Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Immunol., 21 November 2025

Sec. Nutritional Immunology

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1681358

This article is part of the Research TopicPolyphenols as Potent Modulators of Inflammation-Associated Non-Communicable DiseasesView all 4 articles

Salvianolic acid B: a promising cardioprotective agent

  • Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China

Salvia miltiorrhiza Bunge [Lamiaceae; Salviae miltiorrhizae radix et rhizoma] is a core herb in traditional Chinese medicine widely used for treating cardiovascular diseases (CVD). Its key bioactive compound, salvianolic acid B (SalB), has emerged as a promising therapeutic agent for CVD. Modern pharmacological studies have demonstrated that SalB exerts comprehensive cardioprotective effects through multiple mechanisms, including antioxidant and anti-inflammatory activities, induction of mitochondrial autophagy, enhancement of endothelial function, anti-fibrotic actions, and improvement of hemorheology. These properties underscore its significant value in both the prevention and treatment of CVD. However, current research on SalB in the context of CVD remains relatively fragmented. To address this gap, this review systematically consolidates existing research findings on SalB in the cardiovascular field, providing an in-depth analysis of its sources, pharmacological mechanisms, efficacy characteristics, compatibility strategies, and dosage form optimization. Furthermore, by integrating both preclinical and clinical data, this review comprehensively evaluates the safety and efficacy of SalB, aiming to offer theoretical support and clear research directions to facilitate the substantive transformation of this traditional bioactive compound into a modern CVD therapeutic drug.

1 Introduction

Salvia miltiorrhiza Bunge [Lamiaceae; Salviae miltiorrhizae radix et rhizoma] is widely used in clinical practice for its therapeutic effects, including coronary artery dilation, anti-myocardial ischemia, microcirculatory improvement, reduction of myocardial oxygen consumption, enhancement of cardiac function, and modulation of blood pressure, lipid levels, anticoagulation, and thrombosis, along with liver protection and immune regulation (13). Salvianolic acid B (SalB), the primary active compound in S. miltiorrhiza, has shown significant potential in the prevention and treatment of cardiovascular diseases (CVD) owing to its diverse pharmacological properties.

SalB demonstrates a broad spectrum of biological activities, including antioxidant, anti-inflammatory, anti-thrombotic, anti-apoptotic, anti-aging, and vascular and mitochondrial protection (46), establishing it as a central focus in CVD research. Numerous studies have emphasized SalB’s ability to neutralize reactive oxygen species, thereby reducing oxidative stress-induced cellular damage (79), which is critical for CVD prevention. Moreover, SalB inhibits platelet aggregation and thrombosis (1012), playing a key role in the prevention of acute cardiovascular events such as myocardial infarction and stroke. Its multi-target mechanism of action further supports its broad applicability in treating cardiovascular conditions. Additionally, its natural origin and low toxicity make SalB a promising candidate for long-term adjuvant therapy, positioning it as a potential cardioprotective agent.

Currently, S. miltiorrhiza injection and salvianolate are extensively used in clinical practice for the treatment of coronary heart disease, angina, cerebral infarction, and related liver, kidney, and lung disorders, with SalB as the primary active component. However, a monomeric preparation of SalB has yet to be developed, limiting the full utilization of its medicinal potential. This review systematically examines the sources, limitations, and pharmacological mechanisms of SalB, providing a comprehensive summary of recent advancements in CVD research, drug compatibility, and the latest developments in formulation design and clinical studies. The review aims to provide theoretical support and practical guidance for the clinical translation of SalB, addressing existing research fragmentation and promoting its transformation into a modern cardiovascular therapeutic agent. Furthermore, the in-depth analysis of SalB’s pharmacological characteristics and mechanisms of action presented here serves as a valuable template for future pharmaceutical chemistry research, paving the way for the development of novel structural drugs with enhanced efficacy, reduced toxicity, and improved pharmacokinetic properties.

2 The source and limitations of SalB

SalB (C36H30O16) is a monomeric compound extracted from the dried roots of S. miltiorrhiza Bunge (1315). As one of the most significant water-soluble components of S. miltiorrhiza, SalB is distinguished by its high concentration and potent bioactivity (1618). Maximizing the extraction of SalB from S. miltiorrhiza is therefore critical for enhancing its medicinal value. Current methods for SalB separation and purification include water extraction, ethanol precipitation, macroporous resin adsorption, ultrasonic extraction, high-speed countercurrent chromatography, subcritical water extraction, and supercritical CO2 extraction (1921). However, the molecular structure of SalB, formed by the condensation of three molecules of danshensu and one molecule of caffeic acid, contains ester bonds and a five-membered ring, which makes it prone to ester hydrolysis and ring-opening reactions (2224). Moreover, while the parent compound of SalB can be quantified in systemic circulation following oral administration, its absolute bioavailability is severely limited by factors such as intestinal instability, poor membrane permeability, and susceptibility to structural degradation, all of which restrict its systemic exposure (2527). These challenges pose safety risks in clinical settings and hinder the use of SalB in oral liquid formulations. Therefore, to address the challenge of its low oral bioavailability, several strategies can be employed. First, modifications to dosage formsecati as nanoparticles, injectable hydrogels, and core-shell nanofibersles, enhance SalB’s stability and delivery efficiency. Additionally, structural modifications to SalB itself represent an effective means to optimize its pharmacokinetic properties and improve bioavailability. Moreover, selecting high-quality S. miltiorrhiza varieties, can increase SalB extraction yields. Simultaneously, the entire extraction processionus raw material selection to extraction, purification, and storageation,y be systematically optimized, incorporating advanced technologies like supercritical CO2 extraction and subcritical water extraction. These innovations have significantly enhanced the therapeutic efficacy of SalB and paved the way for future drug development.

