Open Access

Research progress on the molecular mechanisms of Saikosaponin D in various diseases (Review)

  • Authors:
    • Simin Gu
    • Yiyuan Zheng
    • Chong Chen
    • Jing Liu
    • Yanping Wang
    • Junmin Wang
    • Yong Li
  • View Affiliations

  • Published online on: December 24, 2024     https://doi.org/10.3892/ijmm.2024.5478
  • Article Number: 37
  • Copyright: © Gu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bupleurum, a Traditional Chinese Medicine (TCM) herb, is widely used in China and other Asian countries to manage chronic liver inflammation and viral hepatitis. Saikosaponin D (SSD), a triterpenoid saponin extracted from Bupleurum, exhibits extensive pharmacological properties, including anti‑inflammatory, antioxidant, anti‑apoptotic, anti‑fibrotic and anti‑cancer effects, making it a therapeutic candidate for numerous diseases. Clarifying the targets and molecular mechanisms underlying TCM compounds is essential for scientifically validating TCM's therapeutic roles in disease prevention and treatment, as well as for identifying novel therapeutic targets and lead compounds. This analysis comprehensively examines SSD's mechanisms across various conditions, such as myocardial injury, pulmonary diseases, hepatic disorders, renal pathologies, neurological disorders, diabetes and cancer. In addition, challenges and potential solutions encountered in SSD research are addressed. SSD is posited as a promising monomer for multifaceted therapeutic applications and this article aims to enhance researchers' understanding of the current landscape of SSD studies, offering strategic insights to guide future investigations. 

Introduction

As awareness of health and wellness grows, the benefits of Traditional Chinese Medicine (TCM) in health care and disease treatment are increasingly recognized. TCM compounds, valued for their targeted therapeutic effects and minimal adverse reactions (1-3), are gaining market traction. Guided by TCM principles, researchers employ modern scientific and technological methods to investigate the pharmacodynamics, pharmacology, toxicology and clinical applications of TCM compounds. Extensive studies have elucidated the active ingredients and mechanisms of these compounds (2), leading to significant advancements. For instance, Qinggan Huoxue Recipe has been shown to mitigate alcoholic liver disease (ALD) progression by inhibiting the liver X receptor-lysophosphatidylcholine acyl-transferase 3 signaling pathway (4). In addition, Danggui Buxue Decoction may improve diabetic nephropathy by modulating insulin resistance, chronic inflammation, and lipid accumulation (5). In Jiangzhi Granule, Xiang et al (6) identified that kaempferol, a monomer from lotus leaf, alleviates liver damage in non-alcoholic steatohepatitis mice by reducing endoplasmic reticulum stress. Ginsenosides from ginseng have been demonstrated to exhibit anti-inflammatory, antioxidant and anti-tumor activities (7,8), alongside neuroprotective effects through the protein kinase B (AKT)/cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) pathway (9). Such findings support the development of innovative therapeutics.

Bupleurum, a widely used TCM herb in China and other Asian countries for managing chronic liver inflammation and viral hepatitis (10,11), is a key component in classic TCM formulas such as Xiao-Chai-Hu-Tang (12), Chaihu-Shugan-San (13) and Xiaoyaosan (14). Known for its anti-inflammatory, anti-infective and hepatoprotective effects (15-18), Bupleurum has yielded >100 triterpene saponins, including saikosaponin (SS)A, B, C and D, with SSA and SSD as primary bioactive components (11). Studies have demonstrated SSD's diverse pharmacological effects, including anti-inflammatory, antioxidant, anti-apoptotic, anti-fibrotic and anti-cancer properties (19), underscoring its therapeutic potential across a spectrum of conditions such as myocardial injury (20), lung injury (21), non-alcoholic fatty liver disease (NAFLD) (22), liver fibrosis (23), glomerulonephritis (24), diabetes (25), depression (26), Alzheimer's disease (AD) (27) and various cancers (28,29).

Elucidating the action targets and molecular mechanisms of TCM compounds is instrumental in scientifically validating their roles in disease prevention and treatment, as well as in identifying novel therapeutic targets and lead compounds. Our team has dedicated extensive research to understanding SSD's mechanisms in liver fibrosis (30-33), while valuable studies by other researchers on SSD in additional diseases also deserve close examination. This review synthesizes the mechanisms of SSD across a range of diseases from its discovery to the present, highlighting current challenges and proposing strategic solutions in SSD research. SSD emerges as a promising monomer with multifaceted therapeutic potential. This article will enhance researchers' understanding of SSD's current research status and offer strategic guidance for future studies.

Chemical structure of SSD

SSD, a triterpene saponin compound, dissolves well in methanol and ethanol but has limited solubility in water. Structurally, it resembles steroids (Fig. 1) (34,35) and possesses a chemical formula of C42H68O13 with a molecular weight of 780.98 (35). The extracted SSD is a white powder, which will be considered for oral administration in the future. The main administration methods of SSD in existing animal experimental studies are gastric lavage and intraperitoneal injection (24,25).

Pharmacological effects of SSD

SSD possesses diverse therapeutic properties, including anti-inflammatory, antipyretic, analgesic, antioxidant, anti-apoptotic, antiviral, antifibrosis, immune-regulation and anti-tumor activities (19), with specific mechanisms supported by studies (Fig. 2).

Investigations into SSD's anti-inflammatory mechanisms reveal its potent inhibition of lipopolysaccharide (LPS)-induced inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and pro-inflammatory cytokines (TNF-α and IL-6) in RAW264.7 cells by blocking nuclear factor-κB (NF-κB) activation (36). Furthermore, SSD was reported to significantly reduce cerulein-induced apoptosis and inflammation in pancreatic AR42J cells by modulating the mitogen-activated protein kinase (MAPK) signaling pathway (37). Heat stress, a trigger for reactive oxygen species (ROS) production, leads to oxidative tissue damage (38). Zhang et al (39) reported that SSD mitigated heat stress-induced oxidative damage in LLC-PK1 cells by promoting antioxidant enzymes and heat shock protein 72. The imbalance between cellular defense mechanisms and free radical production is known as oxidative stress (OS). Research by Lin et al (40) demonstrated that SSD significantly reduces H2O2-induced malondialdehyde (MDA) and lactate dehydrogenase (LDH) release, while enhancing superoxide dismutase (SOD) activity and antioxidant capacity, thereby protecting against PC12-cell apoptosis. The primary mechanism involves SSD's ability to eliminate ROS and disrupt MAPK-mediated oxidative injury, which prevents H2O2-induced PC12-cell death (40). An additional study showed SSD's ability to reduce OS through the phosphatidylinositol-3 kinase (PI3K)/AKT/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, improving muscle atrophy in chronic kidney disease (CKD) models (41).

SSD has shown antiviral activity against enterovirus A71 (EV-A71), which causes hand, foot and mouth disease, a virus known to trigger autophagy (42). Li et al (43) found that SSD inhibits EV-A71 infection by effectively suppressing viral RNA replication and subsequent viral protein synthesis, preventing EV-A71-induced cell death.

In anti-fibrosis research, Sun et al (44) demonstrated that SSD inhibits human embryonic lung fibroblast proliferation and collagen production by modulating the transforming growth factor β (TGF-β1)/Smads signaling pathway, exerting a significant antifibrotic effect.

SSD has also shown immunomodulatory benefits, as it regulates Type 1 T-helper cell (Th1)/Th2 and Th17/T-regulatory cell (Treg) imbalances and alleviates Hashimoto's thyroiditis (HT) severity in mice by promoting M2 macrophage differentiation (45).

Among saikosaponins, SSD displays the strongest anti-tumor activity, targeting various cancers through multiple pathways (46-48). For instance, SSD inhibits non-small cell lung cancer cell proliferation and induces apoptosis by disrupting the signal transducer and activator of transcription 3 (STAT3) pathway (48) and also suppresses triple-negative breast cancer cell growth through β-catenin signaling inhibition (49).

These pharmacological insights underscore SSD's promising potential as a therapeutic agent.

Research progress on SSD in various diseases

SSD demonstrates a wide range of therapeutic properties, including anti-inflammatory, antipyretic, analgesic, antioxidant, anti-apoptotic, antiviral, antifibrotic, immune-regulation and anti-tumor activities. Consequently, its pharmacological effects of inhibiting inflammation, oxidative stress, fibrosis and so on are related to the treatment of various diseases, such as myocardial injury (20), lung damage (50), NAFLD (22), liver fibrosis (23), glomerulonephritis (51), diabetes (25), depression (26), AD (27) and cancers (52,53) (Fig. 3). Specific molecular mechanisms are detailed in Tables I and II.

Table I

SSD inhibits the molecular mechanisms underlying the development of several diseases.

Table I

SSD inhibits the molecular mechanisms underlying the development of several diseases.

DiseaseMechanism(Refs.)
Myocardial damageSSD → p38MAPK↓ → CAT, GPx↑, MDA, ROS↓ → OS↓(20)
Lung damageSSD → neutrophils, MPO, MIP, IL-6, TNF-α↓, TGF-β1, IL-10↑ → inflammation↓; SSD → caspase-3, bax↓, bcl-2↑ → apoptosis↓(21,55)
Pulmonary fibrosisSSD → TGF-β1/Smads↓ → Col-1, α-SMA↓ → fibrosis↓; SSD → angiopoietin/Tie2 pathway↓ → ANG-1, ANG-2, Tie2↓ → Angiogenesis↓(44,57)
Acute liver injurySSD → NF-κB, STAT3↓ → IL-6, CCL2↓, IL-10↑ → inflammation↓; SSD → NLRP3 inflammasome, OS↓ → NLRP3, ASC, IL-1β, IL-18↓; SOD, GPx, CAT↑, MDA, MSP↓ → inflammation and OS↓(59-61)
Alcoholic liver diseaseSSD → ALT, AST, TG, TC, TNF-α, MDA↓ GPx, T-SOD↑ → inflammation and OS↓(63-65)
NAFLDSSD → INSIG/SREBP1c, Fas, Acaca↓ → fatty acid synthesis↓; SSD → PPARα, COX1, CPT1α↑ → fatty acid oxidation↑(22,67, 68)
Liver fibrosisSSD → GPER1/autophagy → Col-1, α-SMA↓ → fibrosis↓; SSD → ERβ↑ → ROS/NLRP3 pathway↓ → inflammation↓(32,73)
Functional dyspepsiaSSD → ghrelin, SP↑, caspases-3, bax↓, bcl-2↑ → autophagy and apoptosis↓(74)
Ulcerative colitisSSD → NF-κB↓ → IL-6, TNF-α, IL-1β↓, IL-10↑ → inflammation↓(75)
Chronic pancreatitisSSD → PI3K/AKT/mTOR↑ → autophagy↓ → fibrosis↓; SSD → MAPK → inflammation and apoptosis↓(37,77)
NephritisSSD → TGF-β1, macrophages, CD8+T↓ → inflammation↓; SSD → TCF7/FOSL1/MMP-9 → inflammation and apoptosis↓(24,51)
Chronic kidney diseaseSSD → PI3K/AKT/Nrf2↑ → ROS↓(41)
NephrotoxicitySSD → MAPK, NF-κB↓ → TNF-α, IL-1β, IL-6, NO, iNOS↓ → inflammation↓(82)
Renal failureSSD → TGF-β1/BMP7/Gremlinl/Smad↓ → fibrosis↓(84)
Alzheimer's diseaseSSD → NF-κB↓ → amyloid β, astrocytes, hippocampal microglia↓; SSD → Nrf2 pathway↑ → OS↓(27,87)
DepressionSSD → p-CREB, BDNF↑ → neuron generation↑; SSD → NF-κB, miR-155↓, FGF-2↑, Homer1-mGluR5 and mTOR pathways↑ → depressive behavior↓; SSD → MGB1/TLR4/NF-κB↓ → inflammation, microglia↓ → depressive behavior↓(91-94)
Diabetes complicationsSSD → SIRT3↑ → OS↓ → DN↓; SSD → AQP1/RhoA/ROCK↓ → inflammation (IL-6, TNF-α, IL-1β)↓, NVC↑ → DPN↓(25,79)
ThyroiditisSSD → TPOAb, IFN-γ(Th1), IL-17(Th17)↓ M2 macrophage polarization↑ → HT↓(45)
OsteoarthritisSSD → PI3K/AKT/mTOR↓ → inflammation and autophagy↓; SSD → Nrf2/HO-1/ROS↑ → inflammation and ECM damage↓; SSD → microRNA-199-3p↑ → TCF4↑ → inflammation↓(98-100)

[i] ADRB2, β2-adrenergic receptors; ANG, angiotensin; ASC, adapter protein apoptosis associated speck-like protein containing a CARD; ALT, alanine aminotransferase; AKT, protein kinase B; AST, aspartate aminotransferase; BDNF, brain-derived neurotrophic factor; CAT, catalase; COX-2, Cyclooxygenase-2; C/EBPβ, CCAAT enhancer binding protein beta; CPT1α, carnitine palmitoyltransferase-1 alpha; CREB, cyclic adenosine monophosphate (cAMP) response element-binding protein; ERβ, estrogen receptor β; FOSL1, Fos-like antigen 1; FTO, fat mass and obesity-associated protein; GPx, glutathione peroxidase; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; LDH, lactate dehydrogenase; MAPK, mitogen-activated protein kinases; MDA, malondialdehyde; MPO, myeloperoxidase; MMP-9, matrix metalloproteinase-9; m6A, mRNA N6-methyladenosine; MSP, MDA and superoxide production; NCV, nerve conduction velocity; NF-κB, nuclear factor-κB; NLRP3, NLR family pyrin domain containing 3; Nrf2, nuclear factor erythroid 2-related factor 2; OS, oxidative stress; PI3K, phosphatidylinositol-3 kinase; ROS, reactive oxygen species; SOD, superoxide dismutase; SP, substance P; SSD, Saikosaponin D; STAT3, signal transducer and activator of transcription 3; TCF4, transcription factor-4; TC, total cholesterol; TG, triglycerides; TGF-β1, transforming growth factor β; TLR4, Toll-like receptor 4;TNF-α, tumor necrosis factor-α; TPOAb, thyroid peroxidase antibody; T-SOD, total superoxide dismutase.

