higgins, P.D. Colorectal cancer in inflammatory bowel disease. Clin. Colon Rectal Surg. 2018, 31, 168–178. [Google Scholar]
  10. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  11. Jung, K.W.; Won, Y.J.; Kang, M.J.; Kong, H.J.; Im, J.S.; Seo, H.G. Prediction of cancer incidence and mortality in Korea, 2022. Cancer Res. Treat. 2022, 54, 345–351. [Google Scholar] [CrossRef] [PubMed]
  12. Kang, Y.J.; Jang, J.Y.; Kwon, Y.H.; Lee, J.H.; Lee, S.; Park, Y.; Jung, Y.S.; Im, E.; Moon, H.R.; Chung, H.Y.; et al. MHY2245, a Sirtuin inhibitor, induces cell cycle arrest and apoptosis in HCT116 human colorectal cancer cells. Int. J. Mol. Sci. 2022, 23, 1590. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, Q.; Li, Z.; Guo, J.; Li, W.; You, F. Effect of Chinese herbal medicine formula on progression-free survival among patients with metastatic colorectal cancer: Study protocol for a multi-center, double-blinded, randomized, placebo-controlled trial. PLoS One 2022, 17, e0275058. [Google Scholar] [CrossRef]
  14. Gao, Y.; Snyder, S.A.; Smith, J.N.; Chen, Y.C. Anticancer properties of baicalein: A review. Med. Chem. Res. 2016, 25, 1515–1523. [Google Scholar] [CrossRef] [Green Version]
  15. Xiang, L.; Gao, Y.; Chen, S.; Sun, J.; Wu, J.; Meng, X. Therapeutic potential of Scutellaria baicalensis Georgi in lung cancer therapy. Phytomedicine 2022, 95, 153727. [Google Scholar] [CrossRef] [PubMed]
  16. Zhou, X.; Fu, L.; Wang, P.; Yang, L.; Zhu, X.; Li, C.G. Drug-herb interactions between Scutellaria baicalensis and pharmaceutical drugs: Insights from experimental studies, mechanistic actions to clinical applications. Biomed. Pharmacother. 2021, 138, 111445. [Google Scholar] [CrossRef] [PubMed]
  17. Yuan, Q.J.; Zhang, Z.Y.; Hu, J.; Guo, L.P.; Shao, A.J.; Huang, L.Q. Impacts of recent cultivation on genetic diversity pattern of a medicinal plant, Scutellaria baicalensis (Lamiaceae). BMC Genet 2010, 11, 29. [Google Scholar] [CrossRef] [Green Version]
  18. Wang, Z.L.; Wang, S.; Kuang, Y.; Hu, Z.M.; Qiao, X.; Ye, M. A comprehensive review on phytochemistry, pharmacology, and flavonoid biosynthesis of Scutellaria baicalensis. Pharm. Biol. 2018, 56, 465–484. [Google Scholar] [CrossRef] [Green Version]
  19. 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] [Green Version]
  20. Seyedian, S.S.; Nokhostin, F.; Malamir, M.D. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. J. Med. Life 2019, 12, 113–122. [Google Scholar] [CrossRef]
  21. Ungaro, R.; Mehandru, S.; Allen, P.B.; Peyrin-Biroulet, L.; Colombel, J.F. Ulcerative colitis. Lancet 2017, 389, 1756–1770. [Google Scholar] [CrossRef] [PubMed]
  22. Ocansey, D.K.W.; Zhang, L.; Wang, Y.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. Exosome-mediated effects and applications in inflammatory bowel disease. Biol. Rev. Camb. Philos. Soc. 2020, 95, 1287–1307. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, D.; Saikam, V.; Skrada, K.A.; Merlin, D.; Iyer, S.S. Inflammatory bowel disease biomarkers. Med. Res. Rev. 2022, 42, 1856–1887. [Google Scholar] [CrossRef] [PubMed]
  24. Høivik, M.L.; Moum, B.; Solberg, I.C.; Henriksen, M.; Cvancarova, M.; Bernklev, T. Work disability in inflammatory bowel disease patients 10 years after disease onset: Results from the IBSEN Study. Gut 2013, 62, 368–375. [Google Scholar] [CrossRef]
  25. Bernstein, C.N.; Blanchard, J.F.; Rawsthorne, P.; Yu, N. The prevalence of extraintestinal diseases in inflammatory bowel disease: A population-based study. Am. J. Gastroenterol. 2001, 96, 1116–1122. [Google Scholar] [CrossRef]
  26. Kim, E.R.; Chang, D.K. Colorectal cancer in inflammatory bowel disease: The risk, pathogenesis, prevention and diagnosis. World J. Gastroenterol. 2014, 20, 9872–9881. [Google Scholar] [CrossRef]
  27. Porter, R.J.; Arends, M.J.; Churchhouse, A.M.D.; Din, S. Inflammatory bowel disease-associated colorectal cancer: Translational risks from mechanisms to medicines. J. Crohns Colitis 2021, 15, 2131–2141. [Google Scholar] [CrossRef]
  28. Lucafò, M.; Curci, D.; Franzin, M.; Decorti, G.; Stocco, G. Inflammatory bowel disease and risk of colorectal cancer: An overview from pathophysiology to pharmacological prevention. Front. Pharmacol. 2021, 12, 772101. [Google Scholar] [CrossRef]
  29. Kameyama, H.; Nagahashi, M.; Shimada, Y.; Tajima, Y.; Ichikawa, H.; Nakano, M.; Sakata, J.; Kobayashi, T.; Narayanan, S.; Takabe, K.; et al. Genomic characterization of colitis-associated colorectal cancer. World J. Surg. Oncol. 2018, 16, 121. [Google Scholar] [CrossRef] [Green Version]
  30. Potack, J.; Itzkowitz, S.H. Colorectal cancer in inflammatory bowel disease. Gut Liver 2008, 2, 61–73. [Google Scholar] [CrossRef] [Green Version]
  31. Cai, Z.; Wang, S.; Li, J. Treatment of inflammatory bowel disease: A comprehensive review. Front. Med. 2021, 8, 765474. [Google Scholar] [CrossRef] [PubMed]
  32. Na, S.Y.; Moon, W. Perspectives on current and novel treatments for inflammatory bowel disease. Gut Liver 2019, 13, 604–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rangel, E.B. Tacrolimus in pancreas transplant: A focus on toxicity, diabetogenic effect and drug-drug interactions. Expert Opin. Drug Metab. Toxicol. 2014, 10, 1585–1605. [Google Scholar] [CrossRef] [PubMed]
  34. Reinisch, W.; Sandborn, W.J.; Hommes, D.W.; D’Haens, G.; Hanauer, S.; Schreiber, S.; Panaccione, R.; Fedorak, R.N.; Tighe, M.B.; Huang, B.; et al. Adalimumab for induction of clinical remission in moderately to severely active ulcerative colitis: Results of a randomised controlled trial. Gut 2011, 60, 780–787. [Google Scholar] [CrossRef] [PubMed]
  35. Sandborn, W.J.; van Assche, G.; Reinisch, W.; Colombel, J.F.; D’Haens, G.; Wolf, D.C.; Kron, M.; Tighe, M.B.; Lazar, A.; Thakkar, R.B. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology 2012, 142, e42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rutgeerts, P.; Sandborn, W.J.; Feagan, B.G.; Reinisch, W.; Olson, A.; Johanns, J.; Travers, S.; Rachmilewitz, D.; Hanauer, S.B.; Lichtenstein, G.R.; et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 2005, 353, 2462–2476. [Google Scholar] [CrossRef] [Green Version]
  37. Targan, S.R.; Hanauer, S.B.; van Deventer, S.J.; Mayer, L.; Present, D.H.; Braakman, T.; DeWoody, K.L.; Schaible, T.F.; Rutgeerts, P.J. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn’s disease cA2 study group. N. Engl. J. Med. 1997, 337, 1029–1035. [Google Scholar] [CrossRef] [Green Version]
  38. Hanauer, S.B.; Sandborn, W.J.; Rutgeerts, P.; Fedorak, R.N.; Lukas, M.; MacIntosh, D.; Panaccione, R.; Wolf, D.; Pollack, P. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn’s disease: The CLASSIC-I trial. Gastroenterology 2006, 130, 323–333; quiz 591. [Google Scholar] [CrossRef] [Green Version]
  39. Schreiber, S.; Khaliq-Kareemi, M.; Lawrance, I.C.; Thomsen, O.; Hanauer, S.B.; McColm, J.; Bloomfield, R.; Sandborn, W.J. Maintenance therapy with certolizumab pegol for Crohn’s disease. N. Engl. J. Med. 2007, 357, 239–250. [Google Scholar] [CrossRef]
  40. Sandborn, W.J.; Hanauer, S.B.; Rutgeerts, P.; Fedorak, R.N.; Lukas, M.; MacIntosh, D.G.; Panaccione, R.; Wolf, D.; Kent, J.D.; Bittle, B.; et al. Adalimumab for maintenance treatment of Crohn’s disease: Results of the CLASSIC II trial. Gut 2007, 56, 1232–1239. [Google Scholar] [CrossRef] [Green Version]
  41. Hanauer, S.B.; Feagan, B.G.; Lichtenstein, G.R.; Mayer, L.F.; Schreiber, S.; Colombel, J.F.; Rachmilewitz, D.; Wolf, D.C.; Olson, A.; Bao, W.; et al. Maintenance infliximab for Crohn’s disease: The ACCENT I randomised trial. Lancet 2002, 359, 1541–1549. [Google Scholar] [CrossRef] [PubMed]