3 The pharmacological action pathway of SalB

SalB is a multi-target natural compound with remarkable pharmacological activity. By modulating various signaling pathways, it exerts biological effects such as anti-oxidation, anti-inflammation, anti-fibrosis, anti-cancer, cardiovascular protection, and neuroprotection. For instance, SalB regulates TGF-β1/Smad (28), MAPK/ERK (28), NF-κB/IκB (29), miR-152/PTCH1/Hh (30), CD36/PI3K/AKT (31), FGF19/FGFR4 (32), EZH2/PTEN/AKT (33), SIRT1-autophagy (34), PDGF-C/PDGFR-α (35), AMPK/FOXO1/miR-148a-3p (36), and HPSE/SDC1 (37) signaling pathways to exert anti-fibrotic effects (Figure 1A). It also regulates TRIM8/GPX1 (38), AKT/GSK3β (39), Nrf2/Keap1 (40), Nrf2/Nox4 (41), PI3K/AKT (42), JNK/MAPK (43), AKT/mTOR/4EBP1 (44), MKK3/6-p38MAPK/ATF2 (44), MEK/ERK1/2 (45), miR-19a/SIRT1 (46), and SIRT3/FOXO1/SOD2 (47) signaling pathways for antioxidant effects (Figure 1B) and modulates NF-κB/NLRP3 (48), LTR4/MyD88/NLRP3 (49), MAPKs/NF-κB (50), CD36/PI3K/AKT (31), IL2/STAT5 (51), TLR4/MyD88/TRAF6 (52), miR-486a-5p/FOXO1 (53), and AKT/mTOR (54) signaling pathways to exert anti-inflammatory effects (Figure 1C). In addition, SalB regulates AKT/mTOR (55), Hippo/YAP (56), PI3K/AKT/HIF-1α (57), TGF-β1/Smad (58), mortalin/RECK/STAT3 (59), and NDRG2/PTEN/PI3K/AKT (60) signaling pathways to promote anti-cancer effects (Figure 1C) and exerts neuroprotective effects by modulating CD40/NF-κB (61), IGF-1/AKT (62), ERK/CREB/BDNF (63), and miRNA-1/MLCK (64) signaling pathways (Figure 1D). Furthermore, SalB confers myocardial protection by regulating circTRRAP/miR-214-3p/SOX6 (65), ERK1/2/GATA4 (66), Nrf2/ARE (67), PI3K/AKT/HMGB1 (68), and TLR4/NF-κ-B/TNF-α (69) signaling pathways (Figure 1D). Notably, the pharmacological effects of SalB are not isolated but interact through intricate signaling pathways in various pathological states, forming an extensive regulatory network. For example, SalB exerts anti-fibrotic effects via the CD36/PI3K/AKT pathway while simultaneously exhibiting antioxidant, anti-inflammatory, and anti-cancer effects through the AKT/mTOR pathway, thereby establishing a multi-layered regulatory mechanism. Moreover, SalB synergistically inhibits oxidative stress and inflammation through the Nrf2/NLRP3 and NF-κB/NLRP3 pathways. This multi-pathway synergy significantly enhances SalB’s therapeutic potential, offering a unique advantage in treating complex diseases.

Figure 1
A flowchart illustrating various biological pathways influenced by Sal B is divided into four sections. Section A: Antifibrosis, shows interactions involving PI3K, FOXO1, SIRT1, and TGF-β. Section B: Antioxidant, includes SIRT3, AKT, and Nrf2 pathways. Section C: Anti-inflammatory, highlights pathways involving NF-κB, TLR4, and STAT5. Section D: Neuroprotection and Myocardial Protection, features AKT, IGF-1, TNFα, and GATA4 pathways. Each section visualizes Sal B’s role in cellular and molecular processes for therapeutic effects.

Figure 1. The pharmacological action pathway of SalB. (A) The mechanism of SalB in inhibiting fibrosis. (B) The mechanism by which SalB alleviates ROS-induced injury. (C) The mechanism of SalB’s anti-inflammatory and anti-tumor effects. (D) The mechanism by which SalB regulates neuronal function and myocardial injury.

