Skip to main content

Exploring the immunometabolic potential of Danggui Buxue Decoction for the treatment of IBD-related colorectal cancer

Abstract

Danggui Buxue (DGBX) decoction is a classical prescription composed of Astragali Radix (AR) and Angelicae Sinensis Radix (ASR), used to enrich blood, and nourish Qi in Chinese medicine, with the potential to recover energy and stimulate metabolism. Chronic inflammation is a risk factor in the development of inflammatory bowel disease (IBD)-related colorectal cancer (CRC). More importantly, AR and ASR have anti-inflammatory and anti-cancer activities, as well as prefiguring a potential effect on inflammation-cancer transformation. We, therefore, aimed to review the immunometabolism potential of DGBX decoction and its components in this malignant transformation, to provide a helpful complement to manage the risk of IBD-CRC. The present study investigates the multifaceted roles of DGBX decoction and its entire components AR and ASR, including anti-inflammation effects, anti-cancer properties, immune regulation, and metabolic regulation. This assessment is informed by a synthesis of scholarly literature, with more than two hundred articles retrieved from PubMed, Web of Science, and Scopus databases within the past two decades. The search strategy employed utilized keywords such as “Danggui Buxue”, “Astragali Radix”, “Angelicae Sinensis Radix”, “Inflammation”, and “Metabolism”, alongside the related synonyms, with a particular emphasis on high-quality research and studies yielding significant findings. The potential of DGBX decoction in modulating immunometabolism holds promise for the treatment of IBD-related CRC. It is particularly relevant given the heterogeneity of CRC and the growing trend towards personalized medicine, but the precise and detailed mechanism necessitate further in vivo validation and extensive clinical studies to substantiate the immunometabolic modulation and delineate the pathways involved.

Graphical Abstract

Highlights

  • Danggui Buxue decoction is a classical prescription consisting of Astragali Radix and Angelicae Sinensis Radix with the efficacy of tonifying blood and invigorating qi in traditional Chinese medicine.

  • Astragali Radix and Angelicae Sinensis Radix, both have a variety of pharmacological activities, including anti-inflammation and anti-cancer effects.

  • In view of inflammation and malignant transformation in inflammatory bowel disease-related colorectal cancer and the curative effects of Astragali Radix and Angelicae Sinensis Radix, immunometabolism modulation potential of DGBX were reviewed and discussed in the present study.

Danggui Buxue (DGBX) decoction is a classical prescription consisting of Astragali Radix (AR) and Angelicae Sinensis Radix (ASR) in traditional Chinese medicine. Given the documented anti-inflammatory and anti-neoplastic properties, this review aims to discuss the potential of mitigating the inflammation-cancer transformation and to offer an immunometabolic adjunct in IBD-CRC risk management.

Overview of IBD and CRC

Pathogenesis and epidemiological characteristics of IBD

IBD includes ulcerative colitis (UC) and Crohn’s disease (CD) and is a chronic inflammatory disease occurring in the gastrointestinal tract [1]. UC characteristically initiates in the rectum, and subsequently spreads to the entire colon in a continuous manner, while CD predominately involves the terminal ileum and perianal region with a discontinuous pattern of involvement extending throughout the gastrointestinal tract [2]. UC inflammation in the mucous membrane leads to ulcers and bloody diarrhea [3]. CD typically involves abdominal pain, chronic diarrhea, weight of loss, and fatigue [4]. In the past, IBD was regarded as a Western disease; however, in the twenty-first century, the incidence and prevalence of IBD are increasing worldwide. Although still lower than in Western countries, the incidence and prevalence of IBD in Asia is increasing over time [5]. Therefore, comprehending the evolving epidemiological patterns and pathogenesis thereof is crucial in addressing the escalating global burden. The pathogenesis of IBD is related to heredity, the intestinal microbe, the environment, and immunity [6]. Genome-wide associated studies of genes and genetic loci involved in IBD identified 99 non-overlapping genetic risk loci and revealed the exact role of disease-related genes. Nucleotide-binding oligomerization domain containing protein 2 (NOD2), for example, is appropriately regulated to maintain intestinal homeostasis [2]. Tens of thousands of microbes living in the human gut are involved in the regulation of health and disease [7], and the human gut contains more than 2000 species of microbes, including Firmicutes, Bacteroides, Actinomycetes, and Proteus [8]. Patients with IBD have significantly less microbial diversity than that of healthy individuals [9]. Environmental determinants, such as tobacco smoking, appendectomy, oral contraceptive use, and dietary habits, exert distinct influences on the risk profiles for celiac disease and CD. Appendectomy has different effects on UC and CD, with a general protective effect and reduced risk of UC, and with an increased risk of stenosis and reduced risk of anal fistula in CD [10, 11]. Contraceptive use in women with a history of smoking is also associated with the occurrence and development of IBD [12, 13]. Even though there is no conclusive evidence that dietary factors are directly related to the pathogenesis of IBD, low-fiber and high-fat foods have been proposed as risk factors [14]. In healthy people, the initial immune response is rigorously regulated, this regulation determines immune tolerance or defensive inflammatory responses, and some disturbances in the balance of these responses may lead to IBD [15]. IBD in patients who are failing to achieve effective disease control may ultimately lead to the development of cancer.

Pathogenesis and epidemiological characteristics of colorectal cancer

CRC is the fourth leading cause of cancer-related deaths and the most common malignancy worldwide [16]. In 2020, there were nearly 4.56 million newly diagnosed cancer cases and 30 million cancer deaths in China [17]. Of the 147,000 people diagnosed with colorectal cancer, approximately 53,000 will eventually die. Despite variations in CRC incidence and mortality by age, ethnicity, and geographic location, a concerning trend of escalating incidence and mortality rates has been observed for CRC [18]. The susceptibility to CRC is influenced by a spectrum of individual-specific factors, encompassing age, lifestyle, and a history of chronic disease. IBD patient are notably at an elevated risk for the development of CRC. Chronic inflammation is postulated to foster aberrant cell proliferation, and prolonged exposure to inflammation can lead to cellular atypia, potentially culminating in the formation of neoplastic lesion [19,20,21]. The occurrence and development of CRC goes through several stages, including normal mucosal epithelium, abnormal crypt foci, microadenoma, and finally the malignant tumor. The progression from normal mucosal epithelium to abnormal crypts is ordinarily considered to be the onset of dysplasia, and a single dysplasia crypt is considered the first histological manifestation of a tumor [22]. Adenomatous polyps progressing to sporadic CRC typically undergo a protracted period of development, and CRC associated with colitis is believed to evolve through multiple stages of precursor lesions, ranging from inflammation to low-grade dysplasia, high-grade dysplasia, and finally, CRC [23]. CRC is not an abrupt occurrence; hence, timely detection and treatment during its formation can effectively prevent it.

Risk and epidemiological characteristics of CRC in IBD

Patients with IBD have an elevated risk of developing CRC, and chronic inflammation leads to dysplastic precursor lesions that may appear in multiple regions of the colon through a local carcinization process. Patients with IBD are at 2–6 times higher risk of developing CRC compared to the general population. IBD-related colorectal cancer accounts for approximately 2% of total annual CRC mortality and 10–15% of annual mortality in patients with IBD [24]. The pathogenesis of IBD-related CRC diverges from that of sporadic CRC, typically manifesting through a distinct sequence characterized by chronic inflammation, dysplastic transformation, and eventual carcinomatous progression. Research has demonstrated that intestinal inflammation can lead to the dysregulation of the host's immune response and a disruption in the homeostasis of the intestinal microbiota. The gut microbiota plays a crucial role in maintaining intestinal homeostasis by impeding pathogen colonization and modulating immune cell networks. Bacteroides fragilis, Fusobacterium nucleatum, and Porphyromonas gingivalis are known to be closely related to IBD-CRC [25]. Intestinal microbiota and their metabolites modulate the metabolic pathways of immune cells, thereby ameliorating IBD within the gastrointestinal tract and augmenting the efficacy of CRC immunotherapy [26,27,28,29]. Colonoscopy and staging biopsies should be performed in patients with long-term IBD since early detection of dysplasia is critical for the prevention of CRC [30]. A slow transition from IBD to cancer is associated with chronic inflammation, so reducing inflammation caused by colitis is a preventive approach and strategy to decrease risk of IBD-CRC [31,32,33]. Chemoprophylaxis is also one of the main means of continuous and complete control of inflammation [34, 35]. The risk of IBD patients developing CRC has decreased recently, which may be due to early monitoring and appropriate treatments.

Immunometabolism regulation in IBD-related CRC

Immune regulation in the tumor microenvironment (TME)

The TME is a cellular environment in which the tumor exists, and the continuous interaction between tumor cells and the surrounding microenvironment plays a crucial role in the genesis, progression, and metastasis of tumors. This complex microenvironment consists of tumor cells, stromal cells, and extracellular matrix. Stromal cells include immune cells and the cytokines or chemokines secreted by these cells [36, 37]. Immune cells play an important role in tumorigenesis, including innate immune cells, such as natural killer (NK) cells, macrophages, dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), and adaptive immune cells, such as T cells and B cells [38, 39]. The cytotoxic activity of NK cells is primarily mediated through two well-characterized mechanisms, one is the release of cytotoxic granules containing perforin and granzymes, and the other is the secretion of pro-inflammatory cytokines. NK cells from IBD patients exhibit a diminished production of interferon-gamma (IFN-γ), yet an increased secretion of tumor necrosis factor-alpha (TNF-α) [40]. Elevated levels of TNF-α have been correlated with the presence of aberrant crypt foci within colorectal polyps [41]. The dynamic equilibrium between M1 and M2 macrophage polarization is a critical determinant of the inflammatory microenvironment and has profound implications for tumor and inflammation [42,43,44,45]. Clinical observations have highlighted a significant association between the overexpression of M2 macrophages and the progression of CRC [46, 47]. DCs function as specialized antigen presenting cells, whereas MDSCs consist of monocytes and polymorphonuclear immature bone marrow cells. In the CRC microenvironment, MDSCs represent the predominant immunosuppressive cell population within the TME and play a critical role in promoting immune resistance [48,49,50,51,52,53]. T cells play a pivotal role in orchestrating the immune response against CRC, rendering them one of the most critical components of immune system. Activated CD8+ T cells have cytotoxic effects on CRC cells, while activated CD4+ T cells can differentiate into subtypes that promote or inhibit tumor growth. Tumor-infiltrating B lymphocytes are considered the main effector cells of the humoral adaptive immune response, and B cells are recognized in the immune system for their ability to produce antibodies and secrete pro-inflammatory and anti-inflammatory cytokines regulating CRC progression [54]. Immune cells play a multifaceted role in the pathogenesis of CRC, influencing the survival, proliferation, and metastatic potential of CRC cells, and actively participating in the regulation of cancer progression. The activation and differentiation of these immune cells are accompanied by significant metabolic reprogramming, which is essential for their functional capabilities. The unique metabolic characteristics of immune cells also have a profound impact on their ability to perform their immune functions [38, 55].