  42. Allez, M.; Karmiris, K.; Louis, E.; Van Assche, G.; Ben-Horin, S.; Klein, A.; Van der Woude, J.; Baert, F.; Eliakim, R.; Katsanos, K.; et al. Report of the ECCO pathogenesis workshop on anti-TNF therapy failures in inflammatory bowel diseases: Definitions, frequency and pharmacological aspects. J. Crohns Colitis 2010, 4, 355–366. [Google Scholar] [CrossRef]
  43. D’Haens, G.R.; Panaccione, R.; Higgins, P.D.; Vermeire, S.; Gassull, M.; Chowers, Y.; Hanauer, S.B.; Herfarth, H.; Hommes, D.W.; Kamm, M.; et al. The London position statement of the world congress of gastroenterology on biological therapy for IBD with the European Crohn’s and colitis organization: When to start, when to stop, which drug to choose, and how to predict response? Am. J. Gastroenterol. 2011, 106, 199–212; quiz 213. [Google Scholar] [CrossRef]
  44. Ben-Horin, S.; Chowers, Y. Review article: Loss of response to anti-TNF treatments in Crohn’s disease. Aliment. Pharmacol. Ther. 2011, 33, 987–995. [Google Scholar] [CrossRef] [PubMed]
  45. Toruner, M.; Loftus, E.V., Jr.; Harmsen, W.S.; Zinsmeister, A.R.; Orenstein, R.; Sandborn, W.J.; Colombel, J.F.; Egan, L.J. Risk factors for opportunistic infections in patients with inflammatory bowel disease. Gastroenterology 2008, 134, 929–936. [Google Scholar] [CrossRef]
  46. Andersen, N.N.; Jess, T. Risk of infections associated with biological treatment in inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 16014–16019. [Google Scholar] [CrossRef]
  47. Siegel, C.A.; Marden, S.M.; Persing, S.M.; Larson, R.J.; Sands, B.E. Risk of lymphoma associated with combination anti-tumor necrosis factor and immunomodulator therapy for the treatment of Crohn’s disease: A meta-analysis. Clin. Gastroenterol. Hepatol. 2009, 7, 874–881. [Google Scholar] [CrossRef] [Green Version]
  48. Lea, T. Caco-2 Cell Line. In The Impact of Food Bioactives on Health: In Vitro and Ex Vivo Models; Verhoeckx, K., Cotter, P., López-Expósito, I., Kleiveland, C., Lea, T., Mackie, A., Requena, T., Swiatecka, D., Wichers, H., Eds.; Springer International Publishing: Cham, Switzerland, 2015; pp. 103–111. [Google Scholar]
  49. Lozoya-Agullo, I.; Araújo, F.; González-Álvarez, I.; Merino-Sanjuán, M.; González-Álvarez, M.; Bermejo, M.; Sarmento, B. Usefulness of Caco-2/HT29-MTX and Caco-2/HT29-MTX/Raji B coculture models to predict intestinal and colonic permeability compared to Caco-2 monoculture. Mol. Pharm. 2017, 14, 1264–1270. [Google Scholar] [CrossRef] [PubMed]
  50. Borgonetti, V.; Cocetta, V.; Biagi, M.; Carnevali, I.; Governa, P.; Montopoli, M. Anti-inflammatory activity of a fixed combination of probiotics and herbal extract in an in-vitro model of intestinal inflammation by stimulating Caco-2 cells with LPS-conditioned THP-1 cells medium. Minerva Pediatr. 2022, 74, 511–518. [Google Scholar] [CrossRef]
  51. Andrews, C.; McLean, M.H.; Durum, S.K. Cytokine tuning of intestinal epithelial function. Front. Immunol. 2018, 9, 1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Weber, L.; Kuck, K.; Jürgenliemk, G.; Heilmann, J.; Lipowicz, B.; Vissiennon, C. Anti-inflammatory and barrier-stabilising effects of myrrh, coffee charcoal and chamomile flower extract in a co-culture cell model of the intestinal mucosa. Biomolecules 2020, 10, 1033. [Google Scholar] [CrossRef] [PubMed]
  53. Lanevskij, K.; Didziapetris, R. Physicochemical QSAR analysis of passive permeability across Caco-2 monolayers. J. Pharm. Sci. 2019, 108, 78–86. [Google Scholar] [CrossRef] [Green Version]
  54. Jarc, T.; Novak, M.; Hevir, N.; Rižner, T.L.; Kreft, M.E.; Kristan, K. Demonstrating suitability of the Caco-2 cell model for BCS-based biowaiver according to the recent FDA and ICH harmonised guidelines. J. Pharm. Pharmacol. 2019, 71, 1231–1242. [Google Scholar] [CrossRef] [PubMed]
  55. Xu, R.; Zhu, H.; Hu, L.; Yu, B.; Zhan, X.; Yuan, Y.; Zhou, P. Characterization of the intestinal absorption of morroniside from Cornus officinalis Sieb. et Zucc via a Caco-2 cell monolayer model. PLoS One 2020, 15, e0227844. [Google Scholar] [CrossRef]
  56. Zhou, G.; Kong, W.S.; Li, Z.C.; Xie, R.F.; Yu, T.Y.; Zhou, X. Effects of qing chang suppository powder and its ingredients on IL-17 signal pathway in HT-29 cells and DSS-induced Mice. Phytomedicine 2021, 87, 153573. [Google Scholar] [CrossRef] [PubMed]
  57. Zweibaum, A.; Laburthe, M.; Grasset, E.; Louvard, D. Use of cultured cell lines in studies of intestinal cell differentiation and function. Compr. Physiol. 2010, 223–255. [Google Scholar] [CrossRef]
  58. Adamczak, M.I.; Hagesaether, E.; Smistad, G.; Hiorth, M. An in vitro study of mucoadhesion and biocompatibility of polymer coated liposomes on HT29-MTX mucus-producing cells. Int. J. Pharm. 2016, 498, 225–233. [Google Scholar] [CrossRef] [PubMed]
  59. O’Connell, L.; Winter, D.C.; Aherne, C.M. The role of organoids as a novel platform for modeling of inflammatory bowel disease. Front. Pediatr. 2021, 9, 624045. [Google Scholar] [CrossRef]
  60. Nozaki, K.; Mochizuki, W.; Matsumoto, Y.; Matsumoto, T.; Fukuda, M.; Mizutani, T.; Watanabe, M.; Nakamura, T. Co-culture with intestinal epithelial organoids allows efficient expansion and motility analysis of intraepithelial lymphocytes. J. Gastroenterol. 2016, 51, 206–213. [Google Scholar] [CrossRef] [Green Version]
  61. Noel, G.; Baetz, N.W.; Staab, J.F.; Donowitz, M.; Kovbasnjuk, O.; Pasetti, M.F.; Zachos, N.C. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci. Rep. 2017, 7, 45270. [Google Scholar] [CrossRef] [Green Version]
  62. Poletti, M.; Arnauts, K.; Ferrante, M.; Korcsmaros, T. Organoid-based models to study the role of host-microbiota interactions in IBD. J. Crohns Colitis 2021, 15, 1222–1235. [Google Scholar] [CrossRef]
  63. Dutta, D.; Clevers, H. Organoid culture systems to study host-pathogen interactions. Curr. Opin. Immunol. 2017, 48, 15–22. [Google Scholar] [CrossRef] [PubMed]
  64. Kayisoglu, O.; Weiss, F.; Niklas, C.; Pierotti, I.; Pompaiah, M.; Wallaschek, N.; Germer, C.T.; Wiegering, A.; Bartfeld, S. Location-specific cell identity rather than exposure to GI microbiota defines many innate immune signalling cascades in the gut epithelium. Gut 2021, 70, 687–697. [Google Scholar] [CrossRef] [PubMed]
  65. Ojo, B.A.; VanDussen, K.L.; Rosen, M.J. The promise of patient-derived colon organoids to model Ulcerative Colitis. Inflamm. Bowel. Dis. 2022, 28, 299–308. [Google Scholar] [CrossRef] [PubMed]
  66. Lee, C.; Hong, S.N.; Kim, E.R.; Chang, D.K.; Kim, Y.H. Epithelial regeneration ability of Crohn’s disease assessed using patient-derived intestinal organoids. Int. J. Mol. Sci. 2021, 22, 6013. [Google Scholar] [CrossRef]
  67. Lesuffleur, T.; Barbat, A.; Dussaulx, E.; Zweibaum, A. Growth adaptation to methotrexate of HT-29 human colon carcinoma cells is associated with their ability to differentiate into columnar absorptive and mucus-secreting cells. Cancer Res. 1990, 50, 6334–6343. [Google Scholar]
  68. Pan, F.; Han, L.; Zhang, Y.; Yu, Y.; Liu, J. Optimization of Caco-2 and HT29 co-culture in vitro cell models for permeability studies. Int.J. Food Sci. Nutr. 2015, 66, 680–685. [Google Scholar] [CrossRef]
  69. Kleiveland, C.R. Co-cultivation of Caco-2 and HT-29MTX. In The Impact of Food Bioactives on Health: In Vitro and Ex Vivo Models; Verhoeckx, K., Cotter, P., López-Expósito, I., Kleiveland, C., Lea, T., Mackie, A., Requena, T., Swiatecka, D., Wichers, H., Eds.; Springer: Cham, Switzerland, 2015; Chapter 13; p. 135. [Google Scholar]
  70. Ponce de León-Rodríguez, M.D.C.; Guyot, J.P.; Laurent-Babot, C. Intestinal in vitro cell culture models and their potential to study the effect of food components on intestinal inflammation. Crit. Rev. Food Sci. Nutr. 2019, 59, 3648–3666. [Google Scholar] [CrossRef]