Table II

SSD inhibits the molecular mechanisms underlying the development of cancers.

Table II

SSD inhibits the molecular mechanisms underlying the development of cancers.

DiseaseMechanism(Refs.)
Thyroid cancerSSD → P53, bax↑, bcl-2↓ → apoptosis↑; SSD → P21↑, CDK2, cyclin D1↓ → cell cycle inhibition↑(101)
Lung cancerSSD → TGFα-JNK-p53, TGFα-Rassfia-Mst1↑; NF-κB/NLRP3/caspase-1/GSDMD↑; STAT3/Bcl-2 pathway↓ → apoptosis↑; SSD → TGFβ-p53/p21/p27/p15/p16, TGFα-Rassfia-cyclin D1↑ → cell proliferation↓(46,50, 103)
Breast cancerSSD → β-catenin↓ → apoptosis↑; SSD → P-gp↓ → drug resistance↓(49,105)
Liver cancerSSD → p-STAT3/HIF-1α, p-STAT3/C/EBPβ↓ → COX-2↓ → proliferation↓, apoptosis↑; SSD → P53, bax↑, bcl-2, HIF-1α↓ → radiosensitivity and apoptosis↑; SSD → SENP5↑ → Glil SUMO↓ → chemosensitivity↑(111-116)
Intrahepatic cholangiocarcinomaSSD → ADRB2/Glycolysis pathway → drug resistance↓(130,131)
Gastric cancerSSD → IKKβ/NF-κB pathway↓ → apoptosis and autophagy↓; SSD → caspase-3, bcl-2, IL-2↑, MMP1, MMP2 ↓ → apoptosis↑, migration and invasion↓(28,133)
Pancreatic cancerSSD → MKK4/JNK pathway↑ → proliferation↓; SSD → PI3K/AKT/mTOR↓ → M2 polarization↓ → immunosuppression↓(47,135)
Kidney cancerSSD → EGFR/p38↓ → p53↑ → cell cycle arrest and apoptosis↑(136)
Prostate cancerSSD → GSK3β↓ → Wnt/β-catenin↓ → EMT, CSC↓; SSD → p53, p21 ↑ → cell cycle arrest↑; SSD → caspaes3 ↑ → apoptosis↑(138,139)
Ovarian cancerSSD → Drp1, Opa1 ↑, MMP↓ → mitochondrial division and cell cycle arrest↑ → apoptosis↑(141)
Endometrial cancerSSD → p21, cyclin B↑ → G2/M cycle arrest↑; SSD → ROS, bcl-2↑, mitochondrial membrane potential↓ → apoptosis↑; SSD → Ras/Raf/MEK/ERK, JNK and p38 MAPK pathway → tumor metastasis↓(142)
OsteosarcomaSSD → p53, p21, p27↑, cyclin D1↓ → proliferation↓; SSD → cytochrome C, bax/bcl-2 ratio, caspase-3, caspase-8, caspase-9↑ → apoptosis↑(144,145)
GlioblastomaSSD → JNK, caspase-3↑, AKT, ERK↓ → apoptosis↑(146)
MedulloblastomaSSD → Hedgehog pathway↓ → tumor growth↓(151)
Acute myeloid leukemiaSSD → FTO↓ → m6A RNA methylation↑ → tumor drug resistance↓(155)

[i] ADRB2, β2-adrenergic receptors; AKT, protein kinase B; CDK2, cyclin-dependent kinase 2; COX-2, cyclooxygenase-2; CSC, cancer stem cell; Drp1, dynamin-related protein 1; EGFR, epidermal growth factor receptor; EMT, epithelial-to-mesenchymal transition; FTO, fat mass and obesity-associated protein; GSK3β, glycogen synthase kinase 3β; GSDMD, gasdermin D; HIF-1α, hypoxia-inducible factor-1α; IL-6, interleukin-6; MAPK, mitogen-activated protein kinases; MMP, mitochondrial membrane potential; m6A, mRNA N6-methyladenosine; NF-κB, nuclear factor-κB; NLRP3, NLR family pyrin domain containing 3; Nrf2, nuclear factor erythroid 2-related factor 2; Opa1, optic atrophy 1; PI3K, phosphatidylinositol-3 kinase; P-gp, P-glycoprotein; ROS, reactive oxygen species; SENP5, sentrin-specific proteases; SSD, Saikosaponin D; STAT3, signal transducer and activator of transcription 3; TGF-β1, transforming growth factor β; UCMS, unpredictable chronic mild stress.

Myocardial damage

The oxidative damage induced by anticancer drugs such as doxorubicin (DOX) can result in irreversible cardiotoxicity (54). Zhang et al (20) found that DOX administration led to cardiac damage and dysfunction, as well as reduced survival in mice. In H9c2 cells, DOX treatment increased LDH leakage, cardiomyocyte apoptosis, myocardial fibrosis and reduced cardiomyocyte volume. SSD protects cardiomyocytes from DOX-induced cardiotoxicity by inhibiting excessive OS through the p38-MAPK signaling pathway (20).

Lung-related diseases
Lung damage

In a rat model of ventilator-induced lung injury (VILI), SSD reduced pulmonary neutrophil infiltration, myeloperoxidase levels and pro-inflammatory cytokines (MIP-2, IL-6 and TNF-α), while elevating anti-inflammatory mediators (TGF-β1 and IL-10) (21). SSD also reduced OS and apoptosis in lung tissue by downregulating caspases-3 and pro-apoptotic protein Bax, while upregulating the anti-apoptotic protein Bcl-2. These effects suggest that SSD may alleviate VILI by suppressing inflammation, OS and apoptosis (21). Another study demonstrated that SSD reduced LPS-induced inflammation and apoptosis in MLE-12 lung epithelial cells and, in vivo, decreased pathological damage, inflammation and apoptosis in cecum ligation and perforation-induced mouse septic acute lung injury model (55).

Pulmonary fibrosis

Idiopathic pulmonary fibrosis, a group of heterogeneous parenchymal lung disorders characterized by chronic progressive fibrosis, represents one of the most severe interstitial lung diseases (56). Sun et al (44) reported that SSD inhibits human embryonic lung fibroblast proliferation and collagen production [Collagen-1 (Col-1), α-smooth muscle actin (α-SMA)] via modulation of the TGF-β1/Smads pathway, exerting an antifibrotic effect. A recent study revealed that SSD reduces the expression of angiotensin-1 (Ang-1), Ang-2 and Tie-2 (tyrosine kinase receptors with immunoglobulin and epidermal growth factor homology domains-2) in the angiopoietin/Tie2 pathway, which may help alleviate pulmonary inflammation in bleomycin-induced mice (administered via intratracheal injection) by inhibiting angiogenesis and fibrosis (57).

Liver diseases
Acute liver injury

Excessive acetaminophen (APAP) intake can lead to acute liver injury, which can be fatal and necessitates effective pharmacological intervention (58). Liu et al (59) found that SSD protected mice from APAP-induced hepatotoxicity by primarily inhibiting NF-κB and STAT3-mediated inflammatory signaling pathways (IL-6, C-C motif chemokine ligand 2, IL-10). Another study indicated that SSD mitigated carbon tetrachloride-induced acute hepatocyte damage by suppressing OS and NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in HL-7702 cells (60). Similar protective effects were observed in animal models (61), where SSD administration led to reduced MDA and superoxide production in carbon tetrachloride-treated mouse liver tissue while enhancing SOD, glutathione peroxidase (GPx) and catalase activities. In addition, protein levels of caspase 1, NLRP3, apoptosis-associated speck-like protein, IL-1β and IL-18 were diminished (61), supporting conclusions drawn from in vitro experiments.

ALD

ALD, encompassing a range of liver damage due to chronic excessive alcohol intake, has become a significant global health concern (62). Hepatic stellate cells (HSCs) play a pivotal role in ALD progression, with activated HSCs acting as a primary source of extracellular matrix (ECM) in the liver (63). Jiang et al (64) demonstrated that SSD suppressed proliferation and promoted apoptosis in acetaldehyde-activated rat HSC-T6 cells by stimulating autophagosome formation. SSD treatment increased caspase-3 and Bax expression while reducing Ki67 and Bcl-2 levels. Despite its therapeutic potential, SSD has poor bioavailability, stability and solubility, which limits its clinical application. A promising study aimed to enhance SSD's efficacy by formulating SSD-loaded liposomes using a thin-film hydration method (65). Compared to the group treated with SSD alone, mice administered SSD liposomes showed significantly lower serum alanine aminotransferase, aspartate aminotransferase, MDA, TNF-α, total cholesterol and triglyceride levels in liver homogenates, while GPx and total SOD levels were significantly elevated. These observations suggest that SSD liposomes exhibit enhanced hepatoprotective effects in alleviating alcoholic hepatitis in mice, likely due to their improved antioxidative and anti-inflammatory properties (65).

NAFLD

Adipogenesis is the developmental process by which preadipocytes differentiate into mature adipocytes, leading to fat accumulation (66). Research has shown that SSD suppresses adipogenesis in 3T3-L1 adipocytes by promoting AMP-activated protein kinase phosphorylation and its substrate acetyl-CoA carboxylase during the early stage of adipogenesis, while concurrently inhibiting the phosphorylation of the ERK1/2 and p38 MAPK pathways (67). In a study focused on SSD's effects on NAFLD, SSD was found to significantly reduce fatty acid biosynthesis by downregulating fatty acid synthase (FASN) and acetyl-CoA carboxylases α (ACACA) expression and to enhance fatty acid degradation through increased expression of COX1 and carnitine palmitoyltransferase-1α (CPT1α) (22). Building on these outcomes, Gu et al (68) conducted further investigations using a high-fat diet (HFD)- and fructose water-induced metabolic dysfunction-associated fatty liver disease mouse model along with HepG2 cells, primary mouse hepatocytes and adipocytes. They discovered that SSD acts as a potent peroxisome proliferator-activated receptor α (PPARα) activator, promoting fatty acid oxidation in hepatocytes and adipocytes while upregulating insulin-induced genes 1 and 2 (INSIG1/2) expression, which inhibits sterol regulatory element-binding protein 1c (SREBP-lc) maturation and thereby reduces fatty acid production. Of note, the lipid-regulating effects of SSD were nullified by the PPARα inhibitor GW6471, indicating that SSD mitigates metabolism-related fatty liver disorder via coordinated modulation of the PPARα and INSIG/SREBP-1c pathways (68).

Liver fibrosis

Previous studies highlighted SSD's protective effects against CCl4-induced liver fibrosis (23,69-71), though detailed mechanisms were initially unclear. Recent research has provided deeper insights. In vitro experiments demonstrated that SSD induced apoptosis in HSC-T6 and LX-2 cells through both caspase-3-dependent and -independent pathways, promoting Bax and Bak translocation to mitochondria, which disrupts mitochondrial function and membrane potential, ultimately releasing apoptotic factors (72). Further investigations using TGF-β and CCl4 to establish liver fibrosis models in vitro and in vivo revealed that SSD significantly reduced fibrosis markers Col-1 and α-SMA. Importantly, SSD inhibited fibrosis progression in primary HSCs and TGF-β-treated LX-2 cells by modulating the G protein-coupled estrogen receptor 1/autophagy pathway (73). An additional study noted that SSD attenuated oxidative stress-induced HSC-T6 activation and fibrosis progression through regulation of estrogen receptor β (Erβ), suggesting that SSD may act as a novel phytoestrogen with estrogen-like effects (31). Lin et al (32) demonstrated that SSD combats liver fibrosis via the ERβ/NLRP3 inflammasome (NLRP3, IL-18 and IL-1β) pathway. Further supporting this, Zhang et al (33) found that SSD reduces liver fibrosis by stimulating the ERβ pathway and downregulating the ROS/NLRP3 inflammasome. Collectively, these outcomes underscore SSD's strong potential as an anti-liver fibrosis agent.

Intestinal diseases
Functional dyspepsia (FD)

FD ranks among the most prevalent digestive tract disorders. Zeng et al (74) developed a model to investigate SSD's impact on apoptosis, autophagy and the morphology of Cajal intestinal cells in FD. Their findings showed that SSD administration in FD rats prevented inflammatory cell infiltration, restored the normal gastric mucosal structure and significantly upregulated light chain 3 I/II, ghrelin and substance P (SP) protein expression, while reducing apoptosis rates. SSD also markedly improved the ultrastructure and clarity of intestinal Cajal cells (ICCs), suggesting it enhances ICC morphology, regulates excessive autophagy and alleviates gastrointestinal motility disorders in FD by modulating ghrelin and SP levels (74).