Immunometabolism aspects in the TME

Metabolic dysregulation is a defining characteristic of cancer cells and significantly influences the development and progression of CRC. Immunometabolism, the interplay between immune cell function and metabolism, is a critical determinant in cancer progression, particularly in the context of CRC [56]. Abnormal metabolic pathways of cancer include fatty acid, glucose, and amino acid metabolism. Other metabolic pathways include the one-carbon metabolism, pentose phosphate pathway, and nicotinamide adenine dinucleotide phosphate metabolism [57,58,59,60]. Metabolism and immunity are both important components in maintaining the normal operation of human body. They reinforce each other, and the components complement one another, as shown in Fig. 1. Glycolytic metabolism is the process of converting glucose uptake from the extracellular environment to pyruvate and releasing adenosine triphosphate (ATP) [61]. T-cell activation significantly increases glycolytic flux and transports glycolytic pyruvate into the tricarboxylic acid (TCA) cycle [62]. The metabolic profile of CD4+ T cells significantly influence their immune functions, which in turn, can modulate the pathogenesis of IBD [63, 64]. The macrophages undergo differentiation into either M1 or M2 cells [65].In M1 macrophages, the TCA cycle results in metabolite accumulation and enhances cell immune function. Fatty acid oxidation regulates the balance between inflammatory effector and suppressor T cells. Increased fatty acid oxidation and oxidative phosphorylation support Treg differentiation and function. Treg accumulates in inflamed tissues of colitis and is involved in the progression of CRC [66]. The differentiation of M2 macrophages also depends on the fatty acid oxidation program. The fatty acid synthesis pathway produces lipids, which are essential for cell growth and proliferation. Fatty acid synthesis also links innate and adaptive immunity by regulating DCs function. Amino acid metabolism is closely related to the mTOR pathway and nucleotide synthesis, and the metabolism of glutamine, arginine, and tryptophan regulates the activity of immune cells. The intricate metabolic demands shared by cancer and immune cells imply that effective targeting on cancer metabolism necessitates consideration of gene type, tumor type, and the composition of the tumor microenvironment. A comprehensive understanding of their respective roles and mechanisms is essential to realize the cancer metabolic therapy. The main regulation of immunometabolism in the TME involves the various critical signaling pathways in immunity. The phosphatidylinositol 3-kinase (PI3K)/AKT (also known as protein kinase B, PKB)/mammalian target of rapamycin (mTOR) and liver kinase B1-5’ (LKB1)-AMP-activated protein kinase (AMPK) signaling pathways are important in regulating immune metabolism [67]. The PI3K/AKT/mTOR signaling cascade is a critical cellular signaling pathway that governs a myriad of cellular processes, including cell growth, proliferation, metabolism, and survival. mTORC1 is highly activated in the intestinal mucosa of IBD patients, and inhibition of mTORC1 is effective in the treatment of UC [68]. mTORC1 subsequently activates the transcription factor hypoxia-inducible factor 1 (HIF1). In macrophages of IBD patients, glycolysis is significantly enhanced by mTORC1 and HIF-1 [69]. HIF-1 promotes glycolysis and cancer-related inflammation by stimulating hexokinase and pyruvate dehydrogenase kinase, co-inducing glycolytic gene expression with other oncogenes or transcription factors. On the flip side, glycolysis affects immature DCs (iDCs) [38, 67, 70, 71]. mTOR is an effector target of AKT signaling that increases glycolysis and reduces lipid oxidation. This pathway is essential for the differentiation of CD4+ T cells into immunologically specific effector T cells (Teff) or the induction of regulatory T-cell (Treg) subsets [72]. As an energy sensor in cells, AMPK activation reduces the levels of mitochondrial aerobic glycolysis and oxidative phosphorylation, and inhibits the migration, invasion, and metastasis formation of CRC cells [38, 73,74,75,76]. Targeting immunometabolism in the TME represents a highly promising therapeutic strategy [55, 77].

Fig. 1
figure 1

The immunometabolism modulation. LKB1-AMPK signaling pathway and PI3K/AKT/mTOR signaling pathway are the main pathways regulating the metabolism of fatty acids and glucose. Consequently, the metabolic outcomes impact immune cells such as T cells, DCs, M1 and M2 macrophages, thereby influencing immunity. Furthermore, immune responses reciprocally recast metabolic regulation

Astragali Radix, Angelicae Sinensis Radix, and DGBX decoction advances

Astragali Radix (AR, Huang Qi in Chinese), the dried root of Astragalus membranaceus (Fisch.), Bge. var. mongholicus (Bge.) Hsiao or Astragalus membranaceus (Fisch.) Bge., and the components isolated and identified included polysaccharides, saponins, flavonoids, and amino acids [78, 79]. As a traditional Chinese medicine employed in clinical treatment, AR exhibits diverse biological activities, including anti-inflammatory and anti-tumor functions [80,81,82,83]. Angelicae Sinensis Radix (ASR, Dang Gui in Chinese) is the root of Angelica sinensis (Oliv.) Diels [84]. The main chemical components of ASR include organic acids, volatile oil, polysaccharides, and flavonoids. It also has a variety of pharmacological activities, including anti-inflammatory activity, cardiac protection, antioxidant activity, and neuroprotection, as well as functioning in the cardiovascular and cerebrovascular systems [85, 86]. As a Chinese classical prescription, DGBX decoction is recorded with AR and ASR, in a ratio of 5:1. It is a classic recipe to invigorate Qi and tonify the blood [87, 88]. The main effective components in DGBX decoction are polysaccharides, calycosin, formononetin, astragaloside IV, ferulic acid, and ligustilide [89]. DGBX decoction exerts supporting Qi and enriches the blood, enhancing efficacy and reducing toxicity [90]. In recent years, traditional Chinese medicine and the classical prescriptions have been found to be widely used [83, 91], such as AR, ASR, and DGBX decoctions, especially for their anti-cancer activities, immune regulation, and metabolic regulation, as shown in Tables 1, 2 and 3. Accordingly, the schema of the present study is shown in Fig. 2.

Table 1 The main active ingredients derived from AR and ASR or the corresponding TCM prescriptions
Table 2 Immunomodulatory effects of active ingredients derived from AR and ASR or the corresponding TCM prescriptions
Table 3 Metabolic regulation of active ingredients derived from AR and ASR or the corresponding TCM prescriptions
Fig. 2
figure 2

The scheme. DGBX decoction is composed of AR and ASR with a ratio of 5:1. AR and ASR both have anti-inflammatory and anti-cancer effects. Inflammation plays a pivotal role in the pathogenesis and progression of IBD, while anti-cancer effects show significant potential for CRC treatment. Hence, this review aims to comprehensively explore the therapeutic implications of DGBX decoction in IBD-associated CRC. DGBX Danggui Buxue, AR Astragali Radix, ASR Angelicae Sinensis Radix, IBD inflammatory bowel diseases, CRC colorectal cancer

AR and ASR exhibit promising anti-inflammatory properties

By subcutaneous injection of air and Zymosan solution into the back of mice, a Zymosan air–pouch mouse model was established to induce inflammation. The higher dose of aqueous AR extract (100 mg/kg) effectively inhibited the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α, indicating its anti-inflammatory effect through suppression of pro-inflammatory cytokines production. In addition, in lipopolysaccharide (LPS)-induced inflammation of RAW 264.7 cells, AR was found to inhibit the synthesis of inflammatory mediator nitric oxide (NO) and the expression of nitrite oxide synthase (iNOS) [111]. Astragalus polysaccharides and astragaloside IV are the primary bioactive compounds extracted from AR. Astragaloside IV enhances the tyrosine phosphatase activity of CD45 protein to induce T-cell activation, manages the balance of Teff/Treg cells to regulate immunity, and inhibits pro-inflammatory cytokines and nuclear factor-κB (NF-κB) pathways to enhance anti-inflammatory activity [112, 113]. In an orthotopic implantation lung cancer model utilizing C57 BL/6 mice, which was established using 3LL-LUS-IDO cells, astragaloside IV, administered at a dosage of 40 mg/kg, has been demonstrated to effectively suppress the expression of indoleamine 2,3-dioxygenase in vivo. It also down-regulates the population of Tregs while concurrently up-regulating the activity of cytotoxic T lymphocytes to enhance the immune response, thereby showcasing anti-cancer activity [114]. By culturing human lung cancer cells and human mononuclear cells in vitro, it was found that astragaloside IV at a dosage of 40 mg/kg, significantly inhibits the M2 macrophage polarization of tumor-associated macrophages (TAMs) through the modulation of AMPK signaling pathway. This finding was corroborated through parallel experiments conducted on primary human macrophages, which further substantiate the immunomodulatory role of astragaloside IV in regulating macrophage function within the tumor microenvironment [115, 116]. Astragalus polysaccharides, administered at a dosage of 3 mg/kg, exert comparable effects on a lung cancer subcutaneous model in vivo, enhancing the anti-cancer efficacy of cisplatin by modulating the activity of inflammation-associated macrophages. The anti-inflammatory effects of astragalus polysaccharides and astragaloside IV on bovine mammary epithelial cells induced by LPS were also studied. Bovine mammary epithelial cells stimulated with LPS were utilized as an in vitro model of inflammation to investigate the impact of astragalus polysaccharides (an efficacious concentration is 100 μg/mL) and astragaloside IV (an efficacious concentration is 1 mg/mL) on inflamed bovine mammary epithelial cells. It was found that both could significantly reduce the relative expression of IL-6, IL-8, and TNF-α, and activate the Wnt/β-catenin signaling pathway to inhibit inflammation [117]. Atragaloside IV also exerts inhibitory effects on the TLR4/NF-κB signaling pathway and the activation of autophagy, thereby attenuating cellular inflammation by reducing the release of inflammatory mediators [118]. CT26 cells were orthotopically implanted into BALB/c mice to establish a subcutaneous tumor model. Astragaloside III, administered at a dosage of 50 mg/kg in five bi-daily treatments, significantly activated NK cells in tumor environment, thereby enhancing the cytotoxic capacity of NK cells and leading to a notable inhibition of tumor growth. Further assay via co-culture of NK cells with CT26 cells revealed that astragaloside III up-regulated the expression of NK group 2D, Fas and IFN-γ in NK cells, thereby exerting a pronounced suppressive effect on the proliferation of CT26 colorectal tumor cells [119]. Flavonoids isolated from AR alleviate DSS-induced colitis by enhancing mitophagy levels, inhibiting NLRP3 inflammasome activation, and reducing the production of pro-inflammatory cytokines in colon tissue [120].

Calycosin is the predominant isoflavonoid in AR. Calycosin, administered at a dosage of 4.67 mg/kg, effectively reduces the levels of TNF-α and IL-1 in the serum of rats with heart failure induced by ligation of the left anterior descending artery, indicating that calycosin could alleviate the inflammatory response in rats with heart failure. In vitro cardiomyocyte cultures showed that calycosin exerts anti-inflammatory effects via the PI3K-AKT signaling pathway [121]. In glucocorticoid-induced osteonecrosis of the femoral head in rats, calycosin, administered at a dosage of 10 mg/kg, promotes bone formation, inhibits the TLR4/NF-κB pathway, and significantly regulates inflammation, thus effectively alleviating osteonecrosis of the femoral head. In addition, calycosin also inhibits LPS-activated inflammation in vitro by inhibiting the TLR4/NF-κB pathway [122]. Formononetin, a naturally occurring flavonoid derived from AR, has been reported to have immunomodulatory effects [123]. By pre-treatment of LPS-induced mastitis model mice with formononetin, administered at dosages of 10, 20 and 30 mg/kg, myeloperoxidase activity was reduced along with TNF-α and IL-1β production. In vitro experiments using EpH4-Ev cells from mouse mammary epithelial cells stimulated with LPS showed that formononetin, administered at dosages of 10, 20 and 30 μM, inhibits LPS-induced activation of the NF-κB signaling pathway [124]. Taken together, the active component from AR effectively modulates immune cells and cytokines to alleviate inflammatory symptoms.

ASR is also an herb used to regulate the immune system, and its active ingredient acts as an antioxidant and anti-inflammatory agent. Angelica sinensis polysaccharide, extracted from the roots of ASR, is a β-D-pyranoid polysaccharide. It is also a crucial herbal constituent in various traditional formulations utilized for the management of inflammatory responses [125]. Four polysaccharides extracted from different roots of Angelica sinensis have anti-inflammatory activity on intestinal epithelial system, and their activity varies with the difference of structure [126]. Angelica sinensis polysaccharide, administered at a dosage of 40 mg/kg, significantly reduced the levels of TNF, IF-2 and interferon-γ(IFN-γ) in L1210-bearing mice. In addition, angelica sinensis polysaccharide increased the number of lymphocytes, enhanced the ability of macrophages and natural killer cells, and induced a protective immune response [127]. Angelica sinensis polysaccharide, administered at a dosage of 6 mg/kg, significantly reduces the levels of TNF-α, IFN-γ, IL-2, and IL-6 in concanavalin A-induced mouse hepatitis models [128]. Both astragalus polysaccharides and angelica sinensis polysaccharide increase the levels of IL-2 and TNF-α in H22 tumor-bearing mice. Astragalus polysaccharides, administered at a dosage of 400 mg/kg, enhance the phagocytic function of peritoneal macrophages in H22 tumor-bearing mice, while angelica sinensis polysaccharide, administered at a dosage of 200 mg/kg, enhance the activity of T, B lymphocytes, and NK cells, and improve the proportion of lymphocyte subsets in the peripheral blood of H22 tumor-bearing mice. Both significantly inhibit tumor growth in mice [129, 130]. Ligustilide is a bioactive phthalide derivative isolated from ASR, which significantly improves the infiltration of peripheral immune cells, inhibits Th1 immunity, increases Th2 immunity, and re-establishes Th1/Th2 balance [131, 132]. Treatment of human umbilical vein endothelial cells with ligustilide, administered at dosages of 1, 3, 10 μM, significantly inhibits TNF-α and activates the Nrf2/HO-1 signaling pathway, alleviating vascular inflammation, and protecting the blood vessels [133]. Ferulic acid is a phenolic acid isolated from ASR, which has a variety of biological activities, including regulation of inflammation. Ferulic acid was found to improve hepatic oxidative stress and inflammation by activating AMPK in mouse hepatic fibrosis induced by carbon tetrachloride and LPS-induced macrophage inflammation [134]. At an efficacious concentration of 20 μM, ferulic acid inhibits LPS-induced expression of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-1β, and ROS production in macrophages by blocking NLRP3 inflammasome activation [135]. Furthermore, within the concentration range of 1, 2, 4 mM, ferulic acid dose-dependently down-regulates the expression of LC3-II, Beclin 1 and Atg12-Atg5 complex. This modulation of autophagy contributes to its efficacy as an anti-cancer agent by inhibiting the autophagic flux [136]. Additionally, tributyltin ferulate, a derivate of ferulic acid with an efficacious concentration of 400 nM, has been demonstrated to induce autophagic cell death in HCT-116 colon cancer cells, thereby exhibiting anti-tumor properties [137]. Therefore, ASR also effectively mitigates inflammation and modulates immune responses.