  71. Sawasvirojwong, S.; Kittayaruksakul, S. Constitutive androstane receptor inhibits Ca2+-dependent Cl secretion in intestinal epithelial cells. J. Physiol. 2019, 32, 47–52. [Google Scholar]
  72. Baxter, E.W.; Graham, A.E.; Re, N.A.; Carr, I.M.; Robinson, J.I.; Mackie, S.L.; Morgan, A.W. Standardized protocols for differentiation of THP-1 cells to macrophages with distinct M(IFNγ+LPS), M(IL-4) and M(IL-10) phenotypes. J. Immunol. Methods 2020, 478, 112721. [Google Scholar] [CrossRef]
  73. Zhen, D.; Xuan, T.Q.; Hu, B.; Bai, X.; Fu, D.N.; Wang, Y.; Wu, Y.; Yang, J.; Ma, Q. Pteryxin attenuates LPS-induced inflammatory responses and inhibits NLRP3 inflammasome activation in RAW264.7 cells. J. Ethnopharmacol. 2022, 284, 114753. [Google Scholar] [CrossRef] [PubMed]
  74. Thomson, A.; Smart, K.; Somerville, M.S.; Lauder, S.N.; Appanna, G.; Horwood, J.; Sunder Raj, L.; Srivastava, B.; Durai, D.; Scurr, M.J.; et al. The Ussing chamber system for measuring intestinal permeability in health and disease. BMC Gastroenterol. 2019, 19, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Fang, Y.; Cao, W.; Xia, M.; Pan, S.; Xu, X. Transport and interactions of co-incubated bi-functional flavonoids through inhibiting the function of P-Glycoprotein (P-gp) using KB/multidrug-resistant (MDR) cells and rat everted gut sacs. J. Agric. Food Chem. 2022, 70, 1923–1933. [Google Scholar] [CrossRef] [PubMed]
  76. Rahman, S.; Ghiboub, M.; Donkers, J.M.; van de Steeg, E.; van Tol, E.A.F.; Hakvoort, T.B.M.; de Jonge, W.J. The progress of intestinal epithelial models from cell lines to gut-on-chip. Int. J. Mol. Sci. 2021, 22, 13472. [Google Scholar] [CrossRef] [PubMed]
  77. Beaurivage, C.; Kanapeckaite, A.; Loomans, C.; Erdmann, K.S.; Stallen, J.; Janssen, R.A.J. Development of a human primary gut-on-a-chip to model inflammatory processes. Sci. Rep. 2020, 10, 21475. [Google Scholar] [CrossRef] [PubMed]
  78. Katsandegwaza, B.; Horsnell, W.; Smith, K. Inflammatory bowel disease: A review of pre-clinical murine models of human Disease. Int. J. Mol. Sci. 2022, 23, 9344. [Google Scholar] [CrossRef]
  79. Wirtz, S.; Popp, V.; Kindermann, M.; Gerlach, K.; Weigmann, B.; Fichtner-Feigl, S.; Neurath, M.F. Chemically induced mouse models of acute and chronic intestinal inflammation. Nat. Protoc. 2017, 12, 1295–1309. [Google Scholar] [CrossRef]
  80. Chassaing, B.; Aitken, J.D.; Malleshappa, M.; Vijay-Kumar, M. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr. Protoc. Immunol. 2015, 104, 15.25.1–15.25.14. [Google Scholar] [CrossRef] [Green Version]
  81. Singh, K.N.M.; Koli, P.G. Animal models for preclinical drug research on ulcerative colitis: A review. J. Sci. Soc. 2018, 45, 80–83. [Google Scholar] [CrossRef]
  82. Kiesler, P.; Fuss, I.J.; Strober, W. Experimental models of inflammatory bowel diseases. Cell. Mol. Gastroenterol. Hepatol. 2015, 1, 154–170. [Google Scholar] [CrossRef] [Green Version]
  83. Mizoguchi, E.; Low, D.; Ezaki, Y.; Okada, T. Recent updates on the basic mechanisms and pathogenesis of inflammatory bowel diseases in experimental animal models. Intest. Res. 2020, 18, 151–167. [Google Scholar] [CrossRef] [PubMed]
  84. Fu, Y.J.; Xu, B.; Huang, S.W.; Luo, X.; Deng, X.L.; Luo, S.; Liu, C.; Wang, Q.; Chen, J.Y.; Zhou, L. Baicalin prevents LPS-induced activation of TLR4/NF-κB p65 pathway and inflammation in mice via inhibiting the expression of CD14. Acta Pharmacol. Sin. 2021, 42, 88–96. [Google Scholar] [CrossRef] [PubMed]
  85. Zhong, X.; Surh, Y.J.; Do, S.G.; Shin, E.; Shim, K.S.; Lee, C.K.; Na, H.K. Baicalein inhibits dextran sulfate sodium-induced mouse colitis. J. Cancer Prev. 2019, 24, 129–138. [Google Scholar] [CrossRef] [PubMed]
  86. Feng, H.; Zhang, K.; Zhang, K.; Guo, Z.; Liu, Q.; Wang, L.; Wang, X.; Qiu, Z.; Wang, G.; Zhang, J.; et al. Antiviral activity and underlying mechanisms of baicalin against avian infectious bronchitis virus in vitro. Avian Pathol. 2022, 51, 574–589. [Google Scholar] [CrossRef]
  87. 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] [Green Version]
  88. 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]
  89. Dou, J.; Wang, Z.; Ma, L.; Peng, B.; Mao, K.; Li, C.; Su, M.; Zhou, C.; Peng, G. Baicalein and baicalin inhibit colon cancer using two distinct fashions of apoptosis and senescence. Oncotarget 2018, 9, 20089–20102. [Google Scholar] [CrossRef] [Green Version]
  90. Han, C.; Xing, G.; Zhang, M.; Zhong, M.; Han, Z.; He, C.; Liu, X. Wogonoside inhibits cell growth and induces mitochondrial-mediated autophagy-related apoptosis in human colon cancer cells through the PI3K/AKT/mTOR/p70S6K signaling pathway. Oncol. Lett. 2018, 15, 4463–4470. [Google Scholar] [CrossRef] [Green Version]
  91. Li-Weber, M. New therapeutic aspects of flavones: The anticancer properties of Scutellaria and its main active constituents wogonin, baicalein and baicalin. Cancer Treat. Rev. 2009, 35, 57–68. [Google Scholar] [CrossRef]
  92. Liao, H.; Ye, J.; Gao, L.; Liu, Y. The main bioactive compounds of Scutellaria baicalensis Georgi. for alleviation of inflammatory cytokines: A comprehensive review. Biomed. Pharmacother. 2021, 133, 110917. [Google Scholar] [CrossRef]
  93. Yu, C.; Zhang, Z.; Zhang, H.; Zhen, Z.; Calway, T.; Wang, Y.; Yuan, C.S.; Wang, C.Z. Pretreatment of baicalin and wogonoside with glycoside hydrolase: A promising approach to enhance anticancer potential. Oncol. Rep. 2013, 30, 2411–2418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ganguly, R.; Gupta, A.; Pandey, A.K. Role of baicalin as a potential therapeutic agent in hepatobiliary and gastrointestinal disorders: A review. World J. Gastroenterol. 2022, 28, 3047–3062. [Google Scholar] [CrossRef] [PubMed]
  95. Noh, K.; Kang, Y.; Nepal, M.R.; Jeong, K.S.; Oh, D.G.; Kang, M.J.; Lee, S.; Kang, W.; Jeong, H.G.; Jeong, T.C. Role of intestinal microbiota in baicalin-induced drug interaction and its pharmacokinetics. Molecules 2016, 21, 337. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, C.Z.; Wan, J.Y.; Zhang, C.F.; Lu, F.; Chen, L.; Yuan, C.S. Deglycosylation of wogonoside enhances its anticancer potential. J. Cancer Res. Ther. 2018, 14, S594–S599. [Google Scholar] [CrossRef]
  97. Heo, S.H.; Song, J.; Kim, B.J.; Kim, H.; Chang, D.I. Rationale and design to assess the efficacy and safety of HT047 in patients with acute ischemic stroke: A multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase II trial. Medicine 2019, 98, e17655. [Google Scholar] [CrossRef]
  98. Zhang, C.L.; Zhang, S.; He, W.X.; Lu, J.L.; Xu, Y.J.; Yang, J.Y.; Liu, D. Baicalin may alleviate inflammatory infiltration in dextran sodium sulfate-induced chronic ulcerative colitis via inhibiting IL-33 expression. Life Sci. 2017, 186, 125–132. [Google Scholar] [CrossRef]
  99. Feng, J.; Guo, C.; Zhu, Y.; Pang, L.; Yang, Z.; Zou, Y.; Zheng, X. Baicalin down regulates the expression of TLR4 and NFkB-p65 in colon tissue in mice with colitis induced by dextran sulfate sodium. Int. J. Clin. Exp. Med. 2014, 7, 4063–4072. [Google Scholar]
  100. Dou, W.; Mukherjee, S.; Li, H.; Venkatesh, M.; Wang, H.; Kortagere, S.; Peleg, A.; Chilimuri, S.S.; Wang, Z.T.; Feng, Y.; et al. Alleviation of gut inflammation by Cdx2/Pxr pathway in a mouse model of chemical colitis. PLoS One 2012, 7, e36075. [Google Scholar] [CrossRef] [Green Version]
  101. Zhu, L.; Xu, L.Z.; Zhao, S.; Shen, Z.F.; Shen, H.; Zhan, L.B. Protective effect of baicalin on the regulation of Treg/Th17 balance, gut microbiota and short-chain fatty acids in rats with ulcerative colitis. Appl. Microbiol. Biotechnol. 2020, 104, 5449–5460. [Google Scholar] [CrossRef]