Ulcerative colitis (UC)

In a separate study on DSS-induced UC in mice, SSD significantly decreased pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) and elevated mRNA levels of the anti-inflammatory cytokine IL-10 (75). The primary mechanism involves SSD's suppression of NF-κB activation and modulation of the gut microbiota to mitigate DSS-induced intestinal inflammation (75).

Chronic pancreatitis (CP)

CP is a progressive fibro-inflammatory syndrome where acinar cell injury initiates inflammation and pancreatic stellate cell (PSC) activation (76). Cui et al (77) found that SSD alleviates pancreatic fibrosis by inhibiting PSC autophagy through the PI3K/AKT/mTOR pathway. Their subsequent research showed that SSD further reduces pancreatic injury by inhibiting acinar cell apoptosis and inflammation via the MAPK signaling pathway (37).

Kidney diseases
Nephritis

Prior research has shown that SSD can prevent aminonucleoside-induced proteinuria in rats (78). Another study indicated that SSD protects renal tubular epithelial cells from high-glucose-induced injury through modulation of SIRT3 (79). Li et al (51) demonstrated that SSD hinders the progression of mesangial proliferative glomerulonephritis in rats by downregulating TGF-β1 and reducing macrophages and CD8+ T lymphocytes. Additionally, it has been suggested that SSD reduces renal inflammation and apoptosis in a sepsis mouse model by inhibiting the transcription factor-7 (TCF7)/Fos-like antigen 1/matrix metalloproteinase (MMP)-9 axis (24), supporting its protective role in renal inflammation.

CKD

In CKD, skeletal muscle atrophy diminishes quality of life and increases morbidity and mortality (80). Huang et al (41) developed a muscle atrophy model using 5/6 nephrectomized mice and dexamethasone-treated C2C12 myotubes. Their results revealed that SSD activates the PI3K/AKT/Nrf2 pathway, reducing OS and alleviating CKD-induced muscle atrophy (41).

Nephrotoxicity

Cisplatin (DDP)-induced nephrotoxicity significantly limits the efficacy of this widely used anticancer drug (81), with inflammation and apoptosis likely contributing to the observed renal damage. Ma et al (82) examined the anti-apoptotic, anti-inflammatory and antioxidant effects of SSD in DDP-induced injury in human renal cortex and HK-2 human proximal tubular epithelial cells. Their findings indicated that SSD substantially increased the survival rate of DDP-treated HK-2 cells, improved nuclear morphology and reduced vesicle-3 activation and apoptosis. In addition, SSD treatment significantly lowered TNF-α, IL-1β and IL-6 secretion, NO generation and iNOS expression. The underlying mechanism suggests that SSD mitigates DDP-induced nephrotoxicity by inhibiting the MAPK and NF-κB signaling pathways (82).

Renal failure

Peritoneal dialysis has the potential to enhance life quality and prolong survival in patients with renal failure; however, peritoneal fibrosis can frequently lead to treatment discontinuation (83). Liu et al (84) found that SSD may alleviate peritoneal fibrosis in renal failure rats by modulating the TGF-β1/BMP7/Gremlin1/Smads pathway, though further research is needed to confirm SSD's mechanism in renal failure.

AD

AD, a neurodegenerative disorder, manifests primarily through dementia, memory loss and language deficits (85). As elevated ROS levels are implicated in OS in AD (86). Du et al (87) showed that SSD alleviates glutamate-induced oxidative cytotoxicity in SH-SY5Y human neuroblastoma cells by activating the Nrf2 pathway, underscoring SSD's neuroprotective effects. However, it has been reported that SSD may impair cognitive function in mice by inhibiting hippocampal neurogenesis through the AKT/forkhead box G1 pathway (27). Another study found that SSD decreases the viability of neural stem/progenitor cells by disrupting neurotrophic factor receptor signaling, thus impairing hippocampal neurogenesis and potentially leading to cognitive deficits (88). These observations suggest a complex role for SSD in AD, with both protective and adverse effects, highlighting the need for further research to clarify SSD's impact on neurodegeneration.

Depression

Depression is a severe, recurrent disorder characterized by persistent low mood, anhedonia and impaired cognitive function (89). Early studies indicated that SSD therapy significantly increased hippocampal neurogenesis in rats exposed to unpredictable chronic mild stress (UCMS), as evidenced by elevated doublecortin levels. In addition, SSD treatment boosted hippocampal neuromolecule levels, such as p-CREB and BDNF, in UCMS rats, suggesting that SSD may counter CMS-induced depressive behaviors partly by enhancing hypothalamic-pituitary-adrenal axis function and supporting hippocampal neurogenesis (90). Further research demonstrated that SSD alleviates depressive-like behavior in UCMS rats by reducing NF-κB and microRNA (miR)-155, while upregulating the fibroblast growth factor 2 (91), homer protein homolog 1-metabotropic glutamate receptor 5 and mTOR signaling pathways (92). A recent study found that SSD alleviates depression in UCMS mice by promoting NLRP3 ubiquitination and suppressing inflammasome activation (26). SSD also reduced LPS-induced depression-like behavior in mice by inhibiting microglial activation and neuroinflammation, potentially through downregulation of the mammaglobin 1/Toll-like receptor 4/NF-κB pathway (93). These insights suggest SSD as a promising candidate for treating depressive disorders.

Diabetes

Diabetes complications include diabetic nephropathy (DN) and diabetic peripheral neuropathy (DPN) (94). Zhao et al (79) showed that SSD protects renal tubular epithelial cells (NRK-52E) from high glucose-induced OS by upregulating SIRT3, elucidating its protective mechanism against DN. Another study revealed that SSD improved body weight, lowered blood glucose levels, alleviated mechanical and thermal hyperalgesia, enhanced nerve conduction velocity and reduced pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) in streptozotocin/HFD-induced DPN in rats. Of note, SSD mitigated DPN by inhibiting the aquaporin 1/Ras homolog family member A/Rho-associated protein kinase signaling pathway (25).

Thyroiditis

HT is an autoimmune condition (95). Du et al (45) reported that SSD therapy reduced lymphocyte infiltration in the thyroid tissue of HT mice, lowered reduced serum thyroid peroxidase antibody levels and suppressed the expression of Th1-type cytokine interferon-γ and Th17-type cytokine IL-17. SSD also shifted the M1/M2 macrophage balance in the spleen toward M2 polarization. These observations suggest that SSD could modulate Th1/Th2 and Th17/Treg imbalances and mitigate HT severity in murine models by promoting M2 macrophage differentiation (45).

Osteoarthritis

Osteoarthritis is the most prevalent joint disease among the elderly, characterized by inflammation and autophagy dysregulation (96). A literature survey by Jiang et al (97) indicated that SSD may inhibit inflammation and modulate autophagic processes by downregulating the PI3K/AKT/mTOR signaling pathway, positioning SSD as a promising treatment option for osteoarthritis. Wu et al (98) demonstrated in their experimental investigation that SSD inhibited IL-1β-induced apoptosis in differentiated ATDC 5 chondrocytes in vitro. Furthermore, SSD effectively prevented cartilage degeneration in the knee joints of mice and reduced the number of osteochondrocytes in the subchondral bone. Of note, SSD activates the Nrf2/heme oxygenase-1/ROS axis, suppresses the production of inflammatory mediators and protects against ECM degradation, thereby delaying the progression of osteoarthritis in destabilization of the medial meniscus model mice (98). A recent study also reported that SSD reduces inflammatory responses in osteoarthritis mice and chondrocytes and mediates autophagy by upregulating miR-199-3p, which targets TCF4 (99). These insights collectively support the notion that SSD mitigates the development of osteoarthritis through its anti-inflammatory and autophagic effects.

Cancer
Thyroid cancer

In thyroid cancer, SSD has been shown to enhance p53 and Bax expression while reducing Bcl-2 levels, promoting apoptosis in thyroid cancer cells. In addition, SSD notably elevates p21 and inhibits cyclin-dependent kinase 2 and cyclin D1, leading to the suppression of human thyroid cancer cell growth (100).

Lung cancer

Prior research has established that SSD inhibits growth and induces programmed cell death in non-small cell lung cancer (NSCLC) cells (A549 and H1299), primarily by inhibiting STAT3 phosphorylation and activating caspase 3 (52). Chen et al (46) further elucidated the anti-cancer mechanism of SSD, revealing that it induces apoptosis in A549 cells through two pathways: TGFα-JNK-p53 and TGFα-Rassfia-Mst1. SSD also inhibits A549 cell proliferation by activating two pathways: TGF-β-p53/p21/p27/p15/p16 and TGF-α/Rassfia/cyclin D1 (46). Drug resistance remains a significant challenge in lung cancer treatment (101). In vivo and in vitro studies by Tang et al (102) indicated that SSD reduced p-STAT3 levels and increased Bcl-2 expression. Downregulation of STAT3 enhanced lung cancer cell responsiveness to gefitinib, suggesting that the combination of SSD and gefitinib may yield improved anti-tumor effects in NSCLC cells, linked to the inhibition of the STAT3/Bcl-2 signaling pathway. Recent investigations have also demonstrated that SSD induces apoptosis in HCC827 and A549 NSCLC cells by increasing ROS levels and activating the NF-κB/NLRP3/caspase-1/gasdermin D pathway (50). These outcomes provide a strong rationale for the use of SSD in the treatment of NSCLC.

Breast cancer

Elevated expression of P-glycoprotein (P-gp) in multidrug-resistant (MDR) cells poses a significant barrier to effective cancer chemotherapy (103). Research indicates that SSD effectively counteracts P-gp-mediated multidrug resistance in MCF-7/adriamycin breast cancer cells (104). Another study corroborated these findings, revealing that the combination of DOX and SSD yielded a stronger anti-cancer effect compared to either DOX or SSD alone (105). Furthermore, Wang et al (49) demonstrated that SSD significantly inhibits β-catenin and its associated downstream target genes, leading to programmed cell death in various triple-negative breast cancer cell lines (HCC1937, MDA-MB-468 and MDA-MB-231 and MCF-7). Fu et al (106) expanded on the mechanisms underlying SSD's anti-breast cancer activity, showing that it inhibits autophagosome-lysosome fusion and induces autophagy-independent apoptosis via caspase-3 activation in MDA-MB-231 cells. A recent untargeted metabolomic analysis highlighted that SSD exerts anti-breast cancer effects by regulating basal metabolism (107), with changes in serum metabolites observed in breast cancer mice following SSD treatment, including alterations in sphingolipid metabolism, glycerophospholipid metabolism and the biosynthetic pathways for phenylalanine, tyrosine and tryptophan (107).

Liver cancer

Hepatocellular carcinoma (HCC) ranks among the most common malignancies (108). In an earlier study utilizing an diethyl nitrosamine (DEN)-induced rat HCC model, SSD was found to inhibit the expression of syndecan-2, MMP-2, MMP-13 and TIMP-2 in liver tissue, suggesting its potential anti-HCC effects (109). COX-2 (110) and C/EBPβ (111) are known to be associated with inflammation and carcinogenesis. One investigation revealed that SSD inhibited the proliferation of HCC SMMC-7721 cells and induced apoptosis by downregulating COX-2 expression and reducing prostaglandin E2 synthesis (112). In addition, SSD was shown to impede DEN-induced liver cancer in rats by suppressing C/EBPβ and COX-2 levels (113). Two further studies indicated that SSD inhibits COX-2 expression via the p-STAT3/HIF-1α pathway (114) and the p-STAT3/C/EBPβ signaling pathway (115), thereby exerting an anti-HCC effect.

The integration of SSD and radiotherapy demonstrates superior efficacy in the treatment of liver cancer compared to their individual applications. Research indicates that SSD enhances the radiosensitivity of SMMC-7721 liver cancer cells by regulating the G0/G1 and G2/M cell cycle checkpoints, which correlates with the upregulation of p53 and Bax and the downregulation Bcl-2 (116). A similar study confirmed that SSD also increases the radiosensitivity of liver cancer cells (SMMC-7721 and HepG2) under hypoxic conditions by suppressing hypoxia-inducible factor-1α, thereby triggering the upregulation of p53 and Bax and the downregulation of Bcl-2 (117). The activation of glioma-associated oncogene (GLI) family proteins and significant protein assimilation are characteristic of HCC cells. Hypoxia activates the sonic hedgehog pathway, which promotes epithelial-to-mesenchymal transition (EMT), invasion and chemosensitivity in HCC cells, with hypoxia-dependent GLI protein activation requiring SUMOylation (118-120). Zhang et al (121) demonstrated that SSD inhibited liver cancer cells (Hep3B) and enhanced chemotherapy sensitivity through sentrin-specific protease 5-dependent inhibition of Gli1 SUMOylation in hypoxic environments. Furthermore, SSD significantly increased radiation-induced apoptosis in SMMC-7721 and MHCC97L liver cancer cells while concurrently inhibiting cell proliferation (122). The introduction of the autophagy inhibitor chloroquine or the mTOR agonist MHY1485 attenuated the pharmacodynamic effects of SSD, suggesting that SSD enhances radiation-induced apoptosis in HCC cells by promoting autophagy through the inhibition of mTOR phosphorylation (123).