Regarding the aspect of inflammation modulation, DGBX decoction regulates immune responses and improves inflammatory symptoms, as shown in Fig. 3. For T lymphocytes, DGBX decoction induces cytokines released from T cells, such as interleukin (IL), granulocyte–macrophage colony-stimulating factor (GM-CSF), IFN-γ, and TNF-α. Phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 is induced to stimulate T lymphocyte proliferation. For macrophages, DGBX decoction treatment increases phagocytosis [138, 139]. Polysaccharides in DGBX decoction induce IκBα degradation, and activate NF-κB signaling pathways, stimulating the immune response. In macrophages, DGBX decoction exerts a pivotal role in host defense mechanisms by dose-dependent suppression of the expression of pro-inflammatory cytokines IL-1β, IL-6, and tumor necrosis factor at both mRNA and protein levels [140]. DGBX decoction significantly reduces the production of pro-inflammatory cytokines, and effectively improves the inflammatory state and pathological structure of DSS-induced IBD model, promoting inflammation resolution. MDSC inhibits the functional activity of CD8+ T activity and improves intestinal inflammation, and DGBX significantly increases the level of MDSC to change the composition of intestinal mucosal immune cells eventually. At the same time, it boosts the proliferation of intestinal epithelial cells and facilitates swift repair of damage to the intestinal mucosal barrier [141, 142]. DGBX decoction attenuates tubulointerstitial fibrosis in rats with unilateral ureteral obstruction by inhibiting the expression of NOD-like receptor family Pyrin domain 3 (NLRP3) inflammasome and significantly reduces the expression of α-smooth muscle actin (α-SMA) representative protein [143]. In 2,4-dinitrochlorobenzene induced mice atopic dermatitis, DGBX decoction significantly inhibits excessive production of IL-4 and IL-5 by Th2 cells, along with a notable reduction in eosinophil and mast cell infiltration, thereby mitigating inflammation and swelling [144]. The potential impact of DGBX decoction on inflammation and immunity is supported by its anti-inflammatory and immunomodulatory effects, mediated by the AR and ASR constituents. Further experimental validation is required to substantiate the immunometabolism potential.

Fig. 3
figure 3

The anti-inflammatory activities of DGBX decoction. DGBX decoction contains polysaccharide, calycosin, formononetin, astragaloside IV, ferulic acid, and ligustilide. These active ingredients interfere with immune cells and modulate cytokines through various signaling pathways to attenuate inflammation

Metabolism modulation aspect

Abnormal metabolism of cancer has highlighted therapeutic targets in recent years. Glucose and amino acids uptake, nutrition acquisition preference, the metabolic intermediates, even the metabolite-driven gene regulation, have been highlighted to explore the novel treatments or targets [57]. AR and ASR both interfere with cell metabolism and improve blood lipids and blood glucose by regulating abnormal cellular metabolic pathways, including fatty acid metabolism and glucose metabolism. AR extract significantly reduces HFD-induced lipid storage, increases the processes of lipolysis and lipid β-oxidation, and alleviates acquired hyperlipidemia in HFD-fed mice by regulating lipid metabolism [170]. Based on pharmacology network analysis and experimental verification, it was found that AR water extract stimulates fat cells and promotes fatty acid metabolism to maintain fatty acid homeostasis [171]. Astragalus polysaccharides at a dosage of 0.25 g/kg regulate cholesterol homeostasis by reducing plasma total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C) in hypercholesterolemia hamsters [172]. Meanwhile, astragalus polysaccharides (700 mg/kg) regulates blood glucose in insulin resistant C57BL/6 J mice by alleviating ER stress [173]. Astragaloside IV, administered at a dosage of 80 mg/kg, alleviates hepatic injury in type 2 diabetes mellitus rats by modulating the AMPK/mTOR pathway, also attenuating dyslipidemia, oxidative stress, and inflammation [174]. Additionally, astragaloside IV, administered at a dosage of 50 mg/kg, exerts hypoglycemic effects in a rat model of diabetes induced by a high-sugar diet combined with streptozotocin by modulating intestinal microbiota [175]. Calycosin-7-glucoside, administered at a dosage of 0.05 mg per mouse, inhibits glycolysis in the db/db mouse model of diabetes mellitus through the activation of AMPK pathway in an inflammatory environment, reducing the inflammatory response and promoting healing of diabetic wounds [176]. Abnormal metabolism in cancer results in different phenotypic characteristics from normal cells, including cell proliferation, migration, invasion, and angiogenesis [177]. Calycosin and Astragaloside IV both inhibit transforming growth factor-β (TGF-β). Calycosin inhibits colorectal cancer cell growth through the PI3K/AKT pathway, upregulates basic leucine zip-ATF-like transcription factor 2 (BATF2) and downregulates plasminogen activator inhibitor-1(PAI-1), and inhibits TGF-β-induced cell migration and enhances the effect of TGF-β induction on cell apoptosis. The mechanism of regulating autophagy is related to the PI3K/AKT/mTOR signaling pathways. Astragalus polysaccharides reduce the levels of p-AKT and p-mTOR in cells, block PI3K/AKT/mTOR signaling pathways, increase autophagy, and alleviate inflammation, to effectively suppress gastric cancer [178,179,180,181]. Angelica sinensis polysaccharide ameliorates the inflammatory response in PC12 cells induced by LPS, attenuates cellular apoptosis, and mitigates cellular damage by down-regulating COX-1 expression and the activation of PI3K/AKT signaling pathway [182]. In addition, Astragaloside IV regulates AMPK, NF-κB, and signal transducer and activator of transcription (STAT) signaling pathways, inhibits the polarization of M2 macrophages, and reduces the progression and metastasis of liver cancer cells and lung cancer cells [116, 183, 184]. Both Astragaloside IV and ligustilide alleviates experimentally DSS-induced colitis. Astragaloside IV, administered at dosages of 50 and 100 mg/kg, effectively inhibits the polarization of M1 macrophages and ameliorates colitis through modulation of STAT signaling pathway. Astragalus saponins reduces the expression level of glycolytic enzymes to attenuate aerobic glycolysis and inflammation, inhibiting colitis eventually. Ligustilide, administered at dosages of 15, 30 and 60 mg/kg, activates peroxisome proliferator-activated receptor γ (PPARγ) and inhibits NF-κB and AP-1 signaling, controlling the expression of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α to alleviate experimental colitis in mice. [156, 185, 186]. ROS are byproducts of cellular metabolism, and the ROS level of cancer cells is higher than that of non-tumor cells. Formononetin mitigates cisplatin-induced nephrotoxicity in LLC-PK1 porcine kidney epithelial cells by suppressing intracellular ROS accumulation and oxidative stress [187]. Similarly, angelica sinensis polysaccharide also inhibits oxidative stress in vivo and in vitro, decrease superoxide dismutase (SOD) activity, and improve acetaminophen-induced acute liver injury to achieve liver protection. Ferulic acid has antioxidant activity, while tributyltin ferulate stimulates ROS production, leading to autophagy activation, showing an obvious anti-tumor effect in colon cancer cells [137, 187, 188]. Astragalus polysaccharides, administered at a dosage of 200 mg/kg, regulate the intestinal microenvironment, including regulating the composition of the intestinal microbiota and its metabolic function, changing the composition of fecal metabolites, reducing the expression levels of IL-1β and IL-6 in serum, weakening the immunosuppressive activity of MDSC, and inhibiting the growth of melanoma in mice [189]. DGBX decoction induces ROS production in the mitochondria of osteoblasts, thereby activating the AMPK pathway, affecting glycolytic capacity, and improving bioenergy [190]. In addition, the potent cardioprotective effect of DGBX decoction is mediated by the regulation of mitochondrial bioenergetics to improve the health status of H9C2 cardiomyoblasts [191]. In conclusion, DGBX decoction and its principal constituents actively participate in metabolic regulation, modulate immune pathways, exerting a therapeutic effect.

Anti-cancer aspect

AR is a traditional tonic herb widely used in the treatment of various cancers. AR aqueous extracts were applied to different cancer cell lines and were found to inhibit a variety of cancer cell growths [211]. AR and its four major bioactive compounds, including calycosin, formononetin, astragaloside IV, and astragalus polysaccharides, were found to have effects on breast cancer cells. Calycosin, at efficacious concentrations of 200 and 400 μM, impedes the migration and invasion of breast cancer cells by suppressing the epithelial-mesenchymal transition process. Formononetin reduces autophagy by regulating mTOR, promotes apoptosis of paclitaxel-resistant triple-negative breast cancer cells, and overcomes paclitaxel resistance [212]. The combination treatment involving formononetin at efficacious concentrations of 40 and 80 μM, in conjunction with metformin, exerts synergistic inhibition of MCF-7 breast cancer cells proliferation and induces apoptosis. Through MDA-MB-231 breast cancer cells in vitro experiments and orthotopic mouse tumor models for in vivo experiments, astragaloside IV was found to inhibit cell viability and invasion of breast cancer cells. Astragalus polysaccharides, administered at concentrations of 100, 200, 500 and 1000 μM, did activate the macrophage-like RAW 264.7 cells in in vitro models to induce apoptosis, thereby inhibiting the viability of MCF-7 cells [78, 213,214,215,216]. Calycosin and astragaloside IV shows anti-tumor activity against CRC and gastric cancer cells. Calycosin, administered at concentrations of 25, 50 and 100 μM, significantly induces apoptosis in HCT116 cells and inhibits cell proliferation and invasion in a dose-dependent manner. Calycosin exhibits significant cytotoxicity against AGS cells, with an IC50 value of 47.18 ± 1.27 μM, while demonstrating minimal toxicity towards normal cells. Astragaloside IV exhibits a dose-dependent inhibition of proliferation in both SW620 and HCT116 cells, while it had no significant effect on the proliferation of normal colonic fetal human cells. N-methyl-N'-nitro-N-nitrosoguanidine was used to induce gastric precancerous lesions (GPL) in a model. Astragaloside IV, at efficacious concentrations of 50 and 100 mg/kg, has been demonstrated to modulate autophagy and apoptosis, thereby exerting a protective effect on gastric mucosal injury and improving both intestinal metaplasia and dysplasia within precancerous gastric lesions [98, 217,218,219]. Astragalus polysaccharides have been shown to participate in a variety of biological processes, encompassing inflammation, metabolism, and carcinogenesis. Cell experiments have shown that astragalus polysaccharides reduce prostate cancer cell proliferation and lipid metabolism in a dose-dependent manner. Utilizing a tumor xenograft model, astragalus polysaccharides, administered at a dosage of 100 mg/kg, have been shown to exert an inhibitory effect on tumor growth via modulation of the miR-138-5p/SIRT1/SREBP1 signaling pathway [220]. Angelica sinensis polysaccharides obtained from ASR are primarily composed of arabinose, glucose, and galactose. Angelica sinensis polysaccharide, at efficacious concentrations of 25, 50, and 100 mg/kg, significantly inhibits tumor growth in H22 tumor-bearing mice by suppressing the production of hepcidin, thereby reducing intracellular iron concentration [221]. Ferulic acid shows inhibitory effects on both Hela and Caski cervical cancer cell lines. By downregulating the expression of MMP-9, ferulic acid suppresses cell invasion in cervical cancer cells. Moreover, ferulic acid inhibits autophagy by decreasing the levels of related proteins LC3-II, Beclin-1, and Atg12-Atg5 in a dose-dependent manner [136]. Ligustilide and two other phthalides extracted from ASR have cytotoxic and anti-proliferative effects on HT-29 [108]. Ligustilide can alter the immunosuppressive function of cancer-associated fibroblasts. Cellular experiments show that ligustilide significantly inhibits prostate cancer and prostate cancer-associated fibroblasts and induces apoptosis of prostate cancer-associated fibroblasts through the TLR4 pathway [222, 223].