  102. Zhu, L.; Shen, H.; Gu, P.Q.; Liu, Y.J.; Zhang, L.; Cheng, J.F. Baicalin alleviates TNBS-induced colitis by inhibiting PI3K/AKT pathway activation. Exp. Ther. Med. 2020, 20, 581–590. [Google Scholar] [CrossRef]
  103. Shen, J.; Cheng, J.; Zhu, S.; Zhao, J.; Ye, Q.; Xu, Y.; Dong, H.; Zheng, X. Regulating effect of baicalin on IKK/IKB/NF-kB signaling pathway and apoptosis-related proteins in rats with ulcerative colitis. Int. Immunopharmacol. 2019, 73, 193–200. [Google Scholar] [CrossRef] [PubMed]
  104. Cui, L.; Feng, L.; Zhang, Z.H.; Jia, X.B. The anti-inflammation effect of baicalin on experimental colitis through inhibiting TLR4/NF-κB pathway activation. Int. Immunopharmacol. 2014, 23, 294–303. [Google Scholar] [CrossRef] [PubMed]
  105. Yao, J.; Cao, X.; Zhang, R.; Li, Y.X.; Xu, Z.L.; Zhang, D.G.; Wang, L.S.; Wang, J.Y. Protective effect of baicalin against experimental colitis via suppression of oxidant stress and apoptosis. Pharmacogn. Mag. 2016, 12, 225–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Zou, Y.; Dai, S.X.; Chi, H.G.; Li, T.; He, Z.W.; Wang, J.; Ye, C.G.; Huang, G.L.; Zhao, B.; Li, W.Y.; et al. Baicalin attenuates TNBS-induced colitis in rats by modulating the Th17/Treg paradigm. Arch. Pharm. Res. 2015, 38, 1873–1887. [Google Scholar] [CrossRef]
  107. Liang, S.; Deng, X.; Lei, L.; Zheng, Y.; Ai, J.; Chen, L.; Xiong, H.; Mei, Z.; Cheng, Y.C.; Ren, Y. The comparative study of the therapeutic effects and mechanism of baicalin, baicalein, and their combination on ulcerative colitis rat. Front. Pharmacol. 2019, 10, 1466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Rizzo, V.; Ferlazzo, N.; Currò, M.; Isola, G.; Matarese, M.; Bertuccio, M.P.; Caccamo, D.; Matarese, G.; Ientile, R. Baicalin-induced autophagy preserved LPS-stimulated intestinal cells from inflammation and alterations of paracellular permeability. Int. J. Mol. Sci. 2021, 22, 2315. [Google Scholar] [CrossRef]
  109. Tao, Y.; Zhan, S.; Wang, Y.; Zhou, G.; Liang, H.; Chen, X.; Shen, H. Baicalin, the major component of traditional Chinese medicine Scutellaria baicalensis induces colon cancer cell apoptosis through inhibition of oncomiRNAs. Sci. Rep. 2018, 8, 14477. [Google Scholar] [CrossRef] [Green Version]
  110. Yang, B.; Bai, H.; Sa, Y.; Zhu, P.; Liu, P. Inhibiting EMT, stemness and cell cycle involved in baicalin-induced growth inhibition and apoptosis in colorectal cancer cells. J. Cancer 2020, 11, 2303–2317. [Google Scholar] [CrossRef]
  111. Ma, W.; Liu, X.; Du, W. Baicalin induces apoptosis in SW480 cells through downregulation of the SP1 transcription factor. Anticancer Drugs 2019, 30, 153–158. [Google Scholar] [CrossRef]
  112. Chen, W.C.; Kuo, T.H.; Tzeng, Y.S.; Tsai, Y.C. Baicalin induces apoptosis in SW620 human colorectal carcinoma cells in vitro and suppresses tumor growth in vivo. Molecules 2012, 17, 3844–3857. [Google Scholar] [CrossRef] [Green Version]
  113. Li, H.; Dong, W.; Hou, J.; He, D. Effects of baicalin regulating PI3K/AKT/GSK-3β pathway on biological behavior of human colon cancer cells. Chin. Arch. TCM. Med. 2021, 39, 241–245. [Google Scholar] [CrossRef]
  114. Yan, G.; Chen, L.; Wang, H.; Wu, S.; Li, S.; Wang, X. Baicalin inhibits LPS-induced inflammation in RAW264.7 cells through miR-181b/HMGB1/TRL4/NF-κB pathway. Am. J. Transl. Res. 2021, 13, 10127–10141. [Google Scholar] [PubMed]
  115. Zhu, W.; Jin, Z.; Yu, J.; Liang, J.; Yang, Q.; Li, F.; Shi, X.; Zhu, X.; Zhang, X. Baicalin ameliorates experimental inflammatory bowel disease through polarization of macrophages to an M2 phenotype. Int. Immunopharmacol. 2016, 35, 119–126. [Google Scholar] [CrossRef] [PubMed]
  116. Hong, T.; Jin, G.B.; Cho, S.; Cyong, J.C. Evaluation of the anti-inflammatory effect of baicalein on dextran sulfate sodium-induced colitis in mice. Planta Med. 2002, 68, 268–271. [Google Scholar] [CrossRef]
  117. Liu, C.; Li, Y.; Chen, Y.; Huang, S.; Wang, X.; Luo, S.; Su, Y.; Zhou, L.; Luo, X. Baicalein restores the balance of Th17/Treg cells via aryl hydrocarbon receptor to attenuate colitis. Mediators Inflamm. 2020, 2020, 5918587. [Google Scholar] [CrossRef]
  118. Luo, X.; Yu, Z.; Deng, C.; Zhang, J.; Ren, G.; Sun, A.; Mani, S.; Wang, Z.; Dou, W. Baicalein ameliorates TNBS-induced colitis by suppressing TLR4/MyD88 signaling cascade and NLRP3 inflammasome activation in mice. Sci. Rep. 2017, 7, 16374. [Google Scholar] [CrossRef] [Green Version]
  119. Wang, C.Z.; Zhang, C.F.; Chen, L.; Anderson, S.; Lu, F.; Yuan, C.S. Colon cancer chemopreventive effects of baicalein, an active enteric microbiome metabolite from baicalin. Int. J. Oncol. 2015, 47, 1749–1758. [Google Scholar] [CrossRef] [Green Version]
  120. Kim, D.H.; Hossain, M.A.; Kang, Y.J.; Jang, J.Y.; Lee, Y.J.; Im, E.; Yoon, J.H.; Kim, H.S.; Chung, H.Y.; Kim, N.D. Baicalein, an active component of Scutellaria baicalensis Georgi, induces apoptosis in human colon cancer cells and prevents AOM/DSS-induced colon cancer in mice. Int. J. Oncol. 2013, 43, 1652–1658. [Google Scholar] [CrossRef] [Green Version]
  121. Havermann, S.; Chovolou, Y.; Humpf, H.U.; Wätjen, W. Modulation of the Nrf2 signalling pathway in Hct116 colon carcinoma cells by baicalein and its methylated derivative negletein. Pharm. Biol. 2016, 54, 1491–1502. [Google Scholar] [CrossRef] [Green Version]
  122. Chen, Z.; Hou, R.; Gao, S.; Song, D.; Feng, Y. Baicalein inhibits proliferation activity of human colorectal cancer cells HCT116 through downregulation of Ezrin. Cell Physiol. Biochem. 2018, 49, 2035–2046. [Google Scholar] [CrossRef]
  123. Su, M.Q.; Zhou, Y.R.; Rao, X.; Yang, H.; Zhuang, X.H.; Ke, X.J.; Peng, G.Y.; Zhou, C.L.; Shen, B.Y.; Dou, J. Baicalein induces the apoptosis of HCT116 human colon cancer cells via the upregulation of DEPP/Gadd45a and activation of MAPKs. Int. J. Oncol. 2018, 53, 750–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kim, S.J.; Kim, H.J.; Kim, H.R.; Lee, S.H.; Cho, S.D.; Choi, C.S.; Nam, J.S.; Jung, J.Y. Antitumor actions of baicalein and wogonin in HT-29 human colorectal cancer cells. Mol. Med. Rep. 2012, 6, 1443–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Rui, X.; Yan, X.I.; Zhang, K. Baicalein inhibits the migration and invasion of colorectal cancer cells via suppression of the AKT signaling pathway. Oncol. Lett. 2016, 11, 685–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chai, Y.; Xu, J.; Yan, B. The anti-metastatic effect of baicalein on colorectal cancer. Oncol. Rep. 2017, 37, 2317–2323. [Google Scholar] [CrossRef]
  127. Qi, Z.; Yin, F.; Lu, L.; Shen, L.; Qi, S.; Lan, L.; Luo, L.; Yin, Z. Baicalein reduces lipopolysaccharide-induced inflammation via suppressing JAK/STATs activation and ROS production. Inflamm. Res. 2013, 62, 845–855. [Google Scholar] [CrossRef] [PubMed]
  128. Fan, G.W.; Zhang, Y.; Jiang, X.; Zhu, Y.; Wang, B.; Su, L.; Cao, W.; Zhang, H.; Gao, X. Anti-inflammatory activity of baicalein in LPS-stimulated RAW264.7 macrophages via estrogen receptor and NF-κB-dependent pathways. Inflammation 2013, 36, 1584–1591. [Google Scholar] [CrossRef]
  129. Kim, Y.J.; Kim, H.J.; Lee, J.Y.; Kim, D.H.; Kang, M.S.; Park, W. Anti-inflammatory effect of baicalein on polyinosinic⁻polycytidylic acid-induced RAW 264.7 mouse macrophages. Viruses 2018, 10, 224. [Google Scholar] [CrossRef] [Green Version]
  130. Huang, S.; Fu, Y.; Xu, B.; Liu, C.; Wang, Q.; Luo, S.; Nong, F.; Wang, X.; Huang, S.; Chen, J.; et al. Wogonoside alleviates colitis by improving intestinal epithelial barrier function via the MLCK/pMLC2 pathway. Phytomedicine 2020, 68, 153179. [Google Scholar] [CrossRef]