Ongoing investigations into the anti-HCC mechanisms of SSD have included a non-targeted metabolomics study revealing that SSD, in combination with neuropilin (NRP)-1 gene knockout, exerts anti-liver cancer effects in vitro, primarily by modulating lipid transport and phospholipid metabolism (124). In addition, the potential interactions between SSD and its hypothetic target, NRP-1, are under investigation. Li et al (125) found that SSD significantly induced the expression and enzyme activity of cytochrome P 450 enzyme (CYP)1A2 and CYP2D6 in HepaRG cells. Another study utilizing molecular docking indicated that SSD substantially suppressed the expression and enzyme activity of CYP3A4 protein in HepaRG cells; however, experimental validation is still required (126).

Intrahepatic cholangiocarcinoma (ICCA)

ICCA is a highly lethal malignant tumor, with systemic chemotherapy using gemcitabine as the primary clinical treatment option (127,128). Norepinephrine (NE) and epinephrine (E) have been shown to promote ICCA cell growth and diminish the efficacy of gemcitabine through stimulation of the β 2-adrenergic receptors (ADRB2) receptor (129). A 2023 study demonstrated that SSD can reverse the adverse effects of gemcitabine in ICCA cells by reducing ADRB2 levels. Furthermore, SSD inhibited drug efflux and glycolysis in ICCA cells by modulating the expression of multidrug resistance 1, ABC subfamily G, isoform 2 protein, hexokinase 2 and glucose transporter 1. This suggests that SSD may counteract NE- and E-induced gemcitabine resistance in intrahepatic cholangiocarcinoma via downregulation of ADRB2 and glycolytic signaling pathways (130).

Gastric cancer (GC)

GC remains a significant contributor to cancer-related mortality, with drug resistance being a critical factor in its poor prognosis (131). Research by Hu et al (28) indicated that SSD enhances the efficacy of DDP by inhibiting the growth and invasiveness of SGC-7901 and SGC-7901/DDP-resistant cells, thereby increasing DDP-induced apoptosis. SSD appears to enhance the sensitivity of GC cells to DDP, potentially through inhibition of the IKKβ/NF-κB pathway, which induces apoptosis and autophagy in GC cells (28). A recent network pharmacology study identified six key targets linking SSD to gastric cancer: VEGFA, IL-2, CASP3, BCL2L1, MMP2 and MMP1. Experimental results demonstrated that SSD induces apoptosis in cancer cells by enhancing caspase-3 activity and elevating Bcl-2 levels, while also inhibiting migration and invasion through upregulation of IL-2 and downregulation of MMP1 and MMP2 (132). These findings indicate that SSD may play a role in overcoming drug resistance in GC treatment.

Pancreatic cancer

Pancreatic cancer remains one of the deadliest malignancies with limited therapeutic options (133). SSD has been shown to inhibit the growth of pancreatic cancer cells (BxPC3, PANC1 and Pan02) and enhance the cleavage of apoptotic proteins caspase-3 and caspase-9 by activating the MAPK kinase 4-JNK signaling cascade (47). An additional in vivo and in vitro study found SSD directly suppresses pancreatic cancer cell apoptosis and invasion while modulating the immunosuppressive microenvironment to reactivate local immune responses. The primary mechanism involves reducing M2 macrophage polarization by downregulating phosphorylated STAT6 levels and the PI3K/AKT/mTOR signaling pathway (134). These findings suggest that SSD holds promise as a potential treatment for pancreatic cancer.

Kidney cancer

Cai et al (135) reported that SSD induces apoptosis in human renal cell adenocarcinoma cells (769-P and 786-O) by upregulating the p53 protein and arresting the cell cycle in the G0/G1 phase, primarily through inhibition of the EGFR/p38 signaling pathway.

Prostate cancer (PC)

PC ranks among the most prevalent malignancies affecting males (136). Research by Yao et al (137) demonstrated that SSD induces apoptosis in DU145 cells via the intrinsic apoptotic pathway. The primary mechanism involves SSD causing cell cycle arrest in G0/G1 phase through upregulation of p53 and p21, along with regulation of Bcl-2 family proteins, dissipation of mitochondrial membrane potential, release of cytochrome into the cytosol and activation of caspase-3, leading to apoptosis. Another study revealed that SSD suppresses the growth, migration and invasion of prostate cancer (DU145 and CWR22Rv1) by reversing EMT and inhibiting the expression and activity of MMP2/9 (138). In addition, SSD impedes the self-renewal capacity of cancer stem cell phenotypes by reducing glycogen synthase kinase 3β phosphorylation, thereby blocking the Wnt/β-catenin signaling pathway (138). These findings position SSD as a promising therapeutic agent for PC.

Ovarian cancer

DDP and its derivatives serve as first-line anticancer drugs for ovarian cancer (139). Studies indicate that SSD sensitizes drug-resistant ovarian cancer cells to DDP-induced apoptosis by promoting mitochondrial fission and causing G2/M phase arrest. The specific mechanism involves the enhancement of calcium signaling, upregulating of mitochondrial fission proteins dynamin-related protein 1 and optic atrophy 1 and inhibition of MMPs (140).

Endometrial cancer

Research conducted by Tang et al (141) revealed that SSD upregulates p21 and Cyclin B, leading to G2/M phase retention in endometrial cancer Ishikawa cells. Furthermore, SSD increased ROS levels, reduced the mitochondrial membrane potential, activated Bcl-2 and caspase family cascades, and mediated both endogenous and exogenous apoptosis pathways in Ishikawa cells. In addition, SSD influences three classic MAPK pathways (Ras/Raf/MEK/ERK, JNK and p38-MAPK pathways) to inhibit cell metastasis (141).

Osteosarcoma

Although human osteosarcoma has a low incidence rate, its prognosis is often poor compared to other cancers (142). A study assessing SSD's therapeutic potential in osteosarcoma demonstrated that it significantly suppresses the proliferation of 143B and MG-63 cells. The underlying mechanism includes the upregulation of tumor protein 53 (p53) and its downstream targets (p21, p27, Bcl-2-like protein 4 and cleaved caspase-3), along with the downregulation of cyclin D1 expression levels (143). Another study within the same year indicated that SSD, either alone or in combination with the JNK inhibitor SP600125, inhibits proliferation, induces apoptosis, and suppresses migration and invasion in human osteosarcoma U2 cells. However, SP600125 alone did not show a significant effect on U2 cells. The mechanism involves enhancement of cytochrome release, elevation of the Bax/Bcl-2 ratio, and activation of caspases -3, -8 and -9, indicating that apoptosis is triggered via both mitochondrial and death receptor pathways (144). These findings provide a theoretical basis for the use of SSD in osteosarcoma treatment.

Glioblastoma (GBM)

An earlier study demonstrated that SSD can inhibit apoptosis in human GBM U87 cells, primarily by significantly suppressing the phosphorylation of AKT and ERK while promoting the expression of phosphorylated JNK and cleaved caspase-3 (145). A subsequent investigation revealed that SSD notably impedes the growth of RG-2, U87-MG and U251 cells in a sugar-dependent manner, leading to a marked increase in the proportion of apoptotic cells. Further research has indicated that SSD induces apoptosis and autophagy by activating endoplasmic reticulum stress in GBM cells, thereby exerting anti-GBM effects (146). Chemotherapy remains a critical adjunctive treatment for glioblastoma; however, resistance to chemotherapy presents an urgent clinical challenge for neuro-oncologists (147). Liang et al (148) discovered that SSD partially suppressed the migration, invasion and apoptosis of LN-229 cells. When combined with temozolomide (TMZ), SSD enhanced the chemotherapeutic efficacy of TMZ by increasing the apoptosis rate and LDH release in LN-229 cells, while also inhibiting the expression of stem cell factors (octamer-binding transcription factor 4, SRY-box transcription factor 2, myelocytomatosis viral oncogene homolog and Kruppel-like factor 4 at both gene and protein levels (148). These insights lay the groundwork for the potential application of SSD in overcoming GBM chemotherapy resistance, although further research is required for a more comprehensive understanding.

Medulloblastoma (MB)

MB is a highly malignant embryonic tumor and the most common primary tumor of the posterior fossa in children (149). A 2021 study demonstrated that SSD inhibits tumor progression in MB-transplanted mice by suppressing the Hedgehog signaling pathway (150). However, further investigations are necessary to elucidate the mechanisms underlying SSD's anti-MB effect.

Acute myeloid leukemia (AML)

AML remains an uncommon but potentially devastating condition with persistently elevated mortality rates (151). The fat mass and obesity-associated protein (FTO), an mRNA N6-methyladenosine (m6A) demethylase, functions as an oncogene that facilitates leukemic oncogene-driven cellular transformation and leukemogenesis (152,153). Research by Sun et al (154) demonstrated that SSD significantly suppressed AML cell proliferation, induced apoptosis and caused cell cycle arrest both in vivo and in vitro. At the molecular level, SSD specifically targets FTO, thereby enhancing m6A RNA methylation, which reduces the stability of downstream gene transcripts and inhibits associated pathways. Notably, SSD also mitigates FTO/m6A-mediated resistance to tyrosine kinase inhibitors in leukemia (154). This investigation reveals promising therapeutic avenues for the treatment of leukemia.

Conclusion and prospects

TCM monomers are single components extracted from TCM, characterized by a clear chemical structure and established pharmacological activity. Compared to compound formulas and proprietary Chinese medicines, TCM monomers offer advantages such as well-defined ingredients, clear mechanisms of action and straightforward quality control. Consequently, they exhibit promising applications in the treatment of various diseases (155-158). SSD is a prominent bioactive component of Bupleurum, demonstrating pharmacological activities that include hepatoprotective, anti-inflammatory, antiviral and anti-tumor effects (15,18,159). The mechanisms underlying these pharmacological actions involve inflammation, OS, immune modulation, autophagy and apoptosis, thereby influencing the progression of numerous diseases (Fig. 4). An unpublished study by our group also identified that SSD can exert anti-hepatic fibrosis effects by modulating the expression of biological clock genes in liver tissue, with findings set for forthcoming publication. These investigations indicate that SSD is a potential monomer for addressing lung diseases, kidney diseases, liver diseases and tumors.

Figure 4

Role of SSD in disease regulation through multiple pathways. SSD participates in disease modulation via multiple pathways: It reduces myocardial damage by inhibiting the MAPK pathway, mitigates pulmonary fibrosis by targeting the Ang/Tie and TGF-β/Smads pathways, and slows non-alcoholic fatty liver disease progression by inhibiting the INSIG/SREBP-1c/FASN and activating the PPARα/CPT1α pathway. SSD also alleviates liver fibrosis by inhibiting the ERβ/ROS/NLRP3 inflammasome pathway, mitigates renal failure through the TGF-β/BMP7/Gremlin1/Smads pathway and reduces renal inflammation via the TCF7/FOSL1/MMP-9 and PI3K/AKT/Nrf2/ROS pathways. In chronic pancreatitis, SSD activates the PI3K/AKT/mTOR/ECM pathway, while in ulcerative colitis, it suppresses inflammation by inhibiting the NF-κB pathway. SSD also alleviates functional dyspepsia through inhibition of the BCL-2/BAX/Caspase-3 pathway. AKT, protein kinase B; Ang, angiotensin; ASC, adapter protein apoptosis associated speck-like protein containing a CARD; BMP7, bone morphogenetic protein 7; Col-1, collagenol-1; α-SMA, α-smooth muscle actin; FASN, fatty acid synthase; CPT1α, carnitine palmitoyltransferase-1 α; ECM, extracellular matrix; FOSL1, Fos-like antigen 1; IL-6, interleukin-6; MAPK, mitogen-activated protein kinases; mTOR, mechanistic target of rapamycin; MMP-9, matrix metalloproteinase-9; NF-κB, nuclear factor-κB; NLRP3, NLR family pyrin domain containing 3; Nrf2, nuclear factor erythroid 2-related factor 2; PI3K, phosphatidylinositol-3 kinase; PPARα, peroxisome proliferator-activated receptor α; ROS, reactive oxygen species; SREBP-lc, sterol regulatory element-binding protein 1c; SSD, Saikosaponin D; TCF7, transcription factor-7; TGF-β1, transforming growth factor β; Tie, angiopoietin/tyrosine kinase with immunoglobulin-like and epidermal growth factor homology; TNF-α, tumor necrosis factor-α.

Natural products and botanical sources serve as vital reservoirs for anti-cancer agents. Several natural compounds and their derivatives, such as Matrine (160), ginsenosides (161), strychnine (162) and berbamine (163), have demonstrated anti-cancer effects. While SSD is a natural substance with significant anti-tumor potential, it is essential to thoroughly understand its possible adverse effects to ensure pharmaceutical safety during its application. Research has identified toxic effects of SSD, including hepatotoxicity, neurotoxicity, hemolysis and cardiotoxicity (88,164,165). Natural compounds derived from plants are generally considered less toxic to normal cells than chemically synthesized drugs, thus offering greater potential safety as anti-cancer therapies (166-169).