4 The therapeutic effect of SalB on CVD

Various preparations derived from S. miltiorrhiza are currently widely used in clinical practice, including S. miltiorrhiza injection, oral liquids, tablets, compound granules, and S. miltiorrhiza-ligustrazine injection, all of which primarily contain active components from S. miltiorrhiza. Additionally, the S. miltiorrhiza extract tanshinone IIA has been developed into a monomeric preparation—sodium tanshinone IIA sulfonate injection—used clinically as an adjunct therapy for coronary heart disease, angina pectoris, and myocardial infarction (MI). Furthermore, preparations containing salvianolic acids, such as salvianolic acids for injection and salvianolate for injection, are employed to promote blood circulation and alleviate collateral obstruction, treating conditions like mild to moderate cerebral infarction and stable angina pectoris in coronary heart disease. However, there are currently no clinical applications for SalB, a primary monomer component of salvianolic acids, in the treatment of cardiovascular and cerebrovascular diseases, with related research still largely in the basic research phase.

4.1 Atherosclerosis

Atherosclerosis is closely linked to vascular endothelial dysfunction and inflammation (7072). SalB alleviates inflammation and thrombosis by inhibiting the activation of the TNF-α-induced NF-κB/NLRP3 pathway and platelet activation (48, 7375). Additionally, SalB protects blood vessels by reducing lipid accumulation in the vessel walls and inhibiting VEGF-induced vascular hyperpermeability, which prevents the uptake of 125I-LDL and thereby slows the progression of atherosclerosis (7678). Furthermore, SalB safeguards endothelial cells by inhibiting arginase activity and the activation of Piezo1 (79, 80). It also prevents NLRP3 inflammasome activation and pyroptosis triggered by endoplasmic reticulum (ER) stress through the suppression of the AMPK/FOXO4/KLF4 and Syndecan-4/Rac1/ATF2 signaling pathways, effectively protecting endothelial progenitor cells from damage (81). Additionally, in LPS-induced human aortic smooth muscle cells and ApoE-/- mice, SalB inhibited the expression of COX-2, MMP-2, and MMP-9 both in vitro and in vivo (82), suggesting that SalB may slow the progression of atherosclerosis through its anti-inflammatory effects and by decreasing vascular matrix degradation. LDL oxidation exacerbates inflammation within the vessel wall and promotes foam cell formation, accelerating atherosclerosis. As an antioxidant, SalB inhibits LDL oxidation and reduces the associated inflammatory response by blocking JAK/STAT1 activation induced by IFN-γ (83, 84). Furthermore, CD36, a high-affinity receptor for oxidized LDL (ox-LDL), is widely expressed in various cell types and plays a critical role in lipid metabolism, inflammation, and atherosclerosis (8587). Previous studies have demonstrated that SalB effectively inhibits the binding of CD36 to its ligand, thereby mitigating its detrimental effects in atherosclerosis and inflammation (88, 89).

SalB also exerts regulatory effects on dendritic cells, macrophages, and pericytes in the context of atherosclerosis. Additionally, SalB interferes with the maturation process of dendritic cells by activating PPARγ, thus reducing their ability to stimulate immune responses (90). It also effectively inhibits inflammation and autophagy dysfunction in LDLR-/- mice and RAW264.7 cells by regulating the MAPKs/NF-κB and AKT/mTOR signaling pathways, reducing the progression of atherosclerosis (50, 91). Additionally, by inhibiting the YAP/TAZ/JNK signaling pathway, SalB reduces the abnormal proliferation of pericytes and the release of inflammatory factors, offering a novel intervention strategy for preventing and treating atherosclerosis (92). The protective mechanism of SalB on atherosclerosis is summarized in Figure 2.

Figure 2
Complex biochemical pathway diagram illustrating the effects of SalB on cellular processes. It includes interactions with TNF-α/LPS, ox-LDL, ROS, and various signaling molecules like JNK, ERK, NF-KB, YAP/TAZ, and others leading to outcomes such as inflammation, apoptosis, and vasodilation. Key elements include cytokines, transcription factors, and stress pathways such as ER stress and mTOR. Various inhibitors and inducers are shown, affecting processes like oxidative stress and immune response.

Figure 2. The protective mechanism of SalB against atherosclerosis (AS). SalB ameliorates AS by modulating multiple signaling pathways, including ERK/JNK/MMP-9/2, ERK/JNK/COX-2/PGE2, NF-κB/ERK/AP-1/PAI-1, PPARγ/TLR4/MyD88/p38MAPK, JAK/STAT1, YAP/TAZ/JNK, Syndecan-4/Rac1/ATF2, AMPK/FOXO4/KLF2, AKT/mTOR, and HO-1/NO/iNOS axes.

In summary, SalB effectively mitigates multiple pathological aspects of atherosclerosis by modulating mechanisms in endothelial cells, macrophages, dendritic cells, pericytes, and immune responses. Its ability to inhibit inflammation, reduce lipid deposition, prevent vascular fibrosis, and enhance endothelial function provides a strong scientific foundation for its potential as a therapeutic agent in the treatment of atherosclerosis.