DGBX decoction influences tumor development, including inducing cell apoptosis and inhibiting metastasis, enhancing immune function, improving chemotherapy sensitivity, and reducing bone marrow suppression, as shown in Fig. 4. Myelosuppression is a frequently encountered adverse effect of most chemotherapy drugs. In gemcitabine-induced myelosuppression mice, DGBX decoction enhances the anti-cancer effect of gemcitabine by regulating the expression of stress response protein Hu antigen R (HuR), deoxycytidine kinase (dCK), and nuclear factor erythroid 2-related factor (Nrf2). Meanwhile, it inhibits the proliferation of cancer cells, increases the number of bone marrow nucleated cells and the level of hematopoietic cytokine thrombopoietin to alleviate myelosuppression induced by gemcitabine, and improves hematopoietic function [224]. In addition, the combination of DGBX decoction and gemcitabine enhances anti-cancer activity, represented by the increased level of granulocyte–macrophage colony-stimulating factor (GM-CSF), the enhanced immune ability, increased deoxycytidine kinase (dCK), and decreased P-glycoprote in a murine lewis lung carcinoma model [225]. Polysaccharide-depleted DGBX decoction partially inhibits the cell viability of colorectal adenocarcinoma cells, enhances the proliferation inhibition effect of 5-fluorouracil (5-FU), induces apoptosis, and increases sensitivity to chemotherapy or radiotherapy [105]. In addition, phase II clinical studies have shown that DGBX decoction prevents chemotherapy-induced myelosuppression in breast cancer patients [226]. According to network pharmacological analysis, 28 active compounds of DGBX decoction were predicted to hit 61 common targets. CT26 cells were employed to develop a murine model of metastatic colon cancer in BALB/c mice. In vivo experiments showed that DGBX decoction alleviates the progression of metastatic breast cancer by upregulating the expression of pro-apoptotic proteins Bax, inducing the activation of Caspase-3, and downregulating the expression of anti-apoptotic protein Bcl-2 to induce apoptosis [106]. DGBX decoction induces autophagic death of colorectal cancer cells and inhibits the growth of colorectal adenocarcinoma by regulating the mTOR/P70S6K signaling pathway and upregulating autophagy related protein 7 (Atg7) [227]. DGBX decoction, particularly its polysaccharide-depleted fraction, potentiates the growth inhibitory effects of 5-fluorouracil and radiation treatment, possibly by inducing autophagy [105]. DGBX decoction also regulates intestinal flora, enhances immunity of mice by regulating Lactobacillus and Odoribacter, and reduces cancer-related bacteria such as Helicobacter and Lactococcus, showing anti-tumor activity [228].

Fig. 4
figure 4

The anti-tumor activity of DGBX decoction. DGBX decoction regulates apoptotic proteins to induce apoptosis of breast cancer cells. Modulation of HuR, dCK and Nrf2 proteins alleviates the suppression of gemcitabine and enhances the anti-cancer effect of gemcitabine. Modulation of autophagic pathways has been shown to induce autophagic cell death in colorectal cancer cells

Immunometabolism potential of DGBX decoction in IBD-related CRC

Promoting intestinal mucosal repair

The intestinal mucosal barrier is essential to prevent bacterial invasion and maintaining intestinal homeostasis. Intestinal epithelial cells and the tight junction complex between epithelial cells serve as mechanical barriers. The disruption of the intestinal mucosal barrier may result in bacteria and toxins invading normal colon tissue, causing local inflammation, and promoting its carcinogenic transformation [229]. AR has the effect of reducing intestinal inflammation. AR extract, administered at dosages of 5, 10, 50 and 100 μg/mL, reduces the expression of TNF-α and the activation of NF-κB, alleviates the inflammatory response of intestinal epithelial cells, and inhibits the destruction of the intestinal mucosal barrier and the increase of permeability caused by inflammation [230]. AR decoction reduces the levels of inflammatory factors, improves the intestinal mucosal injury induced by lipopolysaccharides in mice, and promotes tissue repair [94]. In addition, astragalus polysaccharides promote the proliferation of intestinal epithelial cells in vitro in a dose-dependent manner. Astragalus polysaccharides stimulates the ornithine decarboxylase (ODC) gene to synthesize polyamine organisms and promote the proliferation, migration, and differentiation of intestinal epithelial cells [231]. Astragaloside IV, administered at a dosage of 3 mg/kg, has been demonstrated to attenuate intestinal mucosal injury induced by sepsis through the downregulation of the RhoA/NLRP3 inflammasome signaling pathway [232]. When administered at the early stage of an AOM/DSS model, ASR extract was found to reduce DNA damage and exert an antioxidant effect in epithelial tissues [107]. In rats with 2,4-dinitrobenzene sulphonic acid (DNBS)-induced acute UC, the content of glutathione was decreased by angelica sinensis polysaccharide, and the protective effect on the intestinal mucosa may be attributed to oxidative stress [110]. Ferulic acid, administered at a dosage of 1 μM, can reduce the LPS-induced inflammatory response in human intestinal epithelial model Caco-2 cells, inhibit the activation of MAPK p38 and ERK1/2, inhibit the expression of iNOS, and alleviate intestinal inflammation [233]. DGBX decoction was found to repair intestinal mucosal barriers and improve IBD. DGBX decoction inhibits the activity of CD8+ T cells by increasing the number of MDSC immune cells, to improve intestinal inflammation. DGBX decoction treatment not only regulates immunity, but also promotes the repair of intestinal mucosal damage by accelerating the proliferation of intestinal epithelial cells [141, 142]. Therefore, DGBX decoction exhibits the potential to enhance the restoration of intestinal mucosal injury, alleviate local inflammation, and prevent carcinogenicity, as shown in Fig. 5.

Fig. 5
figure 5

Effects of DGBX decoction and its principal constituents on intestinal barrier. DGBX decoction increases MDSC immune cells and inhibits the activity of CD8 +T cells. Ferulic acid inhibits the expression of MAPK p38, ERK1/2 and iNOS. Astragalus polysaccharides stimulates ODC gene synthesis of polyamine organisms, which promotes the proliferation of intestinal epithelial cells and improves inflammatory symptoms

Balancing intestinal microbiota

Intestinal microbes and their metabolites influence not only the immune response but also the occurrence and development of CRC. Traditional Chinese medicines and their natural compounds are typically administered orally, inevitably interacting with the gut microbiota [234, 235]. Studies have demonstrated that astragalus polysaccharides effectively ameliorate colonic mucosal injury, restore immune homeostasis, and modulate the overall composition of the intestinal microbiota in mice with DSS-induced acute colitis. Furthermore, it normalizes the levels of Firmicutes and Bacteroides to their physiological states. In addition, astragalus polysaccharides after honey processing could increase the proportion of dominant bacteria such as Lactobacillus and Bacteroides, and significantly inhibit the upregulation of Firmicutes and Verrucomicrobia, thereby protecting the intestinal mucosa, affecting the diversity of microbiota, and alleviating the symptoms of colitis in mice. Honey-processed astragalus polysaccharides exhibited superior anti-inflammatory efficacy compared to astragalus polysaccharides in mice with colitis [100, 103]. The main components of Astragalus mongholicus Bunge-Curcuma aromatica Salisb. include calycosin, formononetin, and three astragalosides. The treatment effectively suppresses the proliferation of opportunistic pathogenic gut bacteria, such as Shigella, Streptococcus, and Enterococcus, while promoting the growth of beneficial probiotic gut microbiota including Lactobacillus, Roseburia, and Mucispirillum. At the same time, significant growth of colon cancer in tumor-bearing mice is inhibited and the intestinal barrier damage is repaired [236]. Interestingly, using human gut microbiota to mimic the gut environment, 4-vinylguaiacol (2-methoxy-4-vinylphenol), a metabolite of ferulic acid, exhibits stronger anti-cancer effects than ferulic acid on both chemo-resistant HT-29 and chemotherapy-sensitiveHCT116 cells. Therefore, oral ferulic acid provides a potential method for CRC treatment [237]. DGBX was found to partially restore the balance of intestinal microbiota destroyed by antibiotics and improve the abundance of intestinal microbiota by increasing the prevalence of Bacteroides, Alistipes and Ruminiclostridium [238]. Therefore, the utilization of DGBX decoction for gut microbiota modulation not only ameliorates colitis but also exerts inhibitory effects on colon cancer progression, thus exhibiting promising prospects in the management of IBD-associated CRC.

Clinical research

A formulation developed from the DGBX decoction significantly ameliorates postoperative immunosuppression in cancer patients, sustainably bolsters immune function, and possesses anti-tumor properties, thereby promoting postoperative recovery [239]. In individuals sustaining severe abdominal trauma, there is a notable diminishment in cellular immunity. Clinical trials have evidenced that the administration of Astragalus injection as an adjuvant therapy is efficacious in the restoration of cellular immune function [240]. A Phase II clinical trial was conducted involving a cohort of healthy, naturally postmenopausal women. The study intervention involved the administration of escalating doses of oral DGBX decoction for a period of 12 weeks. Throughout the trial, physiological parameters and adverse events were closely monitored, with blood samples analyzed for a spectrum of health indicators. Notably, no significant alterations were observed in serum levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, or high-density lipoprotein cholesterol in either intra-group or inter-group comparative analyses. Further research is warranted to ascertain the potential therapeutic effects of DBT on blood lipid profiles in comparable populations [241]. Clinical studies also have demonstrated the efficacy of Astragalus extract TA-65 in ameliorating conditions associated with metabolic syndrome, including a significant elevation in high-density lipoprotein (HDL) cholesterol levels accompanied by a concurrent reduction in the low-density lipoprotein (LDL) to HDL cholesterol ratio, and a marked decrease in plasma TNF-α level [242, 243]. Some clinical trials of DGBX decoction and its main components are shown in Table 4.

Table 4 Some clinical trial of DGBX decoction and its main components

Conclusion

Immunometabolism, the intricate interplay between immune cell metabolism and immune function, has emerged as a promising field with potential therapeutic utility in various pathophysiological conditions. The anti-inflammatory and anti-cancer properties of AR and ASR within the traditional Chinese prescription DGBX decoction, prefigures its immunometabolism potential utility in the context of inflammation-cancer transformation, particularly in the setting of IBD-related CRC. It is evidenced by promoting intestinal mucosal repair and balancing intestinal microbiota. While the field of immunometabolism has made significant strides, it is important to acknowledge the limitations inherent in current research methodologies, such as the choice of experimental models, the fundamental biological differences between mice and humans, and clinical verification in the future. Further investigation into the therapeutic application of DGBX decoction for colorectal cancer is imperative, with a particular focus on elucidating its underlying mechanisms of immunometabolism modulation. Concurrently, it is crucial to implement stringent quality control measures and to standardize the production process of DGBX decoction to ensure its safety and reliability for clinical use.

References

  1. Mak WY, et al. The epidemiology of inflammatory bowel disease: east meets west. J Gastroenterol Hepatol. 2020;35(3):380–9.

    Article  PubMed  Google Scholar 

  2. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature. 2011;474(7351):307–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kobayashi T, et al. Ulcerative colitis. Nat Rev Dis Prim. 2020;6(1):73.

    Article  Google Scholar 

  4. Torres J, et al. Crohn’s disease. Lancet. 2017;389(10080):1741–55.

    Article  PubMed  Google Scholar 

  5. Park J, Cheon JH. Incidence and prevalence of inflammatory bowel disease across Asia. Yonsei Med J. 2021;62(2):99–108.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Guan Q. A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res. 2019;2019:7247238.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Kåhrström CT, Pariente N, Weiss U. Intestinal microbiota in health and disease. Nature. 2016;535(7610):47–47.

    Article  PubMed  Google Scholar 

  8. Song M, Chan AT, Sun J. Influence of the gut microbiome, diet, and environment on risk of colorectal cancer. Gastroenterology. 2020;158(2):322–40.

    Article  CAS  PubMed  Google Scholar 

  9. Weingarden AR, Vaughn BP. Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease. Gut Microbes. 2017;8(3):238–52.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Lakatos PL. Environmental factors affecting inflammatory bowel disease: have we made progress? Dig Dis. 2009;27(3):215–25.

    Article  PubMed  Google Scholar 

  11. Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol. 2015;12(4):205–17.

    Article  PubMed  Google Scholar 

  12. Godet PG, May GR, Sutherland LR. Meta-analysis of the role of oral contraceptive agents in inflammatory bowel disease. Gut. 1995;37(5):668–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Khalili H, et al. Oral contraceptives, reproductive factors and risk of inflammatory bowel disease. Gut. 2013;62(8):1153–9.