  131. Sun, Y.; Zhao, Y.; Yao, J.; Zhao, L.; Wu, Z.; Wang, Y.; Pan, D.; Miao, H.; Guo, Q.; Lu, N. Wogonoside protects against dextran sulfate sodium-induced experimental colitis in mice by inhibiting NF-κB and NLRP3 inflammasome activation. Biochem. Pharmacol. 2015, 94, 142–154. [Google Scholar] [CrossRef]
  132. Sun, Y.; Zhao, Y.; Wang, X.; Zhao, L.; Li, W.; Ding, Y.; Kong, L.; Guo, Q.; Lu, N. Wogonoside prevents colitis-associated colorectal carcinogenesis and colon cancer progression in inflammation-related microenvironment via inhibiting NF-κB activation through PI3K/Akt pathway. Oncotarget 2016, 7, 34300–34315. [Google Scholar] [CrossRef] [Green Version]
  133. Yang, Y.Z.; Tang, Y.Z.; Liu, Y.H. Wogonoside displays anti-inflammatory effects through modulating inflammatory mediator expression using RAW264.7 cells. J. Ethnopharmacol. 2013, 148, 271–276. [Google Scholar] [CrossRef] [PubMed]
  134. Zhou, Y.; Dou, F.; Song, H.; Liu, T. Anti-ulcerative effects of wogonin on ulcerative colitis induced by dextran sulfate sodium via Nrf2/TLR4/NF-κB signaling pathway in BALB/c mice. Environ. Toxicol. 2022, 37, 954–963. [Google Scholar] [CrossRef] [PubMed]
  135. Feng, Q.; Wang, H.; Pang, J.; Ji, L.; Han, J.; Wang, Y.; Qi, X.; Liu, Z.; Lu, L. Prevention of wogonin on colorectal cancer tumorigenesis by regulating p53 nuclear translocation. Front. Pharmacol. 2018, 9, 1356. [Google Scholar] [CrossRef] [PubMed]
  136. Yao, J.; Zhao, L.; Zhao, Q.; Zhao, Y.; Sun, Y.; Zhang, Y.; Miao, H.; You, Q.D.; Hu, R.; Guo, Q.L. NF-κB and Nrf2 signaling pathways contribute to wogonin-mediated inhibition of inflammation-associated colorectal carcinogenesis. Cell Death Dis. 2014, 5, e1283. [Google Scholar] [CrossRef]
  137. He, L.; Lu, N.; Dai, Q.; Zhao, Y.; Zhao, L.; Wang, H.; Li, Z.; You, Q.; Guo, Q. Wogonin induced G1 cell cycle arrest by regulating Wnt/β-catenin signaling pathway and inactivating CDK8 in human colorectal cancer carcinoma cells. Toxicology 2013, 312, 36–47. [Google Scholar] [CrossRef]
  138. Zhao, Y.; Zhang, L.; Wu, Y.; Dai, Q.; Zhou, Y.; Li, Z.; Yang, L.; Guo, Q.; Lu, N. Selective anti-tumor activity of wogonin targeting the Warburg effect through stablizing p53. Pharmacol. Res. 2018, 135, 49–59. [Google Scholar] [CrossRef]
  139. Tan, H.; Li, X.; Yang, W.H.; Kang, Y. A flavone, wogonin from Scutellaria baicalensis inhibits the proliferation of human colorectal cancer cells by inducing of autophagy, apoptosis and G2/M cell cycle arrest via modulating the PI3K/AKT and STAT3 signalling pathways. J. BUON 2019, 24, 1143–1149. [Google Scholar]
  140. Chi, Y.S.; Cheon, B.S.; Kim, H.P. Effect of wogonin, a plant flavone from Scutellaria radix, on the suppression of cyclooxygenase-2 and the induction of inducible nitric oxide synthase in lipopolysaccharide-treated RAW 264.7 cells. Biochem. Pharmacol. 2001, 61, 1195–1203. [Google Scholar] [CrossRef]
  141. Wakabayashi, I.; Yasui, K. Wogonin inhibits inducible prostaglandin E(2) production in macrophages. Eur. J. Pharmacol. 2000, 406, 477–481. [Google Scholar] [CrossRef]
  142. Wu, Q.; Xie, S.; Zhu, Y.; Chen, J.; Tian, J.; Xiong, S.; Wu, C.; Ye, Y.; Peng, Y. Wogonin strengthens the therapeutic effects of mesenchymal stem cells in DSS-induced colitis via promoting IL-10 Production. Oxid. Med. Cell Longev. 2021, 2021, 5527935. [Google Scholar] [CrossRef]
  143. Wang, W.; Xia, T.; Yu, X. Wogonin suppresses inflammatory response and maintains intestinal barrier function via TLR4-MyD88-TAK1-mediated NF-κB pathway in vitro. Inflamm. Res. 2015, 64, 423–431. [Google Scholar] [CrossRef] [PubMed]
  144. Huynh, D.L.; Ngau, T.H.; Nguyen, N.H.; Tran, G.B.; Nguyen, C.T. Potential therapeutic and pharmacological effects of wogonin: An updated review. Mol. Biol. Rep. 2020, 47, 9779–9789. [Google Scholar] [CrossRef] [PubMed]
  145. Tabernero, A.; Reimund, J.M.; Chasserot, S.; Muller, C.D.; Andriantsitohaina, R. Cyclooxygenase-2 expression and role of vasoconstrictor prostanoids in small mesenteric arteries from patients with Crohn’s disease. Circulation 2003, 107, 1407–1410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Huang, H.Y.; Zhang, Z.J.; Cao, C.B.; Wang, N.; Liu, F.F.; Peng, J.Q.; Ren, X.J.; Qian, J. The TLR4/NF-κB signaling pathway mediates the growth of colon cancer. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 3834–3843. [Google Scholar] [PubMed]
  147. Capece, D.; Verzella, D.; Flati, I.; Arboretto, P.; Cornice, J.; Franzoso, G. NF-κB: Blending metabolism, immunity, and inflammation. Trends Immunol. 2022, 43, 757–775. [Google Scholar] [CrossRef]
  148. Kim, D.H.; Sung, B.; Chung, H.Y.; Kim, N.D. Modulation of colitis-associated colon tumorigenesis by baicalein and betaine. J. Cancer Prev. 2014, 19, 153–160. [Google Scholar] [CrossRef]
  149. Toiyama, Y.; Araki, T.; Yoshiyama, S.; Hiro, J.; Miki, C.; Kusunoki, M. The expression patterns of Toll-like receptors in the ileal pouch mucosa of postoperative ulcerative colitis patients. Surg. Today 2006, 36, 287–290. [Google Scholar] [CrossRef]
  150. Lu, Y.; Li, X.; Liu, S.; Zhang, Y.; Zhang, D. Toll-like receptors and inflammatory bowel disease. Front. Immunol. 2018, 9, 72. [Google Scholar] [CrossRef] [Green Version]
  151. Dejban, P.; Nikravangolsefid, N.; Chamanara, M.; Dehpour, A.; Rashidian, A. The role of medicinal products in the treatment of inflammatory bowel diseases (IBD) through inhibition of TLR4/NF-kappaB pathway. Phytother. Res. 2021, 35, 835–845. [Google Scholar] [CrossRef]
  152. Zhang, W.; Shen, X.; Xie, L.; Chu, M.; Ma, Y. MicroRNA-181b regulates endotoxin tolerance by targeting IL-6 in macrophage RAW264.7 cells. J. Inflamm. 2015, 12, 18. [Google Scholar] [CrossRef] [Green Version]
  153. Michel, H.E.; Menze, E.T. Tetramethylpyrazine guards against cisplatin-induced nephrotoxicity in rats through inhibiting HMGB1/TLR4/NF-κB and activating Nrf2 and PPAR-γ signaling pathways. Eur. J. Pharmacol. 2019, 857, 172422. [Google Scholar] [CrossRef] [PubMed]
  154. Pfeffer, C.M.; Singh, A.T.K. Apoptosis: A target for anticancer therapy. Int. J. Mol. Sci. 2018, 19, 448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Lopez, J.; Tait, S.W. Mitochondrial apoptosis: Killing cancer using the enemy within. Br. J. Cancer 2015, 112, 957–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef]
  157. Dogan, E.; Kara, H.G.; Kosova, B.; Cetintas, V.B. Metastasis; Sergi, C.M., Ed.; Exon Publications: Brisbane, Australia, 2022; Chapter 12; p. 163. [Google Scholar]
  158. Kaloni, D.; Diepstraten, S.T.; Strasser, A.; Kelly, G.L. BCL-2 protein family: Attractive targets for cancer therapy. Apoptosis 2022. [Google Scholar] [CrossRef]
  159. Salcher, S.; Hagenbuchner, J.; Geiger, K.; Seiter, M.A.; Rainer, J.; Kofler, R.; Hermann, M.; Kiechl-Kohlendorfer, U.; Ausserlechner, M.J.; Obexer, P. C10ORF10/DEPP, a transcriptional target of FOXO3, regulates ROS-sensitivity in human neuroblastoma. Mol. Cancer 2014, 13, 224. [Google Scholar] [CrossRef] [Green Version]
  160. Sanaei, M.J.; Baghery Saghchy Khorasani, A.; Pourbagheri-Sigaroodi, A.; Shahrokh, S.; Zali, M.R.; Bashash, D. The PI3K/Akt/mTOR axis in colorectal cancer: Oncogenic alterations, non-coding RNAs, therapeutic opportunities, and the emerging role of nanoparticles. J. Cell Physiol. 2022, 237, 1720–1752. [Google Scholar] [CrossRef]
  161. Narayanankutty, A. PI3K/ Akt/ mTOR pathway as a therapeutic target for colorectal cancer: A review of preclinical and clinical evidence. Curr. Drug Targets 2019, 20, 1217–1226. [Google Scholar] [CrossRef]
  162. Jang, J.Y.; Im, E.; Kim, N.D. Mechanism of resveratrol-induced programmed cell death and new drug discovery against cancer: A Review. Int. J. Mol. Sci. 2022, 23, 13689. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of baicalin (A), baicalein (B), wogonoside (C), and wogonin (D).