4.2 MI

In 2008, He et al. first reported that SalB exhibited myocardial protection in mice with MI, though these findings were later retracted (93). However, research into SalB’s protective effects against MI continued, with significant advancements in subsequent years. In 2009, Tan et al. demonstrated that pre-treatment of endogenous precursor cells (EPCs) with SalB, followed by transplantation alongside bone marrow mesenchymal stem cells (BMSCs) into the ischemic myocardium, resulted in notable improvements, suggesting SalB’s protective role in MI (94). In 2010, Jiang et al. identified SalB as a competitive inhibitor of matrix metalloproteinase-9 (MMP-9), which effectively reduced ischemic myocardial fibrosis, further supporting its myocardial protective effects (95). Zhao et al. (2012) confirmed Tan et al.’s findings, showing that SalB promoted the expansion of EPCs, improved the ischemic microenvironment, and enhanced BMSC survival and differentiation into cardiomyocytes (96). In 2014, Han et al. discovered that SalB’s protective effect in MI was linked to the promotion of mesenchymal stem cell (MSC) differentiation into endothelial cells, rather than cardiomyocytes (97). These studies collectively suggest that SalB’s myocardial protection is associated with the differentiation of EPCs, BMSCs, and MSCs. Further research has elucidated the mechanisms underlying SalB’s protection against MI, including its role in promoting nitric oxide (NO) production, autophagy, angiogenesis, and inhibiting the NLRP3 inflammasome, apoptosis, and ferroptosis (98103). The compatibility of traditional Chinese medicines has also demonstrated therapeutic advantages in clinical settings, with early studies exploring SalB’s potential in MI treatment. For example, Deng et al. found that combining SalB with ginsenoside Rg1 in a 2:5 ratio improved cardiac contractility in MI rats (104). To address the challenge of SalB’s low bioavailability in MI treatment, various drug carriers have been developed for more effective delivery. Qiu et al. created lipid-polymer mixed nanoparticles (LPNs) for co-delivery of SalB and panax notoginseng saponins (PNS), modified with arginyl-glycyl-aspartic acid (RGD) to form RGD-S/P-LPNs nanoparticles (105). These nanoparticles exhibited excellent serum stability and sustained drug release, significantly enhancing the therapeutic effect on MI in vivo (105). Shoba et al. developed a core-shell nanofiber system designed for phased delivery, releasing SalB from the core and magnesium L-ascorbic acid 2-phosphate (MAAP) from the shell, providing an effective vector for MI treatment (106). Chen et al. designed an elastin-mimicking peptide hydrogel (EMH) for delivering SalB-loaded dopamine nanoparticles (SalB-PDA), forming a SalB-PDA/EMH injectable peptide hydrogel with self-healing and slow-release properties (107). This hydrogel has been shown to inhibit ventricular remodeling and promote angiogenesis, offering a novel approach to MI treatment (107). These innovative drug carriers not only enhance SalB’s efficacy but also open new possibilities for the treatment of MI and related diseases. In summary, the core mechanisms of SalB’s protection in MI primarily involve modulating stem cell behavior, enhancing angiogenesis, and suppressing maladaptive inflammatory responses and cell death pathways. The above content is summarized in Figure 3.

Figure 3
Diagram illustrating the molecular mechanisms involving SalB, focusing on pathways related to ferroptosis, apoptosis, and cardiac function. It features molecules like ROS, MAPK, and SIRT1, with connections showing interactions and effects on processes like autophagy, eNOS activation, and inflammation regulation. The heart icon indicates the target of improved cardiac function.

Figure 3. Protective mechanism of SalB against myocardial infarction (MI). It can play a significant myocardial protective role in MI by regulating Nrf2/ferroptosis, MMP9/α-SMA, BNIP3/caspase3, PI3K/AKT, ATG5/autophagy signaling pathways.

4.3 Myocardial ischemia-reperfusion injury (MI/RI)

In 2011, Qiao et al. investigated the effects of the ethanol extract of S. miltiorrhiza Bunge on MI/RI and found that SalB, a key component of the extract, exhibited protective effects, particularly in preventing MI/R-induced oxidative damage in the myocardium (108). Subsequent studies by Qiao et al. and Gao et al. further confirmed SalB’s myocardial protective effect in MI/RI, showing significant improvements in cardiac function and tissue integrity (109, 110). Further investigations revealed that SalB’s protective mechanisms in MI/R were linked to enhancing cardiac contractility, scavenging reactive oxygen species, reducing inflammation, lipid peroxidation, apoptosis, and ferroptosis, and inhibiting autophagy (111113). Key regulatory mechanisms involved the activation of the PI3K/AKT and SIRT1/MAPK pathways, alongside inhibition of the circTRRAP/miR-214-3p/SOX6 and TRIM8/GPX1 axes (38, 65, 68, 114) (Figure 4). Additionally, Deng et al. demonstrated that combining SalB with ginsenoside Rg1 in a 2:5 ratio could protect against MI and improve MI/RI outcomes (104, 115). Overall, the core mechanisms by which SalB protects against MI/RI—focused on antioxidation, anti-inflammation, and regulation of various cell death pathways—overlap with its effects in MI, underscoring its broad role in counteracting ischemic damage. However, SalB’s role in MI/RI appears to emphasize mitigating reperfusion-specific injuries, such as the acute oxidative burst, distinguishing it from its action in MI alone.