    Article  CAS  PubMed  Google Scholar 

  14. Forbes A. Nutrition and inflammatory bowel disease. Curr Opin Clin Nutr Metab Care. 2020;23(5):350–4.

    Article  PubMed  Google Scholar 

  15. Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009;361(21):2066–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Buccafusca G, et al. Early colorectal cancer: diagnosis, treatment and survivorship care. Crit Rev Oncol Hematol. 2019;136:20–30.

    Article  PubMed  Google Scholar 

  17. Qiu H, Cao S, Xu R. Cancer incidence, mortality, and burden in China: a time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in 2020. Cancer Commun. 2021;41(10):1037–48.

    Article  Google Scholar 

  18. Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Article  PubMed  Google Scholar 

  19. Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014;383(9927):1490–502.

    Article  PubMed  Google Scholar 

  20. Patel SG, et al. The rising tide of early-onset colorectal cancer: a comprehensive review of epidemiology, clinical features, biology, risk factors, prevention, and early detection. Lancet Gastroenterol Hepatol. 2022;7(3):262–74.

    Article  PubMed  Google Scholar 

  21. Mármol I, et al. Colorectal carcinoma: a general overview and future perspectives in colorectal cancer. Int J Mol Sci. 2017;18(1):197.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Fung KY, et al. Colorectal carcinogenesis: a cellular response to sustained risk environment. Int J Mol Sci. 2013;14(7):13525–41.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Wijnands AM, et al. Surveillance and management of colorectal dysplasia and cancer in inflammatory bowel disease: current practice and future perspectives. Eur J Intern Med. 2021;93:35–41.

    Article  CAS  PubMed  Google Scholar 

  24. Keller DS, et al. Colorectal cancer in inflammatory bowel disease: review of the evidence. Tech Coloproctol. 2019;23(1):3–13.

    Article  CAS  PubMed  Google Scholar 

  25. Nagao-Kitamoto H, Kitamoto S, Kamada N. Inflammatory bowel disease and carcinogenesis. Cancer Metastasis Rev. 2022;41(2):301–16.

    Article  CAS  PubMed  Google Scholar 

  26. Michaudel C, Sokol H. The gut microbiota at the service of immunometabolism. Cell Metab. 2020;32(4):514–23.

    Article  CAS  PubMed  Google Scholar 

  27. Michaels M, Madsen KL. Immunometabolism and microbial metabolites at the gut barrier: Lessons for therapeutic intervention in inflammatory bowel disease. Mucosal Immunol. 2023;16(1):72–85.

    Article  CAS  PubMed  Google Scholar 

  28. Leber A, et al. NLRX1 modulates immunometabolic mechanisms controlling the host-gut microbiota interactions during inflammatory bowel disease. Front Immunol. 2018;9:363.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Yuan H, et al. Gut microbiota: a novel and potential target for radioimmunotherapy in colorectal cancer. Front Immunol. 2023;14:1128774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Clarke WT, Feuerstein JD. Colorectal cancer surveillance in inflammatory bowel disease: practice guidelines and recent developments. World J Gastroenterol. 2019;25(30):4148–57.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Nadeem MS, et al. Risk of colorectal cancer in inflammatory bowel diseases. Semin Cancer Biol. 2020;64:51–60.

    Article  PubMed  Google Scholar 

  32. He L, et al. Current advances in for gastrointestinal and other cancers. Front Pharmacol. 2021;12:775084.

    Article  CAS  PubMed  Google Scholar 

  33. He L, et al. The immunomodulatory role of matrix metalloproteinases in colitis-associated cancer. Front Immunol. 2022;13:1093990.

    Article  CAS  PubMed  Google Scholar 

  34. Shah SC, Itzkowitz SH. Colorectal cancer in inflammatory bowel disease: mechanisms and management. Gastroenterology. 2022;162(3):715-730 e3.

    Article  PubMed  Google Scholar 

  35. He L, et al. Current advances in coptidis rhizoma for gastrointestinal and other cancers. Front Pharmacol. 2021;12:775084.

    Article  CAS  PubMed  Google Scholar 

  36. Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221:107753.

    Article  CAS  PubMed  Google Scholar 

  37. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019;79(18):4557–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Biswas SK. Metabolic reprogramming of immune cells in cancer progression. Immunity. 2015;43(3):435–49.

    Article  CAS  PubMed  Google Scholar 

  39. Li MO, et al. Innate immune cells in the tumor microenvironment. Cancer Cell. 2021;39(6):725–9.

    Article  CAS  PubMed  Google Scholar 

  40. Zaiatz Bittencourt V, et al. Dysregulation of metabolic pathways in circulating natural killer cells isolated from inflammatory bowel disease patients. J Crohns Colitis. 2021;15(8):1316–25.

    Article  PubMed  PubMed Central  Google Scholar 

  41. González C, et al. Immunometabolic profile associated with progressive damage of the intestinal mucosa in adults screened for colorectal cancer: association with diet. Int J Mol Sci. 2023;24(22):16451.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Kang J, et al. Tumor-suppressive effect of metformin via the regulation of m2 macrophages and myeloid-derived suppressor cells in the tumor microenvironment of colorectal cancer. Cancers. 2022;14(12):2881.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Funes SC, et al. Implications of macrophage polarization in autoimmunity. Immunology. 2018;154(2):186–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Shapouri-Moghaddam A, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233(9):6425–40.

    Article  CAS  PubMed  Google Scholar 

  45. Mehla K, Singh PK. Metabolic regulation of macrophage polarization in cancer. Trends Cancer. 2019;5(12):822–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhu X, et al. Tumor-associated macrophage-specific CD155 contributes to M2-phenotype transition, immunosuppression, and tumor progression in colorectal cancer. J Immunother Cancer. 2022;10(9): e004219.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Li R, et al. Gut microbiota-stimulated cathepsin K secretion mediates TLR4-dependent M2 macrophage polarization and promotes tumor metastasis in colorectal cancer. Cell Death Differ. 2019;26(11):2447–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. De Cicco P, Ercolano G, Ianaro A. The new era of cancer immunotherapy: targeting myeloid-derived suppressor cells to overcome immune evasion. Front Immunol. 2020;11:1680.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lei X, et al. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020;470:126–33.

    Article  CAS  PubMed  Google Scholar 

  50. Netea-Maier RT, Smit JWA, Netea MG. Metabolic changes in tumor cells and tumor-associated macrophages: a mutual relationship. Cancer Lett. 2018;413:102–9.

    Article  CAS  PubMed  Google Scholar 

  51. Wculek SK, et al. Metabolism of tissue macrophages in homeostasis and pathology. Cell Mol Immunol. 2022;19(3):384–408.

    Article  CAS  PubMed  Google Scholar 

  52. Huang SC, et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat Immunol. 2014;15(9):846–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hossain F, et al. Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies. Cancer Immunol Res. 2015;3(11):1236–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bai Z, et al. Tumor-infiltrating lymphocytes in colorectal cancer: the fundamental indication and application on immunotherapy. Front Immunol. 2021;12:808964.

    Article  CAS  PubMed  Google Scholar 

  55. Tan W, et al. Immunometabolism modulation, a new trick of edible and medicinal plants in cancer treatment. Food Chem. 2021;376:131860.

    Article  PubMed  Google Scholar 

  56. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46.

    Article  CAS  PubMed  Google Scholar 

  57. Counihan JL, Grossman EA, Nomura DK. Cancer metabolism: current understanding and therapies. Chem Rev. 2018;118(14):6893–923.

    Article  CAS  PubMed  Google Scholar 

  58. Zhu L, Zhu X, Wu Y. Effects of glucose metabolism, lipid metabolism, and glutamine metabolism on tumor microenvironment and clinical implications. Biomolecules. 2022;12(4):580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Pålsson-McDermott EM, O’Neill LAJ. Targeting immunometabolism as an anti-inflammatory strategy. Cell Res. 2020;30(4):300–14.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23(1):27–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ganapathy-Kanniappan S, Geschwind JF. Tumor glycolysis as a target for cancer therapy: progress and prospects. Mol Cancer. 2013;12:152.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Steinert EM, Vasan K, Chandel NS. Mitochondrial metabolism regulation of T cell-mediated immunity. Annu Rev Immunol. 2021;39:395–416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lee H, Jeon JH, Kim ES. Mitochondrial dysfunctions in T cells: focus on inflammatory bowel disease. Front Immunol. 2023;14:1219422.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yang W, Yu T, Cong Y. CD4(+) T cell metabolism, gut microbiota, and autoimmune diseases: implication in precision medicine of autoimmune diseases. Precis Clin Med. 2022;5(3):pbac018.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Mantovani A, Sica A, Locati M. Macrophage polarization comes of age. Immunity. 2005;23(4):344–6.

    Article  CAS  PubMed  Google Scholar 

  66. Aristin Revilla S, Kranenburg O, Coffer PJ. Colorectal cancer-infiltrating regulatory T cells: functional heterogeneity, metabolic adaptation, and therapeutic targeting. Front Immunol. 2022;13:903564.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Chou WC, et al. Impact of intracellular innate immune receptors on immunometabolism. Cell Mol Immunol. 2022;19(3):337–51.

    Article  CAS  PubMed  Google Scholar 

  68. Zaiatz Bittencourt V, et al. Targeting immune cell metabolism in the treatment of inflammatory bowel disease. Inflamm Bowel Dis. 2021;27(10):1684–93.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Pan X, et al. Macrophage immunometabolism in inflammatory bowel diseases: from pathogenesis to therapy. Pharmacol Ther. 2022;238:108176.

    Article  CAS  PubMed  Google Scholar 

  70. Saravia J, et al. Signaling networks in immunometabolism. Cell Res. 2020;30(4):328–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25(7):771–84.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Michalek RD, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011;186(6):3299–303.

    Article  CAS  PubMed  Google Scholar 

  73. Wang W, et al. 3,5-diCQA suppresses colorectal cancer cell growth through ROS/AMPK/mTOR mediated mitochondrial dysfunction and ferroptosis. Cell Cycle. 2023;22(18):1951–68.

    Article  CAS  PubMed  Google Scholar 

  74. Liu C, et al. Salicylate induces AMPK and inhibits c-MYC to activate a NRF2/ARE/miR-34a/b/c cascade resulting in suppression of colorectal cancer metastasis. Cell Death Dis. 2023;14(10):707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19(2):121–35.

    Article  CAS  PubMed  Google Scholar 

  76. Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer. 2009;9(8):563–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Tan W, Yang DH, Zhong Z. Editorial: immunometabolic response of natural products in inflammation and cancer. Front Immunol. 2023;14:1335510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Lou Y, et al. Astragali radix and its main bioactive compounds activate the Nrf2-mediated signaling pathway to induce P-glycoprotein and breast cancer resistance protein. J Ethnopharmacol. 2019;228:82–91.

    Article  CAS  PubMed  Google Scholar 

  79. Ye J, et al. Research on the interaction of astragaloside IV and calycosin in Astragalus membranaceus with HMGB1. Chin Med. 2023;18(1):81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Chen Z, et al. Astragali Radix (Huangqi): a promising edible immunomodulatory herbal medicine. J Ethnopharmacol. 2020;258:112895.

    Article  CAS  PubMed  Google Scholar 

  81. Chang X, et al. Advances in chemical composition, extraction techniques, analytical methods, and biological activity of astragali radix. Molecules. 2022;27(3):1058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Li A, et al. Exploration of the main effective constituent and the mechanism in astragali radix in the treatment for doxorubicin-induced nephropathy by integrating metabolomics and molecular docking. J Ethnopharmacol. 2023;305:116074.

    Article  CAS  PubMed  Google Scholar 

  83. He L, et al. Glycyrrhizae Radix et Rhizoma: the popular occurrence of herbal medicine applied in classical prescriptions. Phytother Res. 2023;37(7):3135–60.

    Article  CAS  PubMed  Google Scholar 

  84. Zou W, et al. Anti-inflammatory effect of traditional Chinese medicine preparation penyanling on pelvic inflammatory disease. J Ethnopharmacol. 2021;266:113405.

    Article  CAS  PubMed  Google Scholar 

  85. Chen XP, et al. Phytochemical and pharmacological studies on radix Angelica Sinensis. Chin J Nat Med. 2013;11(6):577–87.

    Article  CAS  PubMed  Google Scholar 

  86. Wei WL, et al. Angelica sinensis in China-A review of botanical profile, ethnopharmacology, phytochemistry and chemical analysis. J Ethnopharmacol. 2016;190:116–41.