Figure 1. Chemical structures of baicalin (A), baicalein (B), wogonoside (C), and wogonin (D).
Ijms 24 01954 g001
Table 1. The effect of baicalin on inflammatory bowel disease and colorectal cancer.
Table 1. The effect of baicalin on inflammatory bowel disease and colorectal cancer.
DiseaseAnimal/Cell ModelsDose and TimesUp-RegulationDown-RegulationRefs.
UCDSS-induced UC mice100 mg/kg, 5 days CD14, IL-6[84]
150 mg/kg/d, 30 daysIκB-αMPO, NO, IL-1β, IL-6,
TNF-α, IL-33, NF-κB p65, p-NF-κB p65, p-IκB-α
[98]
100 mg/kg, twice daily for 7 daysIL-10TLR2, TLR4, TLR9, NF-κB p65, IL-6, IL-13, TNF-α,[99]
20 mg/kg, 13 daysCDX2, MDR1 (colon), Cyp3a11 (colon)TNF-α, IL-6[100]
TNBS-induced UC rat100 mg/kg,
every 2 days
for 14 days
ZO-1, occludin, SOD, GSH, IL-10, Foxp3,
acetic acid (fecal), propionic acid (fecal), butyric acid (fecal)
MUC2, ROS, MDA,
Th17/Treg, IL-17,
RORγt
[101]
100 mg/kg/d,
14 days
IL-10, ZO-1, β-cateninp-PI3K/PI3K, p-AKT/AKT, TNF-α,
IL-6, IL-1β
[102]
90 mg/kg, 4 weeksSOD, CAT, GSH-PxMDA, IL-1β, MPO, PGE2, TNF-α, caspase-3 and -9, Bcl-2/Bax, Cyt c,
NF-κB p65, p-IKKβ/IKKβ, p-IKBα/IKBα
[103]
4 mL baicalin liquid (5 mg/mL),
15 days
MPO activity, ICAM-1, MCP-1, COX-2, TNF-α, IL-1β, IL-6[104]
120 mg/kg, 15 daysCAT, GSH-Px, SODMDA, apoptosis rate, ROS[105]
10 mL/kg,
7 days
Foxp3MPO, TNF-α, IL-1β, IL-12, IFN-γ, IL-17, IL-6, IL-10, TGF-β, RORγt[106]
TNBS-induced UC rat (intestinal mucosal cell)100 mg/kg/d,
14 days
Bcl-2caspase-3 and -9, FasL, Bax[102]
UC rats of various complex factors100 mg/kg, 7 daysSOD, ATPAST, ALT, GGT, TG, MDA, NO, cAMP/cGMP, IL-6, IL-1β, IL-17, NF-κB p65, p38[107]
CRCHT29 cells (LPS-treated)50 μM, 2 or 24 h CD14, MyD88, NF-κB p65[84]
100 ng/mL, 30 minIL-10, Bcl-2, ZO-1,
β-catenin
p-PI3K/PI3K, p-AKT/AKT, TNF-α, IL-6, IL-1β, caspase-3 and -9, FasL, Bax[102]
10 μg/mL, 30 minLC3, ATG5, BECN1,
claudin 1
TNF-α, IL-1β, NF-κB p65[108]
HT29 cells150 μM, 48 hcleaved of caspase-3, apoptosis, HIC1, PDCD4, PTEN, E2F2, E-cadherinmiR-10a, miR-23a, miR-30c, miR-31, miR-151a, miR-205, Bcl-2, c-Myc[109]
RKO and HCT116 cells100 μg/mL, 48 hcleaved of caspase-3, -8, -9, and PARP-1, E-cadherin, Cytokeratin 18, Claudin 1, Smad7XIAP, NF-κB, survivin, Bcl-2, p-AKT(Ser473), cyclin D1, cyclin E1, cyclin B1, N-Cadherin, vimentin, snail, slug, twist, TGF-β1, p-Smad2/3, Smad2/3/4, CD133, CD44, SOX2, OCT4, Nanog[110]
SW480 cells400 μg/mL, 24 or 48 hapoptosis, cleaved of caspase-3 and PARP-1Sp1[111]
SW620 cells200 µM, 24 hsub-G1, caspase-3, -8, and -9 activity, ROS [112]
50 μmol/L, 48 h p-PI3K, p-AKT, p-GSK-3β[113]
HCT116, SW480, and HT29 cells20 µM, 72 hapoptotic cells, p-ERK, p-p38 [89]
HCT116 xenograft200 mg/kg, 32 daysE-cadherin, cleaved of caspase-3 and PARP-1Ki67, vimentin, N-cadherin, CD44, CD133, cyclin D1, cyclin B1[110]
HT29 xenograft100 mg/kg, 3 weekscleaved of caspase-3, miR-204Ki67, c-Myc, miR-10a, miR-23a, miR-30c, miR-31, miR-151a[109]
MiscellaneousRAW 264.7cells (LPS-treated)50 μM, 2 or 24 h NO, IL-6, MHC II,
MyD88, NF-κB p65, CD14
[84]
1.0 μmol/L, 24 h HMGB1, TLR4, p-IκBα, p-p65, p65 (nuclear),
IL-1β, IL-6, COX, iNOS
[114]
p-p65/p-65,
p-IKBα/IKBα
[103]
1.0 × 10−5 MCAT, GSH-Px, SOD, Bcl-2MDA, apoptosis rate, TGF-β1, caspase-3 and -9, cleaved caspase-3 and -9,
Bax, Fas, FasL
[105]
5 × 10−4 MIκBTLR4, NF-κB, p-NF-κB, p-IκB, ICAM-1, MCP-1, COX-2, TNF-α, IL-1β, IL-6[104]
LPS-treated mice100 mg/kg (i.p.),
3 days
CD14, IL-6[84]
Mouse peritoneal macrophages (LPS-treated)50 μM, 24 hIL-10, Arg-1, IRF4TNF-α, IL-23, IRF5, M1/M2 macrophage ratio[115]
AKT, protein kinase B; ALT, alanine aminotransferase; Arg-1, arginase-1; AST, aspartate aminotransferase; ATG5, autophagy-related gene 5; ATP, adenosine triphosphate; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma 2; cAMP, cyclic adenosine monophosphate; CD14, cluster of differentiation 14; CDX2, caudal type homeobox 2; cGMP, cyclic guanosine monophosphate; CAT, catalase; COX, cyclooxygenase; CRC, colorectal cancer; Cyt c, cytochrome c; DSS, dextran sulfate sodium; ERK, extracellular signal-regulated kinase; FasL, Fas Ligand; Foxp3, forkhead box p3; GGT, γ-glutamyl transpeptidase; GSH, glutathione; GSH-Px, glutathione peroxidase; HIC1, hypermethylated in cancer 1; HMGB1, high-mobility group box 1; IFN-γ, interferon gamma; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha; ICAM-1, intercellular adhesion molecule-1; IKKβ, inhibitor of nuclear factor kappa-B kinase subunit beta; IL-, interleukin; iNOS, inducible nitric oxide synthase; i.p., intraperitoneal injection; LC3, microtubule-associated protein 1A/1B-light chain 3; LPS, lipopolysaccharide; MCP-1, monocyte chemoattractant protein 1; MDA, malondialdehyde; MDR1, multidrug resistance protein 1; MyD88, myeloid differentiation factor 88; MPO, myeloperoxidase; NF-κB p65, nuclear factor kappa-light-chain-enhancer of activated B cells p65; NO, nitric oxide; MHC II, major histocompatibility complex class II; HMGB1, high mobility group box 1; p-, phosphorylation; OCT4, octamer-binding transcription factor 4; PARP, poly(ADP-ribose) polymerase; PDCD4, programmed cell death protein 4; PGE2, prostaglandin E2; PI3K, phosphoinositide 3-kinase; PTEN, phosphatase and tensin homologue deleted on chromosome ten; ROS, reactive oxygen species; RORγt, retinoic acid-related orphan receptor gamma t; SOD, superoxide dismutase; SOX2, sex determining region Y-box 2; TG, triglyceride; TGF-β1, transforming growth factor beta-1; TNF-α, tumor necrosis factor α; TLR, toll-like receptor; TNBS, trinitrobenzene sulphonic acid; UC, ulcerative colitis; XIAP, X-linked inhibitor of apoptosis protein; ZO-1, zonula occludens-1.