Figure 4
Circular infographic illustrating the health benefits of SalB across various conditions. Sections include cerebrovascular disease, liver injury, cardiovascular disease, fibrosis-related diseases, cancer, and type 2 diabetes. Each segment lists specific benefits and compounds like Puerarin, Ginsenoside, Silymarin, and others in combination with SalB for treatment. Central focus on SalB, highlighted in yellow with an illustration of its source.

Figure 4. Protective mechanism of SalB on myocardial ischemia/reperfusion (MI/RI) injury. SalB confers cardioprotection against MI/RI by modulating the GPX4/ROS/ferroptosis, TRIM8/GPX1, SIRT3/ROS/NLRP3 inflammasome, SIRT1/ROS/MAPK, RagD/mTOR1, AMPK/PI3K/AKT/eNOS/NO, miR-30a/PI3K/AKT, and circTRRAP/miR-214-3p/SOX6A signaling axes, thereby attenuating oxidative stress, inflammation, apoptosis, and autophagic dysfunction while preserving myocardial function.

4.4 Cardiac hypertrophy

Studies on SalB’s therapeutic effects in cardiac hypertrophy remain limited, but some important findings have emerged. Liu et al. demonstrated that SalB effectively blocked the hypertrophic response in neonatal rat cardiomyocytes exposed to angiotensin II (116). The anti-hypertrophic effect of SalB was linked to its inhibition of poly (ADP-ribose) polymerase-1 (PARP-1), preventing NAD+ depletion in cells (116). This inhibition occurred through SalB’s suppression of the antioxidant functions of NOX2 and NOX4 (116). Further research by Yu et al. using a transverse aortic constriction (TAC)-induced cardiac hypertrophy model revealed that SalB provided superior protection against myocardial damage compared to metoprolol (66). Moreover, Ma et al. suggested that SalB may offer advantages over carvedilol in treating left ventricular hypertrophy, as SalB inhibits both the ERK signaling pathway and the β-adrenergic receptor, whereas carvedilol acts solely as a β-receptor blocker (117). These findings highlight SalB’s potential as a novel therapeutic agent for cardiac hypertrophy.

4.5 Diabetic cardiomyopathy (DCM)

DCM, a condition commonly associated with diabetes mellitus, is characterized by structural and functional abnormalities in the myocardium (118120). SalB exerts significant protective effects against DCM. Specifically, Li et al. found that SalB alleviates diabetic myocardial fibrosis by inhibiting insulin-like growth factor binding protein 3 (IGFBP3) and promoting angiogenesis (121). Furthermore, Luo et al. demonstrated that SalB inhibits the TGF-β1 signaling pathway by upregulating Smad7 expression, which reduces myocardial fibrosis and inflammatory cell infiltration, effectively alleviating diabetic myocardial damage (122). In summary, SalB exhibits substantial therapeutic potential in mitigating myocardial pathological damage caused by diabetes through multiple mechanisms, including anti-fibrosis, anti-inflammation, and pro-angiogenesis, supporting its use in treating DCM.

4.6 Septic cardiomyopathy (SCM)

SCM is a systemic inflammatory response triggered by infection that impairs heart function (123125). Due to its potent anti-inflammatory and antioxidant properties, SalB has shown protective effects against myocardial damage caused by sepsis. Chen et al. found that SalB enhances the mitochondrial unfolded protein response (UPRmt) by activating transcription factor 5 (ATF5), restoring protein folding balance in mitochondria and alleviating mitochondrial damage induced by sepsis (126). The core mechanism of SalB in SCM revolves around mitochondrial protection through ATF5-mediated UPRmt activation, a pathway distinct from its roles in fibrosis inhibition or stem cell regulation observed in conditions like MI, cardiac hypertrophy, or DCM. While SalB’s anti-inflammatory and antioxidant effects are common across various cardiac pathologies, its specific engagement with mitochondrial quality control in SCM underscores its capacity to address disease-specific mechanisms driven by severe infection and metabolic stress. This mechanism forms a theoretical foundation for the clinical application of SalB in treating SCM, offering new approaches for managing this condition.

4.7 Uremic cardiomyopathy (UC)

UC, a heart disease associated with renal failure, is often characterized by myocardial fibrosis, declining cardiac function, and elevated oxidative stress (127129). Ma et al. developed a rat model of UC to study the effects of SalB on cardiac function, ventricular hypertrophy, myocardial fibrosis, and inflammatory markers at various time points (130). Their findings indicated that SalB treatment improved cardiac function, reduced myocardial fibrosis, and alleviated inflammation in UC rats, thereby delaying disease progression. The cardioprotective effect of SalB in UC primarily results from its anti-fibrotic and anti-inflammatory properties, mechanisms that align with its actions in other cardiovascular conditions, such as DCM and cardiac hypertrophy.