Despite its potential, several challenges remain in the research surrounding SSD. Firstly, pharmacokinetic studies are insufficient. Although SSD is categorized as a triterpenoid saponin with poor water solubility, its bioavailability may exceed current expectations. Modern research indicates that bioavailability can be enhanced through methods such as nanoparticle encapsulation and liposomal formulations (65,170). In addition, investigations into SSD metabolism reveal its conversion into specific metabolites (e.g., prosaikogenins and saikogenins) within the body, influenced by various transformation factors including the intestinal flora, gastric acid and enzymes (171,172). However, a comprehensive understanding of the pharmacokinetics of SSD after systemic administration is currently lacking. For instance, while SSD excretion has been documented, the specific metabolic pathways and resulting metabolites are not fully elucidated, hindering a complete grasp of its pharmacokinetic properties. Secondly, the mechanistic research on SSD is limited. Current therapeutic mechanisms attributed to SSD in treating diseases are primarily associated with specific proteins and pathways, with insufficient exploration of the complete upstream and downstream pathways and the interactions between multiple pathways. Although evidence supports SSD's significant preventive and therapeutic effects on liver disease, the precise mechanisms of action are not fully characterized. For instance, despite SSD's recognized therapeutic efficacy for liver diseases, comparative analyses of its mechanisms across various liver conditions are sparse, complicating the differentiation and effective treatment of specific liver diseases in clinical settings. Thirdly, the synergistic effects of SSD with other compounds are underexplored. The treatment of tumors poses significant challenges and the integration of traditional Chinese and Western medicine is becoming increasingly prevalent in oncological therapies (173,174). SSD has shown promise as an anti-cancer monomer; however, research on its application, either alone or in combination with other TCM monomer saponins, remains limited. Furthermore, investigations into the synergistic effects of SSD alongside other compounds, such as SSA and SSC, are scarce. While studies have optimized the extraction processes for SSA and SSD, the interactions among these components within the body and their influence on therapeutic efficacy warrant further examination. Lastly, there is a notable deficiency in clinical application research. Although SSD has demonstrated significant pharmacological activity in preclinical animal studies, there are relatively few investigations into its clinical application. This lack of research restricts its broader clinical utilization and necessitates additional clinical trials to ascertain its safety and efficacy.

In summary, future research on SSD should prioritize pharmacokinetic studies, explore the complex interactions of multiple proteins and pathways and foster collaborative investigations between traditional Chinese and Western medicine. Furthermore, in vivo and in vitro experimental studies on SSD have shown that it is a promising drug in the treatment of various diseases, but to the best of our knowledge, there have been no reports of clinical trials. There should be an increased emphasis on animal studies and clinical trials. Integrating these research approaches will effectively translate basic research findings into clinical practice.

Availability of data and materials

Not applicable.

Authors' contributions

SG: Conceptualization, validation, writing-original draft. YZ and CC: Software, visualization, writing-review & editing. JL: Investigation, methodology. YaW: Visualization, project administration. JuW and YL: Funding acquisition, project administration, formal analysis. All authors have read the manuscript and approved it for publication. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This work was supported by Shanghai Natural Science Foundation (grant no. 22ZR1459400) and Shanghai Science and Technology Innovation Project (grant no. 22S21901100).

References

1 

Zulkifli MH, Abdullah ZL, Mohamed Yusof NIS and Mohd Fauzi F: In silico toxicity studies of traditional Chinese herbal medicine: A mini review. Curr Opin Struct Biol. 80:1025882023. View Article : Google Scholar : PubMed/NCBI

2 

Qian Z, Wang GY, Henning M and Chen Y: Understanding health literacy from a traditional Chinese medicine perspective. J Integr Med. 21:215–220. 2023. View Article : Google Scholar : PubMed/NCBI

3 

Ai C, Zou Y, Liu H, Yang Z and Xi J: Traditional Chinese herbal medicine for allergic diseases: A review. Am J Chin Med. 51:779–806. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Lu Y, Shao M, Xiang H, Wang J, Ji G and Wu T: Qinggan huoxue recipe alleviates alcoholic liver injury by suppressing endoplasmic reticulum stress through LXR-LPCAT3. Front Pharmacol. 13:8241852022. View Article : Google Scholar : PubMed/NCBI

5 

Sun L, Yang Z, Zhao W, Chen Q, Bai H, Wang S, Yang L, Bi C, Shi Y and Liu Y: Integrated lipidomics, transcriptomics and network pharmacology analysis to reveal the mechanisms of Danggui Buxue Decoction in the treatment of diabetic nephropathy in type 2 diabetes mellitus. J Ethnopharmacol. 283:1146992022. View Article : Google Scholar

6 

Xiang H, Shao M, Lu Y, Wang J, Wu T and Ji G: Kaempferol alleviates steatosis and inf lammation during early Non-Alcoholic steatohepatitis associated with liver X Receptor α-Lysophosphatidylcholine acyltransferase 3 signaling pathway. Front Pharmacol. 12:6907362021. View Article : Google Scholar

7 

Paik S, Song GY and Jo EK: Ginsenosides for therapeutically targeting inflammation through modulation of oxidative stress. Int Immunopharmacol. 121:1104612023. View Article : Google Scholar : PubMed/NCBI

8 

Li J, Li F and Jin D: Ginsenosides are promising medicine for tumor and inflammation: A review. Am J Chin Med. 51:883–908. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Zarneshan SN, Fakhri S and Khan H: Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: A mechanistic approach. Pharmacol Res. 177:1060992022. View Article : Google Scholar : PubMed/NCBI

10 

Teng L, Guo X, Ma Y, Xu L, Wei J and Xiao P: A comprehensive review on traditional and modern research of the genus Bupleurum (Bupleurum L., Apiaceae) in recent 10 years. J Ethnopharmacol. 306:1161292023. View Article : Google Scholar : PubMed/NCBI

11 

Sui C, Han WJ, Zhu CR and Wei JH: Recent progress in saikosaponin biosynthesis in bupleurum. Curr Pharm Biotechnol. 22:329–340. 2021. View Article : Google Scholar

12 

Shao S, Jia R, Zhao L, Zhang Y, Guan Y, Wen H, Liu J, Zhao Y, Feng Y, Zhang Z, et al: Xiao-Chai-Hu-Tang ameliorates tumor growth in cancer comorbid depressive symptoms via modulating gut microbiota-mediated TLR4/MyD88/NF-κB signaling pathway. Phytomedicine. 88:1536062021. View Article : Google Scholar

13 

Fan Q, Liu Y, Sheng L, Lv S, Yang L, Zhang Z, Guo J, Fan Y and Hu D: Chaihu-Shugan-San inhibits neuroinflammation in the treatment of post-stroke depression through the JAK/STAT3-GSK3β/PTEN/Akt pathway. Biomed Pharmacother. 160:1143852023. View Article : Google Scholar

14 

Jiao H, Fan Y, Gong A, Li T, Fu X and Yan Z: Xiaoyaosan ameliorates CUMS-induced depressive-like and anorexia behaviors in mice via necroptosis related cellular senescence in hypothalamus. J Ethnopharmacol. 318:1169382024. View Article : Google Scholar

15 

Luo K, Dai RJ, Zeng YB, Chang WJ, Deng YL and Lv F: Triterpenoid saponins from Bupleurum marginatum and their anti-liver fibrotic activities. J Asian Nat Prod Res. 26:858–864. 2024. View Article : Google Scholar : PubMed/NCBI

16 

Kang Y, Gao Y, Li X, Guo X, Liu Z, Li W, Wei J and Qi Y: Bupleurum chinense exerts a mild antipyretic effect on LPS-induced pyrexia rats involving inhibition of peripheral TNF-α production. J Ethnopharmacol. 310:1163752023. View Article : Google Scholar

17 

Zhou J, He X, Sun R, Yu Z, Wang C, Deng S, Zhang B, Huang S, Han C and Li D: Lignans from Bupleurum marginatum and their antioxidant activity. Nat Prod Res. 36:5016–5021. 2022. View Article : Google Scholar

18 

Sun P, Li Y, Wei S, Zhao T, Wang Y, Song C, Xue L, Wang F, Xiao L, Wu J and Qiao M: Pharmacological effects and chemical constituents of bupleurum. Mini Rev Med Chem. 19:34–55. 2019. View Article : Google Scholar

19 

Li XQ, Song YN, Wang SJ, Rahman K, Zhu JY and Zhang H: Saikosaponins: A review of pharmacological effects. J Asian Nat Prod Res. 20:399–411. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Zhang YJ, Wu SS, Chen XM, Pi JK, Cheng YF, Zhang Y, Wang XJ, Luo D, Zhou JH, Xu JY, et al: Saikosaponin D Alleviates DOX-induced cardiac injury in vivo and in vitro. J Cardiovasc Pharmacol. 79:558–567. 2022. View Article : Google Scholar : PubMed/NCBI

21 

Wang HW, Liu M, Zhong TD and Fang XM: Saikosaponin-d attenuates ventilator-induced lung injury in rats. Int J Clin Exp Med. 8:15137–15145. 2015.PubMed/NCBI

22 

Li X, Ge J, Li Y, Cai Y, Zheng Q, Huang N, Gu Y, Han Q, Li Y, Sun R, et al: Integrative lipidomic and transcriptomic study unravels the therapeutic effects of saikosaponins A and D on non-alcoholic fatty liver disease. Acta Pharm Sin B. 11:3527–3541. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Fan J, Li X, Li P, Li N, Wang T, Shen H, Siow Y, Choy P and Gong Y: Saikosaponin-d attenuates the development of liver fibrosis by preventing hepatocyte injury. Biochem Cell Biol. 85:189–195. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Yao T, Zhang L, Fu Y, Yao L, Zhou C and Chen G: Saikosaponin-d alleviates renal inflammation and cell apoptosis in a mouse model of sepsis via TCF7/FOSL1/matrix metalloproteinase 9 inhibition. Mol Cell Biol. 41:e00332212021. View Article : Google Scholar : PubMed/NCBI

25 

Xiang Q, Liu Y and Chen L: Saikosaponin d (SSD) alleviates diabetic peripheral neuropathy by regulating the AQP1/RhoA/ROCK signaling in streptozotocin-induced diabetic rats. Acta Diabetol. 60:805–815. 2023. View Article : Google Scholar : PubMed/NCBI

26 

Gao T, Wang T, Wu L, Tong Y, Tian J, Zhao K and Wang H: Saikosaponin-d alleviates depression by promoting NLRP3 ubiquitination and inhibiting inflammasome activation. Int Immunopharmacol. 127:1113242024. View Article : Google Scholar

27 

Lixing X, Zhouye J, Liting G, Ruyi Z, Rong Q and Shiping M: Saikosaponin-d-mediated downregulation of neurogenesis results in cognitive dysfunction by inhibiting Akt/Foxg-1 pathway in mice. Toxicol Lett. 284:79–85. 2018. View Article : Google Scholar

28 

Hu J, Li P, Shi B and Tie J: Effects and mechanisms of saikosaponin D improving the sensitivity of human gastric cancer cells to cisplatin. ACS Omega. 6:18745–18755. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Chen LL, Xia LY, Zhang JP, Wang Y, Chen JY, Guo C and Xu WH: Saikosaponin D alleviates cancer cachexia by directly inhibiting STAT3. Phytother Res. 37:809–819. 2023. View Article : Google Scholar

30 

Wang P, Ren J, Tang J, Zhang D, Li B and Li Y: Estrogen-like activities of saikosaponin-d in vitro: A pilot study. Eur J Pharmacol. 626:159–165. 2010. View Article : Google Scholar

31 

Que R, Shen Y, Ren J, Tao Z, Zhu X and Li Y: Estrogen receptor-β-dependent effects of saikosaponin-d on the suppression of oxidative stress-induced rat hepatic stellate cell activation. Int J Mol Med. 41:1357–1364. 2018.