Table 2. The effect of baicalein on inflammatory bowel disease and colorectal cancer.
Table 2. The effect of baicalein on inflammatory bowel disease and colorectal cancer.
DiseaseAnimal/Cell ModelsDose and TimesUp-RegulationDown-RegulationRefs.
UCDSS-induced UC mice25 mg/kg, 7 daysIκBαCOX-2, iNOS, cyclin D1, p-IκBα, p-p65, p65, IKKβ,
NF-κB, p-STAT3, STAT3
[85]
20 mg/kg,
10 days
IL-4IFN-γ[116]
40 mg/kg,
10 days
CYP1A1IL-6, IL-10, IL-17, IL-22, TNF-α, TNF-β[117]
20 mg/kg,
13 days
CDX2, MDR1 (colon), Cyp3a11 (colon)TNF-α, IL-6[100]
UC rats of various complex factors100 mg/kg, 7 daysSOD, ATPAST, ALT, GGT, TG, MDA, NO, cAMP/cGMP, IL-6, IL-1β, IL-17, NF-κB p65, p38[107]
CDTNBS-induced CD mice20 mg/kg/d, 10 days iNOS, ICAM-1, MCP-1, COX-2, TNF-α, IL-1β, TLR4, MyD88, p-p65, p-IκBα, p-p38[118]
CRCHCT116 cells60 µM, 48
h
apoptosis, caspase-3 and - 9 [119]
100 µM, 24 hcleavage of PARP, PPARγpro-caspase-3, -8, and -9, p-IκBα, iNOS, p50 (nuclear), p65 (nuclear), MMP-2 and -9[120]
40 µM, 4
h
p-Nrf2/Nrf2[121]
40 µmol/L, 24 or 48 hp53, p21Ezrin, cyclin D1, CDK4[122]
HCT116, A549, and Panc-1 cells40 µM, 48 hapoptotic rate, cleaved of caspase-3 and -9, DEPP, GADD45A, p-JNK, p-ERK, p-p38 [123]
HCT116, SW480, and HT29 cells20 µM, 72 hapoptotic cells, hTERT, p-ERK, p-p38 [89]
HT29 cells100 µM, 48 hcleavage of PARP and
caspase-3, Bax, p53
Bcl-2, p-AKT, p-caspase-
9, p-GSK-3β, survivin,
cyclin D1, cyclin B1
[124]
HT29 xenograft10 mg/kg three times/week, 43 daysapoptotic cells, p53, p21 [124]
LS174T cells25 µM, 48 hPXR, CDX2 [100]
DLD-1 cells30 µM, 24 h MMP-2 and -9, p-AKT[125]
HT29 and DLD1 cells30 µM, 24 h MMP-2 and -9, p-ERK[126]
DLD1 xenograft20 mg/kg/day, 21 days MMP-2 and -9, p-ERK[126]
HCT116 xenograft50 mg/kg, 3 weekly cycle Ki67[122]
MiscellaneousRAW 264.7cells (LPS-treated)50 μM, 2 h or 48 h TLR4, MyD88, IRAK-1,
COX-2, NO, iNOS,
p-JNK, p-ERK1/2, p-p38, MD-2/TLR4
[118]
80 μM, 2 hp-ERK, p-p38, p-JNK, p- JAK1, p-JAK2iNOS, TNF-α, IL-6, IL-1β, NO, PGE2, p-STAT1(Tyr701), p-STAT3(Tyr705), p-STAT3(Ser727)[127]
10 μM, 2 h TNF-α, COX-2, iNOS,
NO, IL-1β, PGE2, p-IκBα
[128]
RAW 264.7 cells (polyinosinic–polycytidylic-acid-treated)100 µM, 24 h NO, calcium release, IL-1α, IL-6, G-CSF, GM-CSF, VEGF, MCP-1, IP-10, LIX, RANTES,
STAT1, LIX, STAT3, CHOP, Fas
[129]
THP-1 cells (LPS-treated)50 μM, 2 h iNOS, COX-2, IL-1α, IL-1β, NLRP3, ASC, caspase-1[118]
EL-4 cells50 μM, 24 hCYP1A1, AHR (nuclear)AHR (cytosol)[117]
colonic lamina propria lymphocyte40 mg/kg,
10 days
CD4+CD25+Foxp3+ T cellsCD4+IL-17+ T cells[117]
mesenteric lymph of mice40 mg/kg,
10 days
CD4+CD25+Foxp3+ T cellsCD4+IL-17+ T cells[117]
AHR, aryl hydrocarbon receptor; AKT, protein kinase B; ALT, alanine aminotransferase; ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; AST, aspartate aminotransferase; ATP, adenosine triphosphate; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma-2; cAMP, cyclic adenosine monophosphate; CD, Crohn’s disease; CDK4, cyclin-dependent kinase4; CDX2, caudal-type homeobox 2; cGMP, cyclic guanosine monophosphate; CHOP, C/EBP-homologous protein; COX-2, cyclooxygenase-2; CRC, colorectal cancer; CYP1A1, Cytochrome P450, family 1, subfamily A, polypeptide 1; DEPP, decidual protein induced by progesterone; DSS, dextran sulfate sodium; ERK, extracellular signal-regulated kinase; GADD45A, growth arrest and DNA-damage-inducible 45 alpha; G-CSF, granulocyte colony-stimulating factor; GGT, γ-glutamyl transpeptidase; GM-CSF, granulocyte macrophage colony-stimulating factor; GSK-3β, glycogen synthase kinase-3 beta; hTERT, human telomerase reverse transcriptase; ICAM-1, intercellular adhesion molecule-1; IFN-γ, interferon gamma; IKKβ, inhibitor of nuclear factor kappa-B kinase subunit beta; IL-, interleukin; iNOS, inducible nitric oxide synthase; IRAK-1, inhibition of interleukin-1 receptor-associated kinase 1; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; p-, phosphorylation; JAK, Janus protein tyrosine kinase; JNK, c- Jun N-terminal kinase; LIX, lipopolysaccharide-induced CXC chemokine; LPS, lipopolysaccharide; MCP-1, monocyte chemotactic protein-1; MDA, malondialdehyde; MDR1, multidrug resistance protein 1; MD-2, myeloid differentiation protein-2; MMP, matrix metalloproteinase; MyD88, myeloid differentiation factor 88; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NO, nitric oxide; NLRP3, nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3; Nrf2, nuclear factor erythroid-2-related factor 2; RANTES, regulated upon activation, expressed by normal T cell and presumably secreted; p-, phosphorylation; PARP, poly(ADP-ribose) polymerase; PGE2, prostaglandin E2; PPARγ, peroxisome proliferator-activated receptors γ; PXR, pregnane X receptor; RANTES, chemokine ligand 5; SOD, superoxide dismutase; STAT, signal transducer and activator of transcription; TNF, tumor necrosis factor; TLR4, toll-like receptor 4; TNBS, trinitrobenzene sulphonic acid; TG, triglyceride; VEGF, vascular endothelial growth factor; UC, ulcerative colitis.
Table 3. The effect of wogonoside on inflammatory bowel disease and colorectal cancer.
Table 3. The effect of wogonoside on inflammatory bowel disease and colorectal cancer.
DiseaseAnimal/Cell ModelsDose and TimesUp-RegulationDown-RegulationRefs.
UCDSS-induced UC mice50 mg/kg, 10 daysoccludin, ZO-1, Claudin1, MLC2IL-13, IFN-γ, MLCK, p-MLC2[130]
50 mg/kg, 10 daysNF-κB p65 (cytosol)MPO activity, iNOS activity, CD11b+F4/80+ monocyte/macrophage, CD11b+Gr-1+ neutrophils, IL-1β, TNF-α, and IL-6 (serum, colonic),
IL-18, IFN-γ, and MIP-1α (colonic), NF-κB p65 (nucleus), p-IkBα (cytosol), p-p65, NF-kB DNA binding activity, cleaved caspase-1 and IL-1β, NLRP3, ASC, caspase-1 activity
[131]
THP-1 cells (LPS-treated)50 µM,
6 h
NF-κB p65 (cytosol)IL-1β, IL-6, TNF-α, NF-κB p65 (nucleus), p-IkBα (cytosol), p-p65, NF-kB DNA binding activity, NLRP3, pro-caspase-1, cleaved caspase-1 and IL-1β, NLRP3, ASC, caspase-1 activity[131]
Caco-2 cells (TNF-α exposure)50 µM,
72 h
occludin, ZO-1, Claudin1, TER, MLC2, F-actinFD4, MLCK, p-MLC2[130]
CRCAOM/DSS mouse100 mg/kg,
105 days
apoptotic cells, NF-κB (cytoplasm)neutrophil (Gr-1+ positive cells), macrophage (F4/80+ positive cells), Ki-67, BrdU, PCNA, IL-1β, IL-6, TNF-α, NF-κB, NF-κB (nucleus), p-p65, PI3K, p-AKT/AKT, p-IKKα/IKKα, p-IkBα/ IkBα, cyclin D1, survivin[132]
HCT116 and HT29 cells (conditioned media from LPS-treated THP-1 cells)150 µM, 24 hNF-κB (cytoplasm)PCNA, p-IKKα/IKKα, p-IκBα/IκBα, cyclin D1, survivin, NF-κB (nucleus), NF-κB fluorescence, NF-κB luciferase activity, PI3K, p-AKT/AKT[132]
LOVO cells62.5 µM, 48 hcaspase-3 and -9, bax, LC3, p62Bcl-2, PI3K, p-AKT, p-mTOR, p-p70S6K[90]
MiscellaneousRAW 264.7 cells (LPS-treated)50 μM, 24 h NO, PGE2, TNF-α, IL-6, iNOS, COX-2[133]
AKT, protein kinase B; AOM, azoxymethane; ASC, apoptosis-associated speck-like protein containing a CARD (caspase recruitment domain); Bax, Bcl-2-associated X protein; BrdU, bromodeoxyuridine; COX-2, cyclooxygenase-2; CRC, colorectal cancer; DSS, dextran sulfate sodium; ERK, extracellular signal-regulated kinase; FD4, FITC-dextran; IFN-γ, interferon gamma; IL-, interleukin; IKKα, IκB kinase alpha; iNOS, inducible nitric oxide synthase; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha; JNK, c-Jun N-terminal kinase; LC3, microtubule-associated protein 1A/1B-light chain 3; LPS, lipopolysaccharide; MIP-1α, macrophage inflammatory proteins-1α; MLC2, myosin light chain2; MLCK, myosin light chain kinase; MPO, myeloperoxidase; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NO, nitric oxide; NLRP3, nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3; p-, phosphorylation-; PCNA, proliferating cell nuclear antigen; PGE2, prostaglandin E2; PI3K, phosphoinositide 3-kinases; p70S6K, ribosomal protein S6 kinase; TER, transmembrane resistance; TNF-α, tumor necrosis factor-α; UC, ulcerative colitis; ZO-1, zonula occludens-1.