    Article  CAS  PubMed  Google Scholar 

  87. Gong G, et al. Danggui Buxue Tang, an ancient Chinese herbal decoction, protects β-amyloid-induced cell death in cultured cortical neurons. BMC Complement Altern Med. 2019;19(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zhu S, et al. UHPLC-TQ-MS coupled with multivariate statistical analysis to characterize nucleosides, nucleobases and amino acids in Angelicae Sinensis radix obtained by different drying methods. Molecules. 2017;22(6):918.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Jin Y, et al. Herb pairs containing Angelicae Sinensis radix (Danggui): a review of bio-active constituents and compatibility effects. J Ethnopharmacol. 2016;181:158–71.

    Article  CAS  PubMed  Google Scholar 

  90. Yang FX, et al. Review of chemical constituents, pharmacological effects and clinical applications of Danggui Buxue Decoction and prediction and analysis of its Q-markers. Zhongguo Zhong Yao Za Zhi. 2021;46(11):2677–85.

    PubMed  Google Scholar 

  91. Chan KI, et al. MYC oncogene: a druggable target for treating cancers with natural products. Aging Dis. 2024;15(2):640–97.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Liu J, et al. Calycosin inhibits intestinal fibrosis on CCD-18Co cells via modulating transforming growth factor-β/Smad signaling pathway. Pharmacology. 2019;104(1–2):81–9.

    Article  CAS  PubMed  Google Scholar 

  93. Wang Q, et al. Calycosin suppresses TGF-β-induced epithelial-to-mesenchymal transition and migration by upregulating BATF2 to target PAI-1 via the Wnt and PI3K/Akt signaling pathways in colorectal cancer cells. J Exp Clin Cancer Res. 2019;38(1):240.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Cui Y, et al. Astragalus membranaceus (Fisch.) Bunge repairs intestinal mucosal injury induced by LPS in mice. BMC Complement Altern Med. 2018;18(1):230.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Lee SY, et al. Astragaloside II promotes intestinal epithelial repair by enhancing L-arginine uptake and activating the mTOR pathway. Sci Rep. 2017;7(1):12302.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Qiao C, et al. Astragaloside II alleviates the symptoms of experimental ulcerative colitis in vitro and in vivo. Am J Transl Res. 2019;11(11):7074–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Zhong Y, et al. Astragaloside IV alleviates ulcerative colitis by regulating the balance of Th17/Treg cells. Phytomedicine. 2022;104:154287.

    Article  CAS  PubMed  Google Scholar 

  98. Wang S, et al. Astragaloside IV inhibits cell proliferation of colorectal cancer cell lines through down-regulation of B7–H3. Biomed Pharmacother. 2018;102:1037–44.

    Article  CAS  PubMed  Google Scholar 

  99. Sun P, et al. Astragaloside IV inhibits human colorectal cancer cell growth. Front Biosci. 2019;24(3):597–606.

    Article  CAS  Google Scholar 

  100. Tang S, et al. Combination of polysaccharides from Astragalus membranaceus and Codonopsis pilosula ameliorated mice colitis and underlying mechanisms. J Ethnopharmacol. 2021;264:113280.

    Article  CAS  PubMed  Google Scholar 

  101. Zhong Y, et al. Astragalus polysaccharide alleviates ulcerative colitis by regulating the balance of Tfh/Treg cells. Int Immunopharmacol. 2022;111:109108.

    Article  CAS  PubMed  Google Scholar 

  102. Lv J, et al. Astragalus polysaccharides protect against dextran sulfate sodium-induced colitis by inhibiting NF-κB activation. Int J Biol Macromol. 2017;98:723–9.

    Article  CAS  PubMed  Google Scholar 

  103. Wu J, et al. Structural differences of polysaccharides from Astragalus before and after honey processing and their effects on colitis mice. Int J Biol Macromol. 2021;182:815–24.

    Article  CAS  PubMed  Google Scholar 

  104. An P, et al. Synergistic antitumor effects of compound-composed optimal formula from Aidi injection on hepatocellular carcinoma and colorectal cancer. Phytomedicine. 2022;103:154231.

    Article  CAS  PubMed  Google Scholar 

  105. Chen ST, et al. The traditional Chinese medicine DangguiBuxue tang sensitizes colorectal cancer cells to chemoradiotherapy. Molecules. 2016;21(12):1677.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Feng SH, et al. Danggui Buxue Decoction in the treatment of metastatic colon cancer: network pharmacology analysis and experimental validation. Drug Des Devel Ther. 2021;15:705–20.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Zhao B, et al. Effect of Angelica sinensis root extract on cancer prevention in different stages of an AOM/DSS mouse model. Int J Mol Sci. 2017;18(8):1750.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Kan WL, et al. Study of the anti-proliferative effects and synergy of phthalides from Angelica sinensis on colon cancer cells. J Ethnopharmacol. 2008;120(1):36–43.

    Article  CAS  PubMed  Google Scholar 

  109. Cheng F, et al. Inhibition of dextran sodium sulfate-induced experimental colitis in mice by Angelica Sinensis polysaccharide. J Med Food. 2020;23(6):584–92.

    Article  CAS  PubMed  Google Scholar 

  110. Wong VK, Yu L, Cho CH. Protective effect of polysaccharides from Angelica sinensis on ulcerative colitis in rats. Inflammopharmacology. 2008;16(4):162–7.

    Article  CAS  PubMed  Google Scholar 

  111. Ryu M, et al. Astragali Radix elicits anti-inflammation via activation of MKP-1, concomitant with attenuation of p38 and Erk. J Ethnopharmacol. 2008;115(2):184–93.

    Article  CAS  PubMed  Google Scholar 

  112. Qi Y, et al. Anti-Inflammatory and Immunostimulatory Activities of Astragalosides. Am J Chin Med. 2017;45(6):1157–67.

    Article  CAS  PubMed  Google Scholar 

  113. Zhong Z, et al. Immunomodulatory potential of natural products from herbal medicines as immune checkpoints inhibitors: Helping to fight against cancer via multiple targets. Med Res Rev. 2022;42(3):1246–79.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Zhang A, et al. Astragaloside IV inhibits progression of lung cancer by mediating immune function of Tregs and CTLs by interfering with IDO. J Cancer Res Clin Oncol. 2014;140(11):1883–90.

    Article  CAS  PubMed  Google Scholar 

  115. Bamodu OA, et al. Astragalus polysaccharides (PG2) enhances the M1 Polarization of macrophages, functional maturation of dendritic cells, and T cell-mediated anticancer immune responses in patients with lung cancer. Nutrients. 2019;11(10):2264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Xu F, et al. Astragaloside IV inhibits lung cancer progression and metastasis by modulating macrophage polarization through AMPK signaling. J Exp Clin Cancer Res. 2018;37(1):207.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Fan J, et al. Astragalus polysaccharides and astragaloside IV alleviate inflammation in bovine mammary epithelial cells by regulating Wnt/β-catenin signaling pathway. PLoS ONE. 2022;17(7): e0271598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ying Y, et al. Induction of autophagy via the TLR4/NF-κB signaling pathway by astragaloside IV contributes to the amelioration of inflammation in RAW264.7 cells. Biomed Pharmacother. 2021;137:111271.

    Article  CAS  PubMed  Google Scholar 

  119. Chen X, et al. Astragaloside III enhances anti-tumor response of NK Cells by elevating NKG2D and IFN-γ. Front Pharmacol. 2019;10:898.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Yu T, et al. Ononin alleviates DSS-induced colitis through inhibiting NLRP3 inflammasome via triggering mitophagy. Immun Inflamm Dis. 2023;11(2): e776.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wang X, et al. Calycosin as a novel PI3K activator reduces inflammation and fibrosis in heart failure through AKT-IKK/STAT3 axis. Front Pharmacol. 2022;13:828061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Zhu D, et al. Calycosin modulates inflammation via suppressing TLR4/NF-κB pathway and promotes bone formation to ameliorate glucocorticoid-induced osteonecrosis of the femoral head in rat. Phytother Res. 2021;35(5):2824–35.

    Article  CAS  PubMed  Google Scholar 

  123. Zhang G, et al. Regulation of drug-metabolizing enzymes and efflux transporters by Astragali radix decoction and its main bioactive compounds: Implication for clinical drug-drug interactions. J Ethnopharmacol. 2016;180:104–13.

    Article  CAS  PubMed  Google Scholar 

  124. Xiang K, et al. Formononetin protects LPS-induced mastitis through suppressing inflammation and enhancing blood-milk barrier integrity via AhR-induced Src inactivation. Front Immunol. 2022;13:814319.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Mo ZZ, et al. Angelica sinensis supercritical fluid CO(2) extract attenuates D-galactose-induced liver and kidney impairment in mice by suppressing oxidative stress and inflammation. J Med Food. 2018;21(9):887–98.

    Article  CAS  PubMed  Google Scholar 

  126. Zou YF, et al. The comparison of preliminary structure and intestinal anti-inflammatory and anti-oxidative activities of polysaccharides from different root parts of Angelica sinensis (Oliv.) Diels. J Ethnopharmacol. 2022;295:115446.

    Article  CAS  PubMed  Google Scholar 

  127. Liu W, et al. Structure characterization and anti-leukemia activity of a novel polysaccharide from Angelica sinensis (Oliv.) Diels. Int J Biol Macromol. 2019;121:161–72.

    Article  CAS  PubMed  Google Scholar 

  128. Wang K, et al. Angelica sinensis polysaccharide attenuates concanavalin A-induced liver injury in mice. Int Immunopharmacol. 2016;31:140–8.

    Article  CAS  PubMed  Google Scholar 

  129. Dong XD, et al. Structural characterization of a water-soluble polysaccharide from Angelica dahurica and its antitumor activity in H22 tumor-bearing mice. Int J Biol Macromol. 2021;193(Pt A):219–27.

    Article  CAS  PubMed  Google Scholar 

  130. Yang B, Xiao B, Sun T. Antitumor and immunomodulatory activity of Astragalus membranaceus polysaccharides in H22 tumor-bearing mice. Int J Biol Macromol. 2013;62:287–90.

    Article  CAS  PubMed  Google Scholar 

  131. Luo S, et al. Ligustilide modulates oxidative stress, apoptosis, and immunity to avoid pathological damages in bleomycin induced pulmonary fibrosis rats via inactivating TLR4/MyD88/NF-KB P65. Ann Transl Med. 2020;8(15):931.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Han L, et al. The neuroprotective effects and probable mechanisms of Ligustilide and its degradative products on intracerebral hemorrhage in mice. Int Immunopharmacol. 2018;63:43–57.

    Article  CAS  PubMed  Google Scholar 

  133. Choi ES, et al. Ligustilide attenuates vascular inflammation and activates Nrf2/HO-1 induction and NO synthesis in HUVECs. Phytomedicine. 2018;38:12–23.

    Article  CAS  PubMed  Google Scholar 

  134. Wu J, et al. Ferulic acid ameliorates hepatic inflammation and fibrotic liver injury by inhibiting PTP1B activity and subsequent promoting AMPK phosphorylation. Front Pharmacol. 2021;12:754976.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Liu Y, et al. Ferulic acid exhibits anti-inflammatory effects by inducing autophagy and blocking NLRP3 inflammasome activation. Mol Cell Toxicol. 2022;18(4):509–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Gao J, et al. The anticancer effects of ferulic acid is associated with induction of cell cycle arrest and autophagy in cervical cancer cells. Cancer Cell Int. 2018;18:102.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Celesia A, et al. ROS-dependent ER stress and autophagy mediate the anti-tumor effects of tributyltin (IV) ferulate in colon cancer cells. Int J Mol Sci. 2020;21(21):8135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Gao QT, et al. A Chinese herbal decoction, Danggui Buxue Tang, prepared from Radix Astragali and Radix Angelicae Sinensis stimulates the immune responses. Planta Med. 2006;72(13):1227–31.

    Article  CAS  PubMed  Google Scholar 

  139. Gao QT, et al. A Chinese herbal decoction, Danggui Buxue Tang, activates extracellular signal-regulated kinase in cultured T-lymphocytes. FEBS Lett. 2007;581(26):5087–93.

    Article  CAS  PubMed  Google Scholar 

  140. Gong AG, et al. Polysaccharide of Danggui Buxue Tang, an ancient Chinese herbal decoction, induces expression of pro-inflammatory cytokines possibly via activation of NFκB signaling in cultured RAW 264.7 cells. Phytother Res. 2017;31(2):274–83.

    Article  CAS  PubMed  Google Scholar 

  141. Li C, et al. Danggui Buxue Decoction ameliorates inflammatory bowel disease by improving inflammation and rebuilding intestinal mucosal barrier. Evid Based Complement Alternat Med. 2021;2021:8853141.