32 

Lin L, Zhou M, Que R, Chen Y, Liu X, Zhang K, Shi Z and Li Y: Saikosaponin-d protects against liver fibrosis by regulating the estrogen receptor-β/NLRP3 inflammasome pathway. Biochem Cell Biol. 99:666–674. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Zhang K, Lin L, Zhu Y, Zhang N, Zhou M and Li Y: Saikosaponin d alleviates liver fibrosis by negatively regulating the ROS/NLRP3 inflammasome through activating the ERβ pathway. Front Pharmacol. 13:8949812022. View Article : Google Scholar

34 

Kumazawa Y, Takimoto H, Nishimura C, Kawakita T and Nomoto K: Activation of murine peritoneal macrophages by saikosaponin a, saikosaponin d and saikogenin d. Int J Immunopharmacol. 11:21–28. 1989. View Article : Google Scholar : PubMed/NCBI

35 

Luo SQ, Lin LZ and Cordell GA: Saikosaponin derivatives from Bupleurum wenchuanense. Phytochemistry. 33:1197–1205. 1993. View Article : Google Scholar : PubMed/NCBI

36 

Lu CN, Yuan ZG, Zhang XL, Yan R, Zhao YQ, Liao M and Chen JX: Saikosaponin a and its epimer saikosaponin d exhibit anti-inflammatory activity by suppressing activation of NF-κB signaling pathway. Int Immunopharmacol. 14:121–126. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Li C, Cui L, Zhang L, Yang L, Zhuo Y, Cui J, Cui N and Zhang S: Saikosaponin D attenuates pancreatic injury through suppressing the apoptosis of acinar cell via modulation of the MAPK signaling pathway. Front Pharmacol. 12:7350792021. View Article : Google Scholar : PubMed/NCBI

38 

Kassis S, Grondin M and Averill-Bates DA: Heat shock increases levels of reactive oxygen species, autophagy and apoptosis. Biochim Biophys Acta Mol Cell Res. 1868:1189242021. View Article : Google Scholar

39 

Zhang BZ, Guo XT, Chen JW, Zhao Y, Cong X, Jiang ZL, Cao RF, Cui K, Gao SS and Tian WR: Saikosaponin-D attenuates heat stress-induced oxidative damage in LLC-PK1 cells by increasing the expression of anti-oxidant enzymes and HSP72. Am J Chin Med. 42:1261–1277. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Lin X, Wu S, Wang Q, Shi Y, Liu G, Zhi J and Wang F: Saikosaponin-D reduces H2O2-induced PC12 cell apoptosis by removing ROS and blocking MAPK-dependent oxidative damage. Cell Mol Neurobiol. 36:1365–1375. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Huang M, Yan Y, Deng Z, Zhou L, She M, Yang Y, Zhang M and Wang D: Saikosaponin A and D attenuate skeletal muscle atrophy in chronic kidney disease by reducing oxidative stress through activation of PI3K/AKT/Nrf2 pathway. Phytomedicine. 114:1547662023. View Article : Google Scholar : PubMed/NCBI

42 

Su W, Huang S, Zhu H, Zhang B and Wu X: Interaction between PHB2 and Enterovirus A71 VP1 Induces Autophagy and Affects EV-A71 Infection. Viruses. 12:4142020. View Article : Google Scholar : PubMed/NCBI

43 

Li C, Huang L, Sun W, Chen Y, He ML, Yue J and Ballard H: Saikosaponin D suppresses enterovirus A71 infection by inhibiting autophagy. Signal Transduct Target Ther. 4:42019. View Article : Google Scholar : PubMed/NCBI

44 

Sun J, Zheng J, Shi X, Qian H, Jin C and Xu L: Saikosaponin D inhibits proliferation and collagen production of human embryonic lung fibroblasts by regulating TGF-β1/Smads signaling pathway. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 35:256–261. 2019.In Chinese. PubMed/NCBI

45 

Du P, Xu J, Jiang Y, Zhao J, Gao C, Fang Y, Yang X, Yang YP and Zhang JA: Saikosaponin-d attenuates hashimoto's thyroiditis by regulating macrophage polarization. J Immunol Res. 2022:74554942022. View Article : Google Scholar : PubMed/NCBI

46 

Chen X, Liu C, Zhao R, Zhao P, Wu J, Zhou N and Ying M: Synergetic and antagonistic molecular effects mediated by the feedback loop of p53 and JNK between Saikosaponin D and SP600125 on lung cancer A549 cells. Mol Pharm. 15:4974–4984. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Lai M, Ge Y, Chen M, Sun S, Chen J and Cheng R: Saikosaponin D inhibits proliferation and promotes apoptosis through activation of MKK4-JNK signaling pathway in pancreatic cancer cells. Onco Targets Ther. 13:9465–9479. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Wu S, Chen W, Liu K, Ren F, Zheng D, Xu F and Wu H: Saikosaponin D inhibits proliferation and induces apoptosis of non-small cell lung cancer cells by inhibiting the STAT3 pathway. J Int Med Res. 48:3000605209371632020. View Article : Google Scholar : PubMed/NCBI

49 

Wang J, Qi H, Zhang X, Si W, Xu F, Hou T, Zhou H, Wang A, Li G, Liu Y, et al: Saikosaponin D from Radix Bupleuri suppresses triple-negative breast cancer cell growth by targeting β-catenin signaling. Biomed Pharmacother. 108:724–733. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Chen M, Hu C, Yang L, Guo Q, Liang Y and Wang W: Saikosaponin-D induces the pyroptosis of lung cancer by increasing ROS and activating the NF-κB/NLRP3/caspase-1/GSDMD pathway. J Biochem Mol Toxicol. 37:e234442023. View Article : Google Scholar

51 

Li P, Gong Y, Zu N, Li Y, Wang B and Shimizu F: Therapeutic mechanism of Saikosaponin-d in anti-Thy1 mAb 1-22-3-induced rat model of glomerulonephritis. Nephron Exp Nephrol. 101:e111–118. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Hsu YL, Kuo PL and Lin CC: The proliferative inhibition and apoptotic mechanism of Saikosaponin D in human non-small cell lung cancer A549 cells. Life Sci. 75:1231–1242. 2004. View Article : Google Scholar : PubMed/NCBI

53 

Wong VK, Zhang MM, Zhou H, Lam KY, Chan PL, Law CK, Yue PY and Liu L: Saikosaponin-d enhances the anticancer potency of TNF-α via overcoming its undesirable response of activating NF-Kappa B signalling in cancer cells. Evid Based Complement Alternat Med. 2013:7452952013. View Article : Google Scholar

54 

Rawat PS, Jaiswal A, Khurana A, Bhatti JS and Navik U: Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed Pharmacother. 139:1117082021. View Article : Google Scholar : PubMed/NCBI

55 

Song L, Lu G and Tao Y: Saikosaponin D attenuates inflammatory response and cell apoptosis of lipopolysaccharide-induced lung epithelial cells. Clin Respir J. 17:1017–1024. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Moss BJ, Ryter SW and Rosas IO: Pathogenic mechanisms underlying idiopathic pulmonary fibrosis. Annu Rev Pathol. 17:515–546. 2022. View Article : Google Scholar

57 

Wu Y, Zhang J, Wang X, Xu Y and Zheng J: Saikosaponin-d regulates angiogenesis in idiopathic pulmonary fibrosis through angiopoietin/Tie-2 pathway. Acta Histochem. 125:1521002023. View Article : Google Scholar : PubMed/NCBI

58 

Jaeschke H, Akakpo JY, Umbaugh DS and Ramachandran A: Novel therapeutic approaches against acetaminophen-induced liver injury and acute liver failure. Toxicol Sci. 174:159–167. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Liu A, Tanaka N, Sun L, Guo B, Kim JH, Krausz KW, Fang Z, Jiang C, Yang J and Gonzalez FJ: Saikosaponin d protects against acetaminophen-induced hepatotoxicity by inhibiting NF-κB and STAT3 signaling. Chem Biol Interact. 223:80–86. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Lin L, Que R, Shen Y, Chen Y, Yan N and Li Y: Saikosaponin-d alleviates carbon-tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL-7702 cell line. Mol Med Rep. 17:7939–7946. 2018.PubMed/NCBI

61 

Chen Y, Que R, Lin L, Shen Y, Liu J and Li Y: Inhibition of oxidative stress and NLRP3 inflammasome by Saikosaponin-d alleviates acute liver injury in carbon tetrachloride-induced hepatitis in mice. Int J Immunopathol Pharmacol. 34:20587384209505932020. View Article : Google Scholar : PubMed/NCBI

62 

Singal AK, Bataller R, Ahn J, Kamath PS and Shah VH: ACG Clinical Guideline: Alcoholic liver disease. Am J Gastroenterol. 113:175–194. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Tsuchida T and Friedman SL: Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 14:397–411. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Jiang H, Liu J, Zhang K and Zeng Q: Saikosaponin D inhibits the proliferation and promotes the apoptosis of rat hepatic stellate cells by inducing autophagosome formation. Evid Based Complement Alternat Med. 2021:54517582021. View Article : Google Scholar : PubMed/NCBI

65 

Yu X, Pan J, Shen N, Zhang H, Zou L, Miao H and Xing L: Development of Saikosaponin D liposome nanocarrier with increased hepatoprotective effect against alcoholic hepatitis mice. J Biomed Nanotechnol. 17:627–639. 2021. View Article : Google Scholar

66 

Byrne CD and Targher G: NAFLD: A multisystem disease. J Hepatol. 62(1 Suppl): S47–S64. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Lim SH, Lee HS, Han HK and Choi CI: Saikosaponin A and D inhibit adipogenesis via the AMPK and MAPK signaling pathways in 3T3-L1 adipocytes. Int J Mol Sci. 22:114092021. View Article : Google Scholar : PubMed/NCBI

68 

Gu Y, Duan S, Ding M, Zheng Q, Fan G, Li X, Li Y, Liu C, Sun R and Liu R: Saikosaponin D attenuates metabolic associated fatty liver disease by coordinately tuning PPARα and INSIG/SREBP1c pathway. Phytomedicine. 103:1542192022. View Article : Google Scholar

69 

Abe H, Sakaguchi M, Odashima S and Arichi S: Protective effect of saikosaponin-d isolated from Bupleurum falcatum L. on CCl4-induced liver injury in the rat. Naunyn Schmiedebergs Arch Pharmacol. 320:266–271. 1982. View Article : Google Scholar : PubMed/NCBI

70 

Dang SS, Wang BF, Cheng YA, Song P, Liu ZG and Li ZF: Inhibitory effects of saikosaponin-d on CCl4-induced hepatic fibrogenesis in rats. World J Gastroenterol. 13:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Chen MF, Huang CC, Liu PS, Chen CH and Shiu LY: Saikosaponin a and saikosaponin d inhibit proliferation and migratory activity of rat HSC-T6 cells. J Med Food. 16:793–800. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Chen MF, Huang SJ, Huang CC, Liu PS, Lin KI, Liu CW, Hsieh WC, Shiu LY and Chen CH: Saikosaponin d induces cell death through caspase-3-dependent, caspase-3-independent and mitochondrial pathways in mammalian hepatic stellate cells. BMC Cancer. 16:5322016. View Article : Google Scholar : PubMed/NCBI

73 

Chen Y, Que R, Zhang N, Lin L, Zhou M and Li Y: Saikosaponin-d alleviates hepatic fibrosis through regulating GPER1/autophagy signaling. Mol Biol Rep. 48:7853–7863. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Zeng Y, Zhou L, Wan Y, Fu T, Xu P, Zhang H and Guan Y: Effects of Saikosaponin D on apoptosis, autophagy, and morphological structure of intestinal cells of cajal with functional dyspepsia. Comb Chem High Throughput Screen. 27:1513–1522. 2023. View Article : Google Scholar : PubMed/NCBI

75 

Li P, Wu M, Xiong W, Li J, An Y, Ren J, Xie Y, Xue H, Yan D, Li M, et al: Saikosaponin-d ameliorates dextran sulfate sodium-induced colitis by suppressing NF-κB activation and modulating the gut microbiota in mice. Int Immunopharmacol. 81:1062882020. View Article : Google Scholar

76 

Habtezion A: Inflammation in acute and chronic pancreatitis. Curr Opin Gastroenterol. 31:395–399. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Cui LH, Li CX, Zhuo YZ, Yang L, Cui NQ and Zhang SK: Saikosaponin d ameliorates pancreatic fibrosis by inhibiting autophagy of pancreatic stellate cells via PI3K/Akt/mTOR pathway. Chem Biol Interact. 300:18–26. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Abe H, Orita M, Konishi H, Arichi S and Odashima S: Effects of saikosaponin-d on aminonucleoside nephrosis in rats. Eur J Pharmacol. 120:171–178. 1986. View Article : Google Scholar : PubMed/NCBI

79 

Zhao L, Zhang H, Bao J, Liu J and Ji Z: Saikosaponin-d protects renal tubular epithelial cell against high glucose induced injury through modulation of SIRT3. Int J Clin Exp Med. 8:6472–6481. 2015.PubMed/NCBI

80 

Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group: KDIGO 2024 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. 105(Suppl): S117–S314. 2024. View Article : Google Scholar

81 

Loren P, Lugones Y, Saavedra N, Saavedra K, Páez I, Rodriguez N, Moriel P and Salazar LA: MicroRNAs involved in intrinsic apoptotic pathway during Cisplatin-induced nephrotoxicity: Potential use of natural products against DDP-induced apoptosis. Biomolecules. 12:12062022. View Article : Google Scholar : PubMed/NCBI

82 

Ma X, Dang C, Kang H, Dai Z, Lin S, Guan H, Liu X, Wang X and Hui W: Saikosaponin-D reduces cisplatin-induced nephrotoxicity by repressing ROS-mediated activation of MAPK and NF-κB signalling pathways. Int Immunopharmacol. 28:399–408. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Tomino Y: Mechanisms and interventions in peritoneal fibrosis. Clin Exp Nephrol. 16:109–114. 2012. View Article : Google Scholar

84 

Ruiqi L, Ming P, Qihang S, Yangyang L, Junli C, Wei L, Chao G, Xinyue L, Kang Y and Hongtao Y: Saikosaponin D inhibits peritoneal fibrosis in rats with renal failure by regulation of TGFβ1/BMP7/Gremlin1/Smad pathway. Front Pharmacol. 12:6286712021. View Article : Google Scholar

85 

Sun Y, Zhang H, Zhang X, Wang W, Chen Y, Cai Z, Wang Q, Wang J and Shi Y: Promotion of astrocyte-neuron glutamate-glutamine shuttle by SCFA contributes to the alleviation of Alzheimer's disease. Redox Biol. 62:1026902023. View Article : Google Scholar : PubMed/NCBI

86 

Bai R, Guo J, Ye XY, Xie Y and Xie T: Oxidative stress: The core pathogenesis and mechanism of Alzheimer's disease. Ageing Res Rev. 77:1016192022. View Article : Google Scholar : PubMed/NCBI