4.8 Doxorubicin-induced cardiomyopathy (DIC)

Doxorubicin (DOX), a widely used chemotherapeutic agent, is known for its cardiotoxicity, which induces oxidative stress, inflammation, and ER stress in cardiomyocytes, ultimately leading to cardiomyocyte apoptosis and heart injury (131133). SalB mitigates ER stress by activating the PI3K/AKT signaling pathway, thereby inhibiting DOX-induced cardiomyocyte apoptosis (134). Additionally, SalB pretreatment prevents calcium overload and ER stress caused by DOX, with its protective mechanism involving the inhibition of TRP channel subfamily members 3 (TRPC3) and 6 (TRPC6) expression (135). These findings confirm that SalB exerts its protective effects against DOX-induced cardiotoxicity primarily by regulating ER stress and calcium homeostasis, highlighting its potential clinical value in treating DIC.

4.9 Cisplatin-induced cardiac injury

Cisplatin, a widely used chemotherapeutic agent, is commonly linked to severe cardiotoxicity and oxidative stress, resulting in cardiac dysfunction (136138). SalB effectively mitigates cisplatin-induced cardiac damage and oxidative stress, with its protective effect closely associated with the activation of the Nrf2 signaling pathway (67, 139). These findings establish Nrf2-mediated antioxidant pathway activation as the core mechanism through which SalB combats cisplatin-induced cardiotoxicity. This redox balance modulation differentiates its action from the calcium homeostasis regulation observed in DIC or the anti-fibrotic effects observed in DCM, highlighting SalB’s ability to selectively target specific pathological triggers.

5 Research on the compatibility and dosage forms of SalB

To overcome the limitations of SalB, numerous strategies, including drug compatibility and dosage form modifications, have been explored. In CVD, combining SalB with ascorbic acid has been shown to enhance the differentiation of MSCs into cardiomyocyte-like cells mediated by valproic acid and 5-azacytidine (140). This combination offers a promising approach for stem cell-based cardiac regeneration therapy. Moreover, combining SalB with astragaloside IV or ginsenoside Re significantly reduces arterial plaque area and lipid deposition (141, 142). Furthermore, the combination of SalB and ginsenoside Rg1 provides notable protection for cardiac function in patients with subacute MI, and it synergistically improves stroke treatment outcomes (143145). Importantly, studies have demonstrated that this combined regimen does not cause any abnormal changes in the tissue structure of the brain, heart, kidneys, liver, or lungs within seven days (146), ensuring the safety of the combination and providing strong support for its clinical application. Beyond CVD, the combined drug strategy of SalB has also been explored in treating cerebrovascular diseases, cancer, liver damage, kidney disease, and oral mucosal fibrosis (Figure 5).

Figure 5
Diagram illustrating the mechanisms by which SalB (salvianolic acid B) affects cardiac function. It shows the interaction with Nrf2 and pathways involving ferroptosis, ROS, and various proteins leading to improved cardiac function. The image includes references to myocardial fibrosis, inflammation, and angiogenesis. Treatments involve CD/GO-SalB/ADSC microsphere and SalB-PDA/EMH. Key elements: iron transport, cystine uptake, and apoptosis pathways. Essential components include CD, GO, ADSC, PDA, and EMH. The diagram emphasizes inhibition of ventricular remodeling and vasodilation.

Figure 5. The combination therapy of SalB. SalB can be combined with ascorbic acid, astragaloside IV, ginsenoside Re, ginsenoside Rg1, puerarin, and tetramethylpyrazine for cardiocerebrovascular diseases; with puerarin and chlorogenic acid for hepatic injury; with tanshinone IIA, SalA, chitosan, and triamcinolone acetonide for fibrosis-related disorders; with formononetin for cancer; and with ginsenoside Rb1 for type 2 diabetes.

In terms of dosage form research, innovative drug carrier systems have been developed in recent years to improve the oral bioavailability, targeted delivery, stability, and efficacy of SalB. Nanoparticles, as a key drug delivery platform, can enhance the therapeutic effects of SalB by improving stability, targeting, and absorption efficiency. For instance, Zhang et al. employed methoxypolyethylene glycol-chitosan (PCS)-derived nanoparticles to load SalB, enhancing its oral delivery capability and kidney-targeted distribution (147). Catechol-modified chitosan nanocarriers can also assemble SalB through coordination, enabling its targeted delivery to the kidneys and enhancing its anti-renal fibrosis effects (148). Moreover, the nanoparticle system developed by Grossi et al., capable of crossing the blood-brain barrier, efficiently delivers SalB to the central nervous system, improving its neuroprotective effects (149).

Liposomes have also been demonstrated to be effective as SalB drug delivery systems. Isacchi et al. developed SalB-loaded liposomes, which significantly improved the bioavailability and prolonged the pharmacodynamic effects of SalB (150), offering a new therapeutic option for treating neuropathic pain. Additionally, Shi et al. encapsulated SalB in liposomes modified with the cell-penetrating peptide TAT to enhance its skin repair potential (151).