    PubMed  PubMed Central  Google Scholar 

  142. Haile LA, et al. Myeloid-derived suppressor cells in inflammatory bowel disease: a new immunoregulatory pathway. Gastroenterology. 2008;135(3):871–81, 881.e1-5.

    Article  CAS  PubMed  Google Scholar 

  143. Wang L, et al. Danggui Buxue Tang attenuates tubulointerstitial fibrosis via suppressing NLRP3 inflammasome in a rat model of unilateral ureteral obstruction. Biomed Res Int. 2016;2016:9368483.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Fang LW, et al. Danggui buxue tang inhibits 2,4-dinitrochlorobenzene: induced atopic dermatitis in mice. Evid Based Complement Alternat Med. 2015;2015:672891.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Xu Y, et al. Calycosin protects HUVECs from advanced glycation end products-induced macrophage infiltration. J Ethnopharmacol. 2011;137(1):359–70.

    Article  CAS  PubMed  Google Scholar 

  146. Chen Y, et al. Reduction of hyperoxic acute lung injury in mice by formononetin. PLoS ONE. 2021;16(1): e0245050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Yang L, et al. Early astragaloside IV administration attenuates experimental autoimmune encephalomyelitis in mice by suppressing the maturation and function of dendritic cells. Life Sci. 2020;249:117448.

    Article  CAS  PubMed  Google Scholar 

  148. Wang YP, et al. Effect of astragaloside IV on T, B lymphocyte proliferation and peritoneal macrophage function in mice. Acta Pharmacol Sin. 2002;23(3):263–6.

    PubMed  Google Scholar 

  149. Wang D, et al. Application of dendritic cells in tumor immunotherapy and progress in the mechanism of anti-tumor effect of Astragalus polysaccharide (APS) modulating dendritic cells: a review. Biomed Pharmacother. 2022;155:113541.

    Article  CAS  PubMed  Google Scholar 

  150. Shao P, et al. Regulation on maturation and function of dendritic cells by Astragalus mongholicus polysaccharides. Int Immunopharmacol. 2006;6(7):1161–6.

    Article  CAS  PubMed  Google Scholar 

  151. Li C, et al. Astragaloside IV ameliorates pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension by restraining the T follicular helper cell response and expanding T follicular regulatory cell response. Phytomedicine. 2022;102:154171.

    Article  CAS  PubMed  Google Scholar 

  152. Liu F, et al. Astragaloside IV exerts anti-tumor effect on murine colorectal cancer by re-educating tumor-associated macrophage. Arch Immunol Ther Exp. 2020;68(6):33.

    Article  Google Scholar 

  153. Li W, et al. Characterization and anti-tumor bioactivity of astragalus polysaccharides by immunomodulation. Int J Biol Macromol. 2020;145:985–97.

    Article  CAS  PubMed  Google Scholar 

  154. Wang Z, et al. Immunomodulatory effect of APS and PSP is mediated by Ca2(+)-cAMP and TLR4/NF-κB signaling pathway in macrophage. Int J Biol Macromol. 2017;94(Pt A):283–9.

    Article  CAS  PubMed  Google Scholar 

  155. Sun Q, et al. PKM2 is the target of a multi-herb-combined decoction during the inhibition of gastric cancer progression. Front Oncol. 2021;11:767116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Tian L, et al. Astragaloside IV alleviates the experimental DSS-induced colitis by remodeling macrophage polarization through STAT signaling. Front Immunol. 2021;12:740565.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Wei Y, et al. Bu-Shen-Yi-Qi formulae suppress chronic airway inflammation and regulate Th17/Treg imbalance in the murine ovalbumin asthma model. J Ethnopharmacol. 2015;164:368–77.

    Article  PubMed  Google Scholar 

  158. Wang W, Liu QB, Jing W. Astragalus membranaceus improves therapeutic efficacy of asthmatic children by regulating the balance of Treg/Th17 cells. Chin J Nat Med. 2019;17(4):252–63.

    CAS  PubMed  Google Scholar 

  159. Huang C, et al. Immunomodulatory mechanism of Bushen Huoxue Recipe alleviates cyclophosphamide-induced diminished ovarian reserve in mouse model. J Ethnopharmacol. 2017;208:44–56.

    Article  PubMed  Google Scholar 

  160. Liu L, et al. Formononetin ameliorates muscle atrophy by regulating myostatin-mediated PI3K/Akt/FoxO3a pathway and satellite cell function in chronic kidney disease. J Cell Mol Med. 2021;25(3):1493–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Zhang WW, et al. Buyang Huanwu Decoction ameliorates ischemic stroke by modulating multiple targets with multiple components: In vitro evidences. Chin J Nat Med. 2018;16(3):194–202.

    CAS  PubMed  Google Scholar 

  162. Wu L, et al. Multi-material basis and multi-mechanisms of the Dahuang Zhechong pill for regulating Treg/Th1 balance in hepatocellular carcinoma. Phytomedicine. 2022;100:154055.

    Article  PubMed  Google Scholar 

  163. Yang J, Xing Z. Ligustilide counteracts carcinogenesis and hepatocellular carcinoma cell-evoked macrophage M2 polarization by regulating yes-associated protein-mediated interleukin-6 secretion. Exp Biol Med. 2021;246(17):1928–37.

    Article  CAS  Google Scholar 

  164. Cao W, et al. Characterizations and anti-tumor activities of three acidic polysaccharides from Angelica sinensis (Oliv.) Diels. Int J Biol Macromol. 2010;46(1):115–22.

    Article  CAS  PubMed  Google Scholar 

  165. Shen J, et al. Effect of Angelica polysaccharide on mouse myeloid-derived suppressor cells. Front Immunol. 2022;13:989230.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Yang T, et al. Immunomodulatory activity of polysaccharide isolated from Angelica sinensis. Int J Biol Macromol. 2006;39(4–5):179–84.

    Article  CAS  PubMed  Google Scholar 

  167. Zhang T, et al. Structures and anti-melanoma activities of two polysaccharides from Angelica sinensis (Oliv.) Diels. Int J Biol Macromol. 2021;183:972–81.

    Article  CAS  PubMed  Google Scholar 

  168. Kuang X, et al. Ligustilide ameliorates neuroinflammation and brain injury in focal cerebral ischemia/reperfusion rats: involvement of inhibition of TLR4/peroxiredoxin 6 signaling. Free Radic Biol Med. 2014;71:165–75.

    Article  CAS  PubMed  Google Scholar 

  169. Uto T, et al. Anti-inflammatory activity of constituents isolated from aerial part of Angelica acutiloba Kitagawa. Phytother Res. 2015;29(12):1956–63.

    Article  CAS  PubMed  Google Scholar 

  170. Wang L, et al. Mechanism of Astragalus membranaceus alleviating acquired hyperlipidemia induced by high-fat diet through regulating lipid metabolism. Nutrients. 2022;14(5):954.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Han Z, et al. Network-driven targeted analysis reveals that Astragali Radix alleviates doxorubicin-induced cardiotoxicity by maintaining fatty acid homeostasis. J Ethnopharmacol. 2022;287:114967.

    Article  CAS  PubMed  Google Scholar 

  172. Cheng Y, et al. Astragalus polysaccharides lowers plasma cholesterol through mechanisms distinct from statins. PLoS ONE. 2011;6(11): e27437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Mao XQ, et al. Hypoglycemic effect of polysaccharide enriched extract of Astragalus membranaceus in diet induced insulin resistant C57BL/6J mice and its potential mechanism. Phytomedicine. 2009;16(5):416–25.

    Article  CAS  PubMed  Google Scholar 

  174. Zhu Y, et al. Astragaloside IV alleviates liver injury in type 2 diabetes due to promotion of AMPK/mTOR-mediated autophagy. Mol Med Rep. 2021;23(6):437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Gong P, et al. Hypoglycemic effect of astragaloside IV via modulating gut microbiota and regulating AMPK/SIRT1 and PI3K/AKT pathway. J Ethnopharmacol. 2021;281:114558.

    Article  CAS  PubMed  Google Scholar 

  176. Chen J, et al. Analysis of the mechanism underlying diabetic wound healing acceleration by Calycosin-7-glycoside using network pharmacology and molecular docking. Phytomedicine. 2023;114:154773.

    Article  CAS  PubMed  Google Scholar 

  177. You M, et al. Signaling pathways in cancer metabolism: mechanisms and therapeutic targets. Signal Transduct Target Ther. 2023;8(1):196.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Tan Y, et al. Astragalus polysaccharide exerts anti-Parkinson via activating the PI3K/AKT/mTOR pathway to increase cellular autophagy level in vitro. Int J Biol Macromol. 2020;153:349–56.

    Article  CAS  PubMed  Google Scholar 

  179. Meng Q, et al. Astragalus polysaccharides inhibits cell growth and pro-inflammatory response in IL-1β-stimulated fibroblast-like synoviocytes by enhancement of autophagy via PI3K/AKT/mTOR inhibition. Apoptosis. 2017;22(9):1138–46.

    Article  CAS  PubMed  Google Scholar 

  180. Wu J, et al. Astragalus polysaccharide enhanced antitumor effects of Apatinib in gastric cancer AGS cells by inhibiting AKT signalling pathway. Biomed Pharmacother. 2018;100:176–83.

    Article  CAS  PubMed  Google Scholar 

  181. Ren Q, et al. Astragalus polysaccharide alleviates LPS-induced inflammation injury by regulating miR-127 in H9c2 cardiomyoblasts. Int J Immunopathol Pharmacol. 2018;32:2058738418759180.

    PubMed  Google Scholar 

  182. Xie Y, et al. Chinese Angelica polysaccharide (CAP) Alleviates LPS-induced inflammation and apoptosis by down-regulating COX-1 in PC12 cells. Cell Physiol Biochem. 2018;49(4):1380–8.

    Article  CAS  PubMed  Google Scholar 

  183. Min L, Wang H, Qi H. Astragaloside IV inhibits the progression of liver cancer by modulating macrophage polarization through the TLR4/NF-κB/STAT3 signaling pathway. Am J Transl Res. 2022;14(3):1551–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Tan W, et al. Deciphering the metabolic role of AMPK in cancer multi-drug resistance. Semin Cancer Biol. 2019;56:56–71.

    Article  CAS  PubMed  Google Scholar 

  185. Guo H, et al. Astragalus saponins inhibit cell growth, aerobic glycolysis and attenuate the inflammatory response in a DSS-induced colitis model. Int J Mol Med. 2019;43(2):1041–8.

    CAS  PubMed  Google Scholar 

  186. Huang Y, et al. Systems pharmacology approach uncovers Ligustilide attenuates experimental colitis in mice by inhibiting PPARγ-mediated inflammation pathways. Cell Biol Toxicol. 2021;37(1):113–28.

    Article  CAS  PubMed  Google Scholar 

  187. Lee H, et al. Inhibition of intracellular ROS accumulation by formononetin attenuates cisplatin-mediated apoptosis in LLC-PK1 cells. Int J Mol Sci. 2018;19(3):813.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Cao P, et al. Angelica sinensis polysaccharide protects against acetaminophen-induced acute liver injury and cell death by suppressing oxidative stress and hepatic apoptosis in vivo and in vitro. Int J Biol Macromol. 2018;111:1133–9.

    Article  CAS  PubMed  Google Scholar 

  189. Ding G, et al. Immunosuppressive activity is attenuated by Astragalus polysaccharides through remodeling the gut microenvironment in melanoma mice. Cancer Sci. 2021;112(10):4050–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Kwan KKL, Dong TTX, Tsim KWK. Danggui Buxue Tang, a Chinese herbal decoction containing Astragali Radix and Angelicae Sinensis Radix, improves mitochrondial bioenergetics in osteoblast. Phytomedicine. 2021;88:153605.

    Article  CAS  PubMed  Google Scholar 

  191. Kwan KKL, et al. Danggui Buxue Tang, a Chinese herbal decoction containing Astragali Radix and Angelicae Sinensis Radix, modulates mitochondrial bioenergetics in cultured cardiomyoblasts. Front Pharmacol. 2019;10:614.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Hu R, et al. Calycosin inhibited autophagy and oxidative stress in chronic kidney disease skeletal muscle atrophy by regulating AMPK/SKP2/CARM1 signalling pathway. J Cell Mol Med. 2020;24(19):11084–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Xu W, et al. Calycosin-7-O-β-D-glucoside attenuates palmitate-induced lipid accumulation in hepatocytes through AMPK activation. Eur J Pharmacol. 2022;925:174988.

    Article  CAS  PubMed  Google Scholar 

  194. Duan X, et al. Calycosin attenuates triglyceride accumulation and hepatic fibrosis in murine model of non-alcoholic steatohepatitis via activating farnesoid X receptor. Phytomedicine. 2017;25:83–92.