87 

Du J, Song D, Li Y, Liu J, Huang X, Li B and Li L: Saikosaponin-D mitigates oxidation in SH-SY5Y cells stimulated by glutamate through activation of Nrf2 pathway: Involvement of PI3K. Neurotox Res. 40:230–240. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Qin T, Yuan Z, Yu J, Fu X, Deng X, Fu Q, Ma Z and Ma S: Saikosaponin-d impedes hippocampal neurogenesis and causes cognitive deficits by inhibiting the survival of neural stem/progenitor cells via neurotrophin receptor signaling in mice. Clin Transl Med. 10:e2432020. View Article : Google Scholar : PubMed/NCBI

89 

Smith K: Mental health: A world of depression. Nature. 515:1812014. View Article : Google Scholar : PubMed/NCBI

90 

Li HY, Zhao YH, Zeng MJ, Fang F, Li M, Qin TT, Ye LY, Li HW, Qu R and Ma SP: Saikosaponin D relieves unpredictable chronic mild stress induced depressive-like behavior in rats: Involvement of HPA axis and hippocampal neurogenesis. Psychopharmacology (Berl). 234:3385–3394. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Chao B, Huang S, Pan J, Zhang Y and Wang Y: Saikosaponin d downregulates microRNA-155 and upregulates FGF2 to improve depression-like behaviors in rats induced by unpredictable chronic mild stress by negatively regulating NF-κB. Brain Res Bull. 157:69–76. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Liu CY, Chen JB, Liu YY, Zhou XM, Zhang M, Jiang YM, Ma QY, Xue Z, Zhao ZY, Li XJ, et al: Saikosaponin D exerts antidepressant effect by regulating Homer1-mGluR5 and mTOR signaling in a rat model of chronic unpredictable mild stress. Chin Med. 17:602022. View Article : Google Scholar : PubMed/NCBI

93 

Su J, Pan YW, Wang SQ, Li XZ, Huang F and Ma SP: Saikosaponin-d attenuated lipopolysaccharide-induced depressive-like behaviors via inhibiting microglia activation and neuroinflammation. Int Immunopharmacol. 80:1061812020. View Article : Google Scholar : PubMed/NCBI

94 

Papatheodorou K, Banach M, Bekiari E, Rizzo M and Edmonds M: Complications of Diabetes 2017. J Diabetes Res. 2018:30861672018. View Article : Google Scholar : PubMed/NCBI

95 

Ralli M, Angeletti D, Fiore M, D'Aguanno V, Lambiase A, Artico M, de Vincentiis M and Greco A: Hashimoto's thyroiditis: An update on pathogenic mechanisms, diagnostic protocols, therapeutic strategies, and potential malignant transformation. Autoimmun Rev. 19:1026492020. View Article : Google Scholar : PubMed/NCBI

96 

Martel-Pelletier J, Barr AJ, Cicuttini FM, Conaghan PG, Cooper C, Goldring MB, Goldring SR, Jones G, Teichtahl AJ and Pelletier JP: Osteoarthritis. Nat Rev Dis Primers. 2:160722016. View Article : Google Scholar : PubMed/NCBI

97 

Jiang J, Meng Y, Hu S, Botchway BOA, Zhang Y and Liu X: Saikosaponin D: A potential therapeutic drug for osteoarthritis. J Tissue Eng Regen Med. 14:1175–1184. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Wu X, Zhao K, Fang X, Lu F, Cheng P, Song X, Zhang W, Yao C, Zhu J and Chen H: Saikosaponin D inhibited IL-1β induced ATDC 5 chondrocytes apoptosis in vitro and delayed articular cartilage degeneration in OA model mice in vivo. Front Pharmacol. 13:8459592022. View Article : Google Scholar

99 

Yan M, Zhang D and Yang M: Saikosaponin D alleviates inflammatory response of osteoarthritis and mediates autophagy via elevating microRNA-199-3p to target transcription Factor-4. J Orthop Surg Res. 19:1512024. View Article : Google Scholar : PubMed/NCBI

100 

Liu RY and Li JP: Saikosaponin-d inhibits proliferation of human undifferentiated thyroid carcinoma cells through induction of apoptosis and cell cycle arrest. Eur Rev Med Pharmacol Sci. 18:2435–2443. 2014.PubMed/NCBI

101 

Passaro A, Jänne PA, Mok T and Peters S: Overcoming therapy resistance in EGFR-mutant lung cancer. Nat Cancer. 2:377–391. 2021. View Article : Google Scholar

102 

Tang JC, Long F, Zhao J, Hang J, Ren YG, Chen JY and Mu B: The Effects and mechanisms by which Saikosaponin-D enhances the sensitivity of human non-small cell lung cancer cells to gefitinib. J Cancer. 10:6666–6672. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Dong J, Yuan L, Hu C, Cheng X and Qin JJ: Strategies to overcome cancer multidrug resistance (MDR) through targeting P-glycoprotein (ABCB1): An updated review. Pharmacol Ther. 249:1084882023. View Article : Google Scholar : PubMed/NCBI

104 

Li C, Guan X, Xue H, Wang P, Wang M and Gai X: Reversal of P-glycoprotein-mediated multidrug resistance is induced by saikosaponin D in breast cancer MCF-7/adriamycin cells. Pathol Res Pract. 213:848–853. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Li C, Xue HG, Feng LJ, Wang ML, Wang P and Gai XD: The effect of saikosaponin D on doxorubicin pharmacokinetics and its MDR reversal in MCF-7/adr cell xenografts. Eur Rev Med Pharmacol Sci. 21:4437–4445. 2017.PubMed/NCBI

106 

Fu R, Zhang L, Li Y, Li B, Ming Y, Li Z, Xing H and Chen J: Saikosaponin D inhibits autophagosome-lysosome fusion and induces autophagy-independent apoptosis in MDA-MB-231 breast cancer cells. Mol Med Rep. 22:1026–1034. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Yang T, Li X, Wang X, Meng X, Zhang Z, Zhao M and Su R: Combination of histological and metabolomic assessments to evaluate the potential pharmacological efficacy of saikosaponin D. J Pharm Biomed Anal. 242:1160012024. View Article : Google Scholar : PubMed/NCBI

108 

Wen N, Cai Y, Li F, Ye H, Tang W, Song P and Cheng N: The clinical management of hepatocellular carcinoma worldwide: A concise review and comparison of current guidelines: 2022 update. Biosci Trends. 16:20–30. 2022. View Article : Google Scholar : PubMed/NCBI

109 

Jia X, Dang S, Cheng Y, Zhang X, Li M, Li Y and Li S: Effects of saikosaponin-d on syndecan-2, matrix metalloproteinases and tissue inhibitor of metalloproteinases-2 in rats with hepatocellular carcinoma. J Tradit Chin Med. 32:415–422. 2012. View Article : Google Scholar

110 

Chen Y, Chen HN, Wang K, Zhang L, Huang Z, Liu J, Zhang Z, Luo M, Lei Y, Peng Y, et al: Ketoconazole exacerbates mitophagy to induce apoptosis by downregulating cyclooxygenase-2 in hepatocellular carcinoma. J Hepatol. 70:66–77. 2019. View Article : Google Scholar

111 

Pang C, Miao H, Zuo Y, Guo N, Sun D and Li B: C/EBPβ enhances efficacy of sorafenib in hepatoblastoma. Cell Biol Int. 45:1897–1905. 2021. View Article : Google Scholar : PubMed/NCBI

112 

He SX, Luo JY, Zhao G, Xu JL, Wang YL, Fu H and Dong L: Effect of saikosaponins-d on cyclooxygenase-2 expression of human hepatocellular carcinoma cell line SMMC-7721. Zhonghua Gan Zang Bing Za Zhi. 14:712–714. 2006.In Chinese. PubMed/NCBI

113 

Lu XL, He SX, Ren MD, Wang YL, Zhang YX and Liu EQ: Chemopreventive effect of saikosaponin-d on diethylinitrosamine-induced hepatocarcinogenesis: Involvement of CCAAT/enhancer binding protein β and cyclooxygenase-2. Mol Med Rep. 5:637–644. 2012.

114 

He S, Lu G, Hou H, Zhao Z, Zhu Z, Lu X, Chen J and Wang Z: Saikosaponin-d suppresses the expression of cyclooxygenase-2 through the phospho-signal transducer and activator of transcription 3/hypoxia-inducible factor-1α pathway in hepatocellular carcinoma cells. Mol Med Rep. 10:2556–2562. 2014. View Article : Google Scholar : PubMed/NCBI

115 

Ren M, McGowan E, Li Y, Zhu X, Lu X, Zhu Z, Lin Y and He S: Saikosaponin-d Suppresses COX2 Through p-STAT3/C/EBPβ signaling pathway in liver cancer: A novel mechanism of action. Front Pharmacol. 10:6232019. View Article : Google Scholar

116 

Wang BF, Dai ZJ, Wang XJ, Bai MH, Lin S, Ma HB, Wang YL, Song LQ, Ma XL, Zan Y, et al: Saikosaponin-d increases the radiosensitivity of smmc-7721 hepatocellular carcinoma cells by adjusting the g0/g1 and g2/m checkpoints of the cell cycle. BMC Complement Altern Med. 13:2632013. View Article : Google Scholar : PubMed/NCBI

117 

Wang BF, Wang XJ, Kang HF, Bai MH, Guan HT, Wang ZW, Zan Y, Song LQ, Min WL, Lin S, et al: Saikosaponin-D enhances radiosensitivity of hepatoma cells under hypoxic conditions by inhibiting hypoxia-inducible factor-1α. Cell Physiol Biochem. 33:37–51. 2014. View Article : Google Scholar

118 

Ciepla P, Konitsiotis AD, Serwa RA, Masumoto N, Leong WP, Dallman MJ, Magee AI and Tate EW: New chemical probes targeting cholesterylation of Sonic Hedgehog in human cells and zebrafish. Chem Sci. 5:4249–4259. 2014. View Article : Google Scholar

119 

Zhang Z, Zhan X, Kim B and Wu J: A proteomic approach identifies SAFB-like transcription modulator (SLTM) as a bidirectional regulator of GLI family zinc finger transcription factors. J Biol Chem. 294:5549–5561. 2019. View Article : Google Scholar : PubMed/NCBI

120 

Hendriks IA and Vertegaal AC: A comprehensive compilation of SUMO proteomics. Nat Rev Mol Cell Biol. 17:581–595. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Zhang CY, Jiang ZM, Ma XF, Li Y, Liu XZ, Li LL, Wu WH and Wang T: Saikosaponin-d Inhibits the hepatoma cells and enhances chemosensitivity through SENP5-dependent inhibition of Gli1 SUMOylation under hypoxia. Front Pharmacol. 10:10392019. View Article : Google Scholar : PubMed/NCBI

122 

Tian YD, Lin S, Yang PT, Bai MH, Jin YY, Min WL, Ma HB and Wang BF: Saikosaponin-d increases the radiosensitivity of hepatoma cells by adjusting cell autophagy. J Cancer. 10:4947–4953. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Wang B, Min W, Lin S, Song L, Yang P, Ma Q and Guo J: Saikosaponin-d increases radiation-induced apoptosis of hepatoma cells by promoting autophagy via inhibiting mTOR phosphorylation. Int J Med Sci. 18:1465–1473. 2021. View Article : Google Scholar : PubMed/NCBI

124 

Lv Y, Hou X, Zhang Q, Li R, Xu L, Chen Y, Tian Y, Sun R, Zhang Z and Xu F: Untargeted metabolomics study of the in vitro anti-hepatoma effect of saikosaponin d in combination with NRP-1 knockdown. Molecules. 24:14232019. View Article : Google Scholar : PubMed/NCBI

125 

Li H, Tang Y, Wang Y, Wei W, Yin C and Tang F: Effects of Saikosaponin D on CYP1A2 and CYP2D6 in HepaRG cells. Drug Des Devel Ther. 14:5251–5258. 2020. View Article : Google Scholar : PubMed/NCBI

126 

Li H, Tang Y, Wei W, Yin C and Tang F: Effects of saikosaponin-d on CYP3A4 in HepaRG cell and protein-ligand docking study. Basic Clin Pharmacol Toxicol. 128:661–668. 2021. View Article : Google Scholar

127 

Edeline J, Bridgewater J, Campillo-Gimenez B, Neveu E, Phelip JM, Neuzillet C, Boudjema K, Rolland Y, Valle JW, Garin E, et al: Chemotherapy with or without selective internal radiation therapy for intrahepatic cholangiocarcinoma: Data from clinical trials. Hepatology. 79:96–106. 2024. View Article : Google Scholar

128 

Kelley RK, Bridgewater J, Gores GJ and Zhu AX: Systemic therapies for intrahepatic cholangiocarcinoma. J Hepatol. 72:353–363. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Kanno N, Lesage G, Phinizy JL, Glaser S, Francis H and Alpini G: Stimulation of alpha2-adrenergic receptor inhibits cholangiocarcinoma growth through modulation of Raf-1 and B-raf activities. Hepatology. 35:1329–1340. 2002. View Article : Google Scholar : PubMed/NCBI