In the development of composite scaffolds, Qin et al. used 3D bioprinting technology to create a SalB-sodium alginate-gelatin composite porous scaffold, which was applied to diabetic wounds. This scaffold was found to accelerate wound healing significantly (152). Similarly, Li et al. loaded SalB into chitosan microspheres, evenly distributing them in three dimensions and fixing them on the surface of porous hydroxyapatite (HA) scaffolds, thus creating a new type of bone tissue engineering scaffold (153). This composite scaffold demonstrated significant effects in promoting cell proliferation, adhesion, and differentiation, highlighting its potential for bone tissue regeneration. Additionally, polylactic acid (PLA) and graphene oxide (GO) have been utilized as composite scaffolds for SalB, enabling sustained release of SalB to promote bone tissue regeneration (154).

Based on the drug-loading capabilities of hydrogels, Chen et al. developed a SalB-polydopamine nanoparticle/elastin-mimic peptide hydrogel with suitable mechanical strength and self-healing properties (107). This hydrogel facilitates myocardial tissue repair and regeneration by enabling the long-term release of SalB directly to the infarct area. In another approach, injectable hydrogels composed of hyaluronic acid and gelatin were designed to deliver SalB and vascular endothelial growth factor, promoting synergistic brain tissue repair (155).

Bioactive glass scaffolds have also been explored for SalB delivery. Wu et al. loaded SalB into a mesoporous bioactive glass scaffold (MBG) via physical adsorption, forming a SalB-MBG scaffold. This scaffold effectively sustained SalB release and significantly promoted new bone formation and angiogenesis in bone defect sites (156). Kan et al. developed SalB microporous osmotic pump controlled-release pellets (157), which exhibited excellent in vitro drug release performance and favorable in vivo pharmacokinetic properties, making them a promising option for treating CVD.

In conclusion, nanoparticles and liposomes improve the delivery efficiency and therapeutic efficacy of SalB by enhancing drug stability, targeting, and absorption. Composite scaffolds and hydrogels offer unique advantages for tissue repair and regeneration, thanks to their customizable and sustainable release properties. Additionally, innovative carrier materials, such as mesoporous bioactive glass scaffolds and controlled-release pellets, have expanded the potential applications of SalB, particularly in regenerative medicine and the treatment of CVD.

6 Preclinical and clinical research status

Preclinical studies have shown that SalB provides significant protective effects across multiple organs, including the heart, liver (158160), brain (161163), kidneys (164166), and lungs (167169), indicating its broad therapeutic potential for treating cardiovascular, liver, renal, neurodegenerative, and pulmonary diseases. Notably, SalB’s cardiovascular protective effects are strongly supported by extensive preclinical animal data, with some indications advancing to clinical trials (Table 1). For example, intravenous administration of SalB for angina pectoris and coronary artery disease has entered Phase I clinical trials (CTR20192236). Cheng et al. assessed the safety, tolerability, and pharmacokinetic profile of SalB in healthy subjects (170), revealing no serious adverse events across all dose groups, with favorable safety and tolerability (170), supporting its potential application in CVD. Additionally, SalB has shown promising results in other clinical settings. A study of 42 patients with oral submucous fibrosis reported that combining triamcinolone acetonide with SalB significantly improved mouth opening and reduced burning sensations, with no adverse effects observed (171). In another clinical trial involving 60 patients with chronic hepatitis B-related liver fibrosis, six months of oral SalB tablets significantly improved liver fibrosis markers without side effects (172). Furthermore, multiple patents covering SalB’s extraction methods and therapeutic strategies for CVD further validate its clinical and methodological value (Table 2). In conclusion, SalB holds substantial promise for clinical translation in cardiovascular therapeutics. With growing clinical evidence, it may become a safe and effective treatment option, opening new pathways for patient care.

Table 1
www.frontiersin.org

Table 1. Preclinical and clinical trial studies on the cardiovascular therapeutic effects of SalB.

Table 2
www.frontiersin.org

Table 2. The relevant patents of SalB in cardiovascular diseases.

7 Conclusions and prospects

SalB, a polyphenolic compound extracted from S. miltiorrhiza Bunge, exhibits remarkable cardiovascular protective potential. Its multi-target mechanism includes a range of biological effects such as antioxidant, anti-inflammatory, anti-fibrotic, anti-thrombotic, and anti-apoptotic activities, along with promoting angiogenesis, making it highly applicable for treating CVD such as MI, MI/R injury, DCM, drug-induced cardiomyopathy, and atherosclerosis. Notably, SalB monomer preparations (injection) have entered clinical trials for CVD, marking a shift from an “active ingredient” in traditional Chinese medicine formulations to a modern “single chemical drug,” signifying a critical advancement in the modernization of traditional Chinese medicine. Therefore, SalB is well-positioned to become a key clinical treatment for CVD. Given this potential, future research should focus on several key areas: In basic research, efforts should be directed toward developing personalized treatment strategies based on SalB’s multi-target mechanisms for various cardiovascular conditions. In applied research, optimizing drug carrier design and formulating new dosage forms is essential to improve SalB’s stability, bioavailability, and clinical efficacy, with the aim of advancing SalB monomer preparations toward market release. In clinical translation, large-scale, multicenter trials are necessary to validate SalB’s safety and efficacy while exploring potential combination therapies with other agents. In conclusion, with continued progress in basic research, drug development, and clinical translation, SalB—an agent with multiple mechanisms of action targeting diverse pathological processes—has the potential to become a mainstream clinical drug for CVD in the form of monomer preparations.