    Article  CAS  PubMed  Google Scholar 

  195. Cheng Y, et al. Plant natural product formononetin protects rat cardiomyocyte H9c2 cells against oxygen glucose deprivation and reoxygenation via inhibiting ROS formation and promoting GSK-3β phosphorylation. Oxid Med Cell Longev. 2016;2016:2060874.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Luo Z, et al. Astragaloside IV ameliorates fat metabolism in the liver of ageing mice through targeting mitochondrial activity. J Cell Mol Med. 2021;25(18):8863–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Zhou B, et al. Astragaloside IV attenuates free fatty acid-induced ER stress and lipid accumulation in hepatocytes via AMPK activation. Acta Pharmacol Sin. 2017;38(7):998–1008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Chen Q, et al. Astragalosides IV protected the renal tubular epithelial cells from free fatty acids-induced injury by reducing oxidative stress and apoptosis. Biomed Pharmacother. 2018;108:679–86.

    Article  CAS  PubMed  Google Scholar 

  199. Dong Z, et al. Astragaloside IV alleviates heart failure via activating PPARα to switch glycolysis to fatty acid β-oxidation. Sci Rep. 2017;7(1):2691.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Liu J, et al. Astragalus polysaccharide stimulates glucose uptake in L6 myotubes through AMPK activation and AS160/TBC1D4 phosphorylation. Acta Pharmacol Sin. 2013;34(1):137–45.

    Article  PubMed  Google Scholar 

  201. Cao Y, et al. Astragalus polysaccharide suppresses doxorubicin-induced cardiotoxicity by regulating the PI3k/Akt and p38MAPK pathways. Oxid Med Cell Longev. 2014;2014:674219.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Luo MJ, et al. Astragalus Polysaccharides alleviate type 2 diabetic rats by reversing the expressions of sweet taste receptors and genes related to glycolipid metabolism in liver. Front Pharmacol. 2022;13:916603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Zhong M, et al. Astragalus mongholicus polysaccharides ameliorate hepatic lipid accumulation and inflammation as well as modulate gut microbiota in NAFLD rats. Food Funct. 2022;13(13):7287–301.

    Article  CAS  PubMed  Google Scholar 

  204. Zhang R, et al. Astragalus polysaccharide improves insulin sensitivity via AMPK activation in 3T3-L1 adipocytes. Molecules. 2018;23(10):2711.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Jiang X, et al. Ligustilide inhibits the proliferation of non-small cell lung cancer via glycolytic metabolism. Toxicol Appl Pharmacol. 2021;410:115336.

    Article  CAS  PubMed  Google Scholar 

  206. Wang K, et al. Chronic administration of Angelica sinensis polysaccharide effectively improves fatty liver and glucose homeostasis in high-fat diet-fed mice. Sci Rep. 2016;6:26229.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Wang K, et al. Protective effects of Angelica sinensis polysaccharide against hyperglycemia and liver injury in multiple low-dose streptozotocin-induced type 2 diabetic BALB/c mice. Food Funct. 2016;7(12):4889–97.

    Article  CAS  PubMed  Google Scholar 

  208. Yao W, et al. The investigation of anti-inflammatory activity of volatile oil of Angelica sinensis by plasma metabolomics approach. Int Immunopharmacol. 2015;29(2):269–77.

    Article  CAS  PubMed  Google Scholar 

  209. Hua Y, et al. Metabonomics study on the hepatoprotective effect of polysaccharides from different preparations of Angelica sinensis. J Ethnopharmacol. 2014;151(3):1090–9.

    Article  CAS  PubMed  Google Scholar 

  210. Qi H, Han Y, Rong J. Potential roles of PI3K/Akt and Nrf2-Keap1 pathways in regulating hormesis of Z-ligustilide in PC12 cells against oxygen and glucose deprivation. Neuropharmacology. 2012;62(4):1659–70.

    Article  CAS  PubMed  Google Scholar 

  211. Lin J, et al. Effects of astragali radix on the growth of different cancer cell lines. World J Gastroenterol. 2003;9(4):670–3.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Li T, et al. Formononetin ameliorates the drug resistance of Taxol resistant triple negative breast cancer by inhibiting autophagy. Am J Transl Res. 2021;13(2):497–514.

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Zhang Z, et al. Calycosin inhibits breast cancer cell migration and invasion by suppressing EMT via BATF/TGF-β1. Aging. 2021;13(12):16009–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Xin M, et al. Formononetin and metformin act synergistically to inhibit growth of MCF-7 breast cancer cells in vitro. Biomed Pharmacother. 2019;109:2084–9.

    Article  CAS  PubMed  Google Scholar 

  215. Jiang K, et al. Astragaloside IV inhibits breast cancer cell invasion by suppressing Vav3 mediated Rac1/MAPK signaling. Int Immunopharmacol. 2017;42:195–202.

    Article  CAS  PubMed  Google Scholar 

  216. Li W, et al. Anti-tumor potential of astragalus polysaccharides on breast cancer cell line mediated by macrophage activation. Mater Sci Eng C Mater Biol Appl. 2019;98:685–95.

    Article  CAS  PubMed  Google Scholar 

  217. Chen J, et al. Calycosin induces apoptosis by the regulation of ERβ/miR-17 signaling pathway in human colorectal cancer cells. Food Funct. 2015;6(9):3091–7.

    Article  CAS  PubMed  Google Scholar 

  218. Zhang Y, et al. Calycosin induces gastric cancer cell apoptosis via the ROS-mediated MAPK/STAT3/NF-κB pathway. Onco Targets Ther. 2021;14:2505–17.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Cai T, et al. The gastric mucosal protective effects of astragaloside IV in mnng-induced GPL rats. Biomed Pharmacother. 2018;104:291–9.

    Article  CAS  PubMed  Google Scholar 

  220. Guo S, et al. Astragalus polysaccharides inhibits tumorigenesis and lipid metabolism through miR-138-5p/SIRT1/SREBP1 pathway in prostate cancer. Front Pharmacol. 2020;11:598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Cheng Y, et al. The effects of polysaccharides from the root of Angelica sinensis on tumor growth and iron metabolism in H22-bearing mice. Food Funct. 2016;7(2):1033–9.

    Article  CAS  PubMed  Google Scholar 

  222. Ma J, et al. Ligustilide inhibits the activation of cancer-associated fibroblasts. Life Sci. 2019;218:58–64.

    Article  CAS  PubMed  Google Scholar 

  223. Ma J, et al. Ligustilide promotes apoptosis of cancer-associated fibroblasts via the TLR4 pathways. Food Chem Toxicol. 2020;135:110991.

    Article  CAS  PubMed  Google Scholar 

  224. Liu Y, et al. Danggui Buxue Decoction enhances the anticancer activity of gemcitabine and alleviates gemcitabine-induced myelosuppression. J Ethnopharmacol. 2021;273:113965.

    Article  CAS  PubMed  Google Scholar 

  225. Sun X, et al. Danggui Buxue Decoction sensitizes the response of non-small-cell lung cancer to gemcitabine via regulating deoxycytidine kinase and P-glycoprotein. Molecules. 2019;24(10):2011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Hong J, et al. Danggui Buxue Decoction, a classical formula of traditional Chinese medicine, fails to prevent myelosuppression in breast cancer patients treated with adjuvant chemotherapy: a prospective study. Integr Cancer Ther. 2017;16(3):406–13.

    Article  PubMed  Google Scholar 

  227. Chen ST, et al. Traditional Chinese medicine Danggui Buxue Tang inhibits colorectal cancer growth through induction of autophagic cell death. Oncotarget. 2017;8(51):88563–74.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Shi XQ, et al. Studies on blood enrichment and anti-tumor effects of combined Danggui Buxue Decoction, Fe and rhEPO based on colon cancer-related anemia model and gut microbiota modulation. Chin J Nat Med. 2021;19(6):422–31.

    CAS  PubMed  Google Scholar 

  229. Ternet C, Kiel C. Signaling pathways in intestinal homeostasis and colorectal cancer: KRAS at centre stage. Cell Commun Signal. 2021;19(1):31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Adesso S, et al. Astragalus membranaceus extract attenuates inflammation and oxidative stress in intestinal epithelial cells via NF-κB activation and Nrf2 response. Int J Mol Sci. 2018;19(3):800.

    Article  PubMed  PubMed Central  Google Scholar 

  231. Zhang CL, et al. Modulation of intestinal epithelial cell proliferation, migration, and differentiation in vitro by Astragalus polysaccharides. PLoS ONE. 2014;9(8): e106674.

    Article  PubMed  PubMed Central  Google Scholar 

  232. Xie S, et al. Astragaloside IV attenuates sepsis-induced intestinal barrier dysfunction via suppressing RhoA/NLRP3 inflammasome signaling. Int Immunopharmacol. 2020;78:106066.

    Article  CAS  PubMed  Google Scholar 

  233. Serreli G, et al. Ferulic acid metabolites attenuate LPS-induced inflammatory response in enterocyte-like cells. Nutrients. 2021;13(9):3152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Yue SJ, et al. Gut microbiota modulation with traditional Chinese medicine: a system biology-driven approach. Pharmacol Res. 2019;148:104453.

    Article  CAS  PubMed  Google Scholar 

  235. Kim J, Lee HK. Potential role of the gut microbiome in colorectal cancer progression. Front Immunol. 2021;12:807648.

    Article  CAS  PubMed  Google Scholar 

  236. Gu J, et al. Standardized Astragalus mongholicus bunge-curcuma Aromatica Salisb. Extract efficiently suppresses colon cancer progression through gut microbiota modification in CT26-bearing mice. Front Pharmacol. 2021;12:714322.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Luo Y, et al. 4-Vinylguaiacol, an active metabolite of ferulic acid by enteric microbiota and probiotics, possesses significant activities against drug-resistant human colorectal cancer cells. ACS Omega. 2021;6(7):4551–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Du R, et al. Danggui Buxue Tang restores antibiotic-induced metabolic disorders by remodeling the gut microbiota. J Ethnopharmacol. 2020;259:112953.

    Article  CAS  PubMed  Google Scholar 

  239. Du QC, Yang KZ, Sun XF. Efficacy of auxiliary therapy with Danggui Buxue Decoction No.1 in treating patients of non-small cell lung cancer at peri-operational stage. Chin J Integr Med. 2009;15(3):184–8.

    Article  PubMed  Google Scholar 

  240. Wu J, et al. Effect of astragalus injection on serious abdominal traumatic patients’ cellular immunity. Chin J Integr Med. 2006;12(1):29–31.

    Article  CAS  PubMed  Google Scholar 

  241. Wang CC, et al. A randomized, double-blind, multiple-dose escalation study of a Chinese herbal medicine preparation (Dang Gui Buxue Tang) for moderate to severe menopausal symptoms and quality of life in postmenopausal women. Menopause. 2013;20(2):223–31.

    Article  PubMed  Google Scholar 

  242. Fernandez ML, et al. TA-65, a telomerase activator improves cardiovascular markers in patients with metabolic syndrome. Curr Pharm Des. 2018;24(17):1905–11.

    Article  CAS  PubMed  Google Scholar 

  243. Li NY, et al. Astragalus membranaceus improving asymptomatic left ventricular diastolic dysfunction in postmenopausal hypertensive women with metabolic syndrome: a prospective, open-labeled. Randomized Controlled Trial Chin Med J (Engl). 2018;131(5):516–26.

    PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank all our colleagues and friends for their sincere and generous help.

Funding

This research was funded by the Gansu Province Science Fund for Distinguished Young Scholars (20JR10RA650), and the Outstanding Youth of the Fundamental Research Funds for the Central Universities (lzujbky-2021-ey21). This study was also supported by the Macao Science and Technology Development Fund (FDCT 001/2023/ALC, 0123/2022/A and 0006/2020/AKP), Natural Science Foundation of Guangdong Province, China (2020A1515010922), Traditional Chinese Medicine Bureau of Guangdong Province, China (20201183), Shenzhen–Hong Kong-Macau S&T Program (Category C) (SGDX2020110309420200), and Research Fund of the University of Macau (CPG2023-00028-ICMS).

Author information

Authors and Affiliations

Authors

Contributions

YZ wrote and revised this manuscript. QK, LH, KC, HG and WX participated in the revision guidance of the manuscript. WT and ZZ conceived and organized this study. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Zhangfeng Zhong or Wen Tan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Y., Kang, Q., He, L. et al. Exploring the immunometabolic potential of Danggui Buxue Decoction for the treatment of IBD-related colorectal cancer. Chin Med 19, 117 (2024). https://doi.org/10.1186/s13020-024-00978-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13020-024-00978-y

Keywords