130 

He H, Guo J, Hu Y, Zhang H, Li X, Zhang J and Jin S: Saikosaponin D reverses epinephrine- and norepinephrine-induced gemcitabine resistance in intrahepatic cholangiocarcinoma by downregulating ADRB2/glycolysis signaling. Acta Biochim Biophys Sin (Shanghai). 55:1404–1414. 2023.PubMed/NCBI

131 

Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Ning N, Li X, Nan Y, Chen G, Huang S, Du Y, Gu Q, Li W and Yuan L: Molecular mechanism of Saikosaponin-d in the treatment of gastric cancer based on network pharmacology and in vitro experimental verification. Naunyn Schmiedebergs Arch Pharmacol. 397:8943–8959. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Klein AP: Pancreatic cancer epidemiology: Understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 18:493–502. 2021. View Article : Google Scholar : PubMed/NCBI

134 

Xu X, Cui L, Zhang L, Yang L, Zhuo Y and Li C: Saikosaponin d modulates the polarization of tumor-associated macrophages by deactivating the PI3K/AKT/mTOR pathway in murine models of pancreatic cancer. Int Immunopharmacol. 122:1105792023. View Article : Google Scholar : PubMed/NCBI

135 

Cai C, Zhang H, Ou Y, Jiang Y, Zhong D, Qi H and Dang Q: Saikosaponin-d suppresses cell growth in renal cell carcinoma through EGFR/p38 signaling pathway. Neoplasma. 64:518–525. 2017. View Article : Google Scholar : PubMed/NCBI

136 

Rizzo A, Santoni M, Mollica V, Fiorentino M, Brandi G and Massari F: Microbiota and prostate cancer. Semin Cancer Biol. 86:1058–1065. 2022. View Article : Google Scholar

137 

Yao M, Yang J, Cao L, Zhang L, Qu S and Gao H: Saikosaponin-d inhibits proliferation of DU145 human prostate cancer cells by inducing apoptosis and arresting the cell cycle at G0/G1 phase. Mol Med Rep. 10:365–372. 2014. View Article : Google Scholar : PubMed/NCBI

138 

Zhong D, Zhang HJ, Jiang YD, Wu P, Qi H, Cai C, Zheng SB and Dang Q: Saikosaponin-d: A potential chemotherapeutics in castration resistant prostate cancer by suppressing cancer metastases and cancer stem cell phenotypes. Biochem Biophys Res Commun. 474:722–729. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Ghosh S: Cisplatin: The first metal based anticancer drug. Bioorg Chem. 88:1029252019. View Article : Google Scholar : PubMed/NCBI

140 

Tsuyoshi H, Wong VKW, Han Y, Orisaka M, Yoshida Y and Tsang BK: Saikosaponin-d, a calcium mobilizing agent, sensitizes chemoresistant ovarian cancer cells to cisplatin-induced apoptosis by facilitating mitochondrial fission and G2/M arrest. Oncotarget. 8:99825–99840. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Tang TT, Jiang L, Zhong Q, Ni ZJ, Thakur K, Khan MR and Wei ZJ: Saikosaponin D exerts cytotoxicity on human endometrial cancer ishikawa cells by inducing apoptosis and inhibiting metastasis through MAPK pathways. Food Chem Toxicol. 177:1138152023. View Article : Google Scholar : PubMed/NCBI

142 

Meltzer PS and Helman LJ: New horizons in the treatment of osteosarcoma. N Engl J Med. 385:2066–2076. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Zhao L, Li J, Sun ZB, Sun C, Yu ZH and Guo X: Saikosaponin D inhibits proliferation of human osteosarcoma cells via the p53 signaling pathway. Exp Ther Med. 17:488–494. 2019.PubMed/NCBI

144 

Gao T, Zhao P, Yu X, Cao S, Zhang B and Dai M: Use of Saikosaponin D and JNK inhibitor SP600125, alone or in combination, inhibits malignant properties of human osteosarcoma U2 cells. Am J Transl Res. 11:2070–2080. 2019.PubMed/NCBI

145 

Li Y, Cai T, Zhang W, Zhu W and Lv S: Effects of Saikosaponin D on apoptosis in human U87 glioblastoma cells. Mol Med Rep. 16:1459–1464. 2017. View Article : Google Scholar : PubMed/NCBI

146 

Liu G, Guan Y, Liu Y, Wang Y, Zhang J, Liu Y and Liu X: Saikosaponin D inducing apoptosis and autophagy through the activation of endoplasmic reticulum stress in glioblastoma. Biomed Res Int. 2022:54895532022. View Article : Google Scholar : PubMed/NCBI

147 

McKinnon C, Nandhabalan M, Murray SA and Plaha P: Glioblastoma: Clinical presentation, diagnosis, and management. BMJ. 374:n15602021. View Article : Google Scholar : PubMed/NCBI

148 

Liang J, Sun J, Liu A, Chen L, Ma X, Liu X and Zhang C: Saikosaponin D improves chemosensitivity of glioblastoma by reducing the its stemness maintenance. Biochem Biophys Rep. 32:1013422022.PubMed/NCBI

149 

Northcott PA, Robinson GW, Kratz CP, Mabbott DJ, Pomeroy SL, Clifford SC, Rutkowski S, Ellison DW, Malkin D, Taylor MD, et al: Medulloblastoma. Nat Rev Dis Primers. 5:112019. View Article : Google Scholar : PubMed/NCBI

150 

Luo J, Wang J, Yang J, Huang W, Liu J, Tan W and Xin H: Saikosaponin B1 and Saikosaponin D inhibit tumor growth in medulloblastoma allograft mice via inhibiting the Hedgehog signaling pathway. J Nat Med. 76:584–593. 2022. View Article : Google Scholar : PubMed/NCBI

151 

Shimony S, Stahl M and Stone RM: Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol. 98:502–526. 2023. View Article : Google Scholar : PubMed/NCBI

152 

Huang Y, Su R, Sheng Y, Dong L, Dong Z, Xu H, Ni T, Zhang ZS, Zhang T, Li C, et al: Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell. 35:677–691.e10. 2019. View Article : Google Scholar : PubMed/NCBI

153 

Li Z, Weng H, Su R, Weng X, Zuo Z, Li C, Huang H, Nachtergaele S, Dong L, Hu C, et al: FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell. 31:127–141. 2017. View Article : Google Scholar

154 

Sun K, Du Y, Hou Y, Zhao M, Li J, Du Y, Zhang L, Chen C, Yang H, Yan F, et al: Saikosaponin D exhibits anti-leukemic activity by targeting FTO/m6A signaling. Theranostics. 11:5831–5846. 2021. View Article : Google Scholar :

155 

Wang S, Long S and Wu W: Application of traditional chinese medicines as personalized therapy in human cancers. Am J Chin Med. 46:953–970. 2018. View Article : Google Scholar : PubMed/NCBI

156 

Luo Y, Wang CZ, Hesse-Fong J, Lin JG and Yuan CS: Application of Chinese medicine in acute and critical medical conditions. Am J Chin Med. 47:1223–1235. 2019. View Article : Google Scholar : PubMed/NCBI

157 

Xue J, Xu Z, Wang Q, Hou H, Wei L, Zhang J, Zhao X, Chen L, Ding F, Ma L, et al: Clinical practice guidelines for prevention and treatment of postoperative gastrointestinal disorder with Integrated Traditional Chinese and Western Medicine (2023). J Evid Based Med. 17:207–223. 2024. View Article : Google Scholar : PubMed/NCBI

158 

Zhang S, Liu M and Zhao L: Effects of different Chinese traditional exercises on mental health during the COVID-19 pandemic: A systematic review and meta-analysis. Front Public Health. 12:14200352024. View Article : Google Scholar : PubMed/NCBI

159 

Li X, Li X, Huang N, Liu R and Sun R: A comprehensive review and perspectives on pharmacology and toxicology of saikosaponins. Phytomedicine. 50:73–87. 2018. View Article : Google Scholar : PubMed/NCBI

160 

Chen MH, Gu YY, Zhang AL, Sze DM, Mo SL and May BH: Biological effects and mechanisms of matrine and other constituents of Sophora flavescens in colorectal cancer. Pharmacol Res. 171:1057782021. View Article : Google Scholar : PubMed/NCBI

161 

Shah MA, Abuzar SM, Ilyas K, Qadees I, Bilal M, Yousaf R, Kassim RMT, Rasul A, Saleem U, Alves MS, et al: Ginsenosides in cancer: Targeting cell cycle arrest and apoptosis. Chem Biol Interact. 382:1106342023. View Article : Google Scholar : PubMed/NCBI

162 

Alam P, Imran M, Gupta DK and Akhtar A: Formulation of transliposomal nanocarrier gel containing strychnine for the effective management of skin cancer. Gels. 9:8312023. View Article : Google Scholar : PubMed/NCBI

163 

Liu L, Yan J, Cao Y, Yan Y, Shen X, Yu B, Tao L and Wang S: Proliferation, migration and invasion of triple negative breast cancer cells are suppressed by berbamine via the PI3K/Akt/MDM2/p53 and PI3K/Akt/mTOR signaling pathways. Oncol Lett. 21:702021. View Article : Google Scholar

164 

Zhou P, Shi W, He XY, Du QY, Wang F and Guo J: Saikosaponin D: Review on the antitumour effects, toxicity and pharmacokinetics. Pharm Biol. 59:1480–1489. 2021. View Article : Google Scholar : PubMed/NCBI

165 

Zhang F, Chen L, Jin H, Shao J, Wu L, Lu Y and Zheng S: Activation of Fas death receptor pathway and Bid in hepatocytes is involved in saikosaponin D induction of hepatotoxicity. Environ Toxicol Pharmacol. 41:8–13. 2016. View Article : Google Scholar

166 

Farhan M: Green Tea Catechins: Nature's way of preventing and treating cancer. Int J Mol Sci. 23:107132022. View Article : Google Scholar : PubMed/NCBI

167 

Islam MR, Akash S, Rahman MM, Nowrin FT, Akter T, Shohag S, Rauf A, Aljohani ASM and Simal-Gandara J: Colon cancer and colorectal cancer: Prevention and treatment by potential natural products. Chem Biol Interact. 368:1101702022. View Article : Google Scholar : PubMed/NCBI

168 

Yang Z, Xiao T, Li Z, Zhang J and Chen S: Novel Chemicals derived from tadalafil exhibit PRMT5 inhibition and promising activities against breast cancer. Int J Mol Sci. 23:48062022. View Article : Google Scholar : PubMed/NCBI

169 

Zhao N, Wang W, Jiang H, Qiao Z, Sun S, Wei Y, Xie X, Li H, Bi X and Yang Z: Natural products and gastric cancer: cellular mechanisms and effects to change cancer progression. Anticancer Agents Med Chem. 23:1506–1518. 2023. View Article : Google Scholar : PubMed/NCBI

170 

Zhang GS, Hu PY, Li DX, He MZ, Rao XY, Luo XJ, Wang YS and Wang YR: Formulations, hemolytic and pharmacokinetic studies on saikosaponin a and saikosaponin d compound liposomes. Molecules. 20:5889–5907. 2015. View Article : Google Scholar : PubMed/NCBI

171 

Shimizu K, Amagaya S and Ogihara Y: Structural transformation of saikosaponins by gastric juice and intestinal flora. J Pharmacobiodyn. 8:718–725. 1985. View Article : Google Scholar : PubMed/NCBI

172 

Liu J, Xue Y, Sun J, Fu R, Ren S, Zhang Z and Song R: Pharmacokinetics and oral bioavailability studies of three saikogenins in rats using a validated UFLC-MS/MS method. J Chromatogr B Analyt Technol Biomed Life Sci. 1124:265–272. 2019. View Article : Google Scholar : PubMed/NCBI

173 

Ye HN, Liu XY and Qin BL: Research progress of integrated traditional Chinese and Western medicine in the treatment of advanced gastric cancer. World J Gastrointest Oncol. 15:69–75. 2023. View Article : Google Scholar : PubMed/NCBI

174 

Ma L, Wang B, Long Y and Li H: Effect of traditional Chinese medicine combined with Western therapy on primary hepatic carcinoma: A systematic review with meta-analysis. Front Med. 11:191–202. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2025
Volume 55 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gu S, Zheng Y, Chen C, Liu J, Wang Y, Wang J and Li Y: Research progress on the molecular mechanisms of Saikosaponin D in various diseases (Review). Int J Mol Med 55: 37, 2025.
APA
Gu, S., Zheng, Y., Chen, C., Liu, J., Wang, Y., Wang, J., & Li, Y. (2025). Research progress on the molecular mechanisms of Saikosaponin D in various diseases (Review). International Journal of Molecular Medicine, 55, 37. https://doi.org/10.3892/ijmm.2024.5478
MLA
Gu, S., Zheng, Y., Chen, C., Liu, J., Wang, Y., Wang, J., Li, Y."Research progress on the molecular mechanisms of Saikosaponin D in various diseases (Review)". International Journal of Molecular Medicine 55.3 (2025): 37.
Chicago
Gu, S., Zheng, Y., Chen, C., Liu, J., Wang, Y., Wang, J., Li, Y."Research progress on the molecular mechanisms of Saikosaponin D in various diseases (Review)". International Journal of Molecular Medicine 55, no. 3 (2025): 37. https://doi.org/10.3892/ijmm.2024.5478