Author contributions

YS: Writing – original draft, Conceptualization. LZ: Validation, Writing – review & editing. SJ: Validation, Writing – review & editing. FS: Conceptualization, Validation, Writing – review & editing. XL: Conceptualization, Validation, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This work was supported by the National Natural Science Foundation of China (82400429), Natural Science Foundation of Shandong Province (ZR2023QH007), Natural Science Foundation of Qingdao Municipality (23-2-1-135-zyyd-jch), and Qingdao Key Medical and Health Discipline Project-Clinical Pharmacy (2025-2027-18).

Acknowledgments

We acknowledge the use of Figdraw in creating Figures 13, 5. We thank Bullet Edits Limited for the linguistic editing and proofreading of the manuscript.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

MAPK, Mitogen-activated protein kinase; ERK, Extracellular signal-regulated kinase; PTCH1, Patched1; Hh, Hedgehog; CD36, Cluster of differentiation 36; PI3K/AKT, Phosphatidylinositol 3-kinases/protein kinase B; FGF19, Fibroblast growth factor 19; FGFR4, Fibroblast growth factor receptor 4; EZH2, Enhancer of zeste homolog 2; PTEN, Phosphatase and tensin homolog; SIRT1, Sirtuin 1; PDGF-C, Platelet-derived growth factor C; PDGFR-α, Platelet-derived growth factor receptor alpha; AMPK, AMP-activated protein kinase; FOXO1, Factor Forkhead Box O1; HPSE, Heparinase; SDC1, Syndecan 1; TRIM8, Tripartite motif containing protein 8; GPX1, Glutathione peroxidase 1; GSK3β, Glycogen synthase kinase 3beta; Nrf2, Nuclear factor E2-related factor 2; Keap1, Kelch-like ECH-associated protein 1; Nox4, NADPH oxidase 4; JNK, C-Jun N-terminal kinase; 4EBP1, 4E-binding protein 1; MKK3/6, Mitogen-activated protein kinase 3/6; ATF2, Activating transcription factor 2; MEK, Mitogen-activated protein kinase; ERK1/2, Extracellular regulated kinase 1/2; SOD2, Superoxide dismutase 2; TLR4, Toll-like receptor 4; MyD88, Myeloid differentiation primary response 88; NLRP3, NOD-like receptor protein 3; TRAF-6, Tumor necrosis factor receptor associated factor 6; RECK, REversion-inducing-Cysteine-rich protein with Kazal motifs; NDRG2, N-myc downstream-regulated gene 2; CREB, cAMP response element binding protein; BDNF, Brain-derived neurotrophic factor; SOX6, SRY-box transcription factor 6; GATA4, GATA binding protein 4; HMGB1, High mobility group box-1; Rac1, RAS-related C3 botulinum toxin substrate 1; BNIP3, Bcl-2/adenovirus E1B 19kDa interacting protein 3; ATG5, Autophagy related 5; RagD, RAG deficiency; AP-1, Activating protein-1; PAI-1, Plasminogen activator inhibitor-1.

References

1. Zhang MX, Huang XY, Song Y, Xu WL, Li YL, and Li C. Astragalus propinquus schischkin and Salvia miltiorrhiza bunge promote angiogenesis to treat myocardial ischemia via Ang-1/Tie-2/FAK pathway. Front Pharmacol. (2022) 13:1103557. doi: 10.3389/fphar.2022.1103557

PubMed Abstract | Crossref Full Text | Google Scholar

2. Zhang GX, Zhang YY, Zhang XX, Wang PQ, Liu J, Liu Q, et al. Different network pharmacology mechanisms of Danshen-based Fangjis in the treatment of stable angina. Acta Pharmacol Sin. (2018) 39:952–60. doi: 10.1038/aps.2017.191

PubMed Abstract | Crossref Full Text | Google Scholar

3. Yuan T, Chen Y, Zhou X, Lin X, and Zhang Q. Effectiveness and safety of Danshen injection on heart failure: Protocol for a systematic review and meta-analysis. Med (Baltimore). (2019) 98:e15636. doi: 10.1097/MD.0000000000015636

PubMed Abstract | Crossref Full Text | Google Scholar

4. Zhao M, Mu F, Lin R, Gao K, Zhang W, Tao X, et al. Chinese medicine-derived salvianolic acid B for disease therapy: A scientometric study. Am J Chin Med. (2024) 52:1359–96. doi: 10.1142/S0192415X2450054X

PubMed Abstract | Crossref Full Text