Table 4. The effect of wogonin on inflammatory bowel disease and colorectal cancer.
Table 4. The effect of wogonin on inflammatory bowel disease and colorectal cancer.
DiseaseAnimal/Cell ModelsDose and TimesUp-RegulationDown-RegulationRefs.
UCDSS-induced UC mice30 mg/kg, 7 daysGST, GSH, SOD, IL-10 (tissue), Bax, caspase-3 and -9, Nrf2, HO-1MPO, NO, TBARS, TNF-α (serum, tissue), IL-6 (serum, tissue), PGE2 (tissue), Bcl-2, COX-2, iNOS, TLR4, p65[134]
CRCAOM/DSS mouse100 mg/kg, 25 weekscleavage of
caspase-3 and -9, IRE1α, PDI
[135]
60 mg/kg,
105 days
IOD of Nrf2-positive cells (surrounding tissues), NF-kB p65 (cytoplasm), Nrf2 (nuclear)IOD of BrdU-, PCNA-, and NF-kB-positive cells (surrounding tissues, tumor tissues),
IOD of IL-6- and 1β-positive cells, IOD of Nrf2-positive cells (tumor tissues), NF-kB p65 (nuclear)
[136]
HCT116 cells (cocultured with LPS-treated THP-1 cells)100 µM, 12 h IL-6, IL1β, PCNA, NF-kB (nuclear), p-IκB, p-IKK(α/β) [136]
HCT116 cells (LPS-treated)50 µM, 8 hNrf2 (nuclear), NQO-1, HO-1NF-kB (nuclear), p-IκB, p-IKKα/β, p-p38, p-ERK, p-JNK, p-AKT, PI3K, Nrf2 (cytoplasm)[136]
HCT116 cells40 µM, 24 hp21, p27, E2F1, p-Rb, GSK-3β, AXIN, p-β-catenin (Ser33/37/Thr41), apoptotic rate cyclin A, cyclin E, cyclin D1, CDK2, CDK4, CDK8, Rb, Wnt3a, Dvl2, LRP6, p-GSK-3β, β-catenin, β-catenin (cytosolic), β-catenin (nuclear), TCF1, TCF3, TCF4, LEF1, c-Myc, survivin [137]
10 µM, 72 hcleavage of
caspase-3 and -9, apoptotic cells, IRE1α, calnexin, PDI, CHOP, DDIT3, XBP1, p53(Ser376), p53(Ser376) (cytoplasm), p53(Ser315)
colony formation rate, LC3-II, ATG12, p53(Ser376) (nuclear), p-p53(Ser15) (nuclear/ cytoplasm)[135]
40 µM, 24 hp53, TIGAR, p-p53(Ser15), p-p53(Ser20), Ac-p53(Lys382)glucose uptake, lactate generation, ATP production, PGM, GLUT1, HK2, PDHK1, LDHA, MDM2, [138]
HCT116 cells xenograft60 mg/kgE2F1β-catenin, cyclin D1, c–Myc, TCF3 [137]
HT29 cells100 µM, 48 hcleavage of PARP and
caspase-3, Bax, p53,
Bcl-2, p-AKT, p-caspase-9, p-GSK-3β, survivin,
cyclin D1, cyclin B1
[124]
HT29 xenograft10 mg/kg three times/week, 43 daysapoptotic cells, p53, p21 [124]
SW48 cells16 μM LC3-II, cleavage of
caspase-3, -8, -9, and PARP, PI3K
p-PI3K, p-AKT, p-STAT3(Tyr705), p-STAT3(Ser727)[139]
SW-480 cells60 μM, 48 hapoptotic cells, caspases-3, -8, and -9 activity [96]
THP-1 cells (LPS-treated)100 µMNrf2 (nuclear), Keap1, HO-1Nrf2 (cytosolic)[136]
MiscellaneousRAW 264.7 cells (LPS-treated)100 µM, 24 h NO, PGE2, iNOS, COX-2[140]
50 µM, 24 h PGE2, COX-2, COX-2 activity[141]
DSS + MSCs10 mg/kg, 8 daysIL-10TNF-α[142]
MSCs + LPS50 μM, 24 hROS, HIF-1α, IL-10, p-GSK-3β, p-GSK-3β/GSK-3β, p-AKT, p-AKT/AKT [142]
Caco-2 cells (LPS-treated)50 μMTEER, ZO-1, claudin-1, IκBFD4, FD10, FD20, IL-1β, IL-6, IL-8, iNOS, COX-2, NF-κB p65, p-IκB, TLR4, MyD88, p-TAK1[143]
Ac-, acetylation; AKT, protein kinase B; AOM, azoxymethane; ATG12, autophagy-related gene 12; ATP, adenosine triphosphate; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma-2; CDK4, cyclin-dependent kinase4; COX-2, cyclooxygenase-2; CHOP, C/EBP-homologous protein; CRC, colorectal cancer; DDIT3(CHOP), DNA damage-inducible transcript 3; DSS, dextran sulfate sodium; Dvl2, disheveled; ERK, extracellular signal-regulated kinase; E2F1, E2F transcription factor 1; FD, fluorescein isothiocyanate-dextran; GLUT1, glucose transporter 1; GSK-3β, glycogen synthase kinase-3 beta; GSH, glutathione; GST, glutathione-s-transferase; HIF-1α, hypoxia-inducible factor 1-alpha; HK2, hexokinase 2; HO-1, heme oxygenase-1; IL-, interleukin; iNOS, inducible nitric oxide synthase; IOD, integrated optical density; IRE1α, inositol-requiring transmembrane kinase endoribonuclease-1 alpha; JNK, c-Jun N-terminal kinase; Keap1, Kelch-like ECH-associated protein 1; LC3-II, microtubule-associated protein 1A/1B-light chain 3-II; LDHA, lactate dehydrogenase A; LEF1, lymphoid enhancer factor1; LPS, lipopolysaccharide; LRP6, low-density lipoprotein receptor-related protein 6; MDM2, mouse double minute 2 homolog; MPO, myeloperoxidase; MSCs, mesenchymal stem cells; MyD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NQO-1, NADPH quinone oxidoreductase-1; NO, nitric oxide; Nrf2, nuclear factor erythroid 2-related factor 2; p-, phosphorylation; PARP, poly(ADP-ribose) polymerase; PCNA, proliferating cell nuclear antigen; PDHK1, pyruvate dehydrogenase kinase 1; PDI, protein disulfide isomerase; PGE2, prostaglandin E2; PGM, phosphoglycerate mutase; PI3K, phosphoinositide 3-kinase; Rb, retinoblastoma; ROS, reactive oxygen species; SOD, superoxide dismutase; STAT3, signal transducer and activator of transcription3; TAK1, transforming growth factor beta-activated kinase 1; TBARS, thiobarbituric acid reactive substances; TCF, T cell factor; TEER, transepithelial electrical resistance; TIGAR, TP53-inducible glycolysis and apoptosis regulator; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor α; UC, ulcerative colitis; XBP1, X-box binding protein 1; ZO-1, zonula occludens-1.
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

Jang, J.Y.; Im, E.; Kim, N.D. Therapeutic Potential of Bioactive Components from Scutellaria baicalensis Georgi in Inflammatory Bowel Disease and Colorectal Cancer: A Review. Int. J. Mol. Sci. 2023, 24, 1954. https://doi.org/10.3390/ijms24031954

AMA Style

Jang JY, Im E, Kim ND. Therapeutic Potential of Bioactive Components from Scutellaria baicalensis Georgi in Inflammatory Bowel Disease and Colorectal Cancer: A Review. International Journal of Molecular Sciences. 2023; 24(3):1954. https://doi.org/10.3390/ijms24031954

Chicago/Turabian Style

Jang, Jung Yoon, Eunok Im, and Nam Deuk Kim. 2023. "Therapeutic Potential of Bioactive Components from Scutellaria baicalensis Georgi in Inflammatory Bowel Disease and Colorectal Cancer: A Review" International Journal of Molecular Sciences 24, no. 3: 1954. https://doi.org/10.3390/ijms24031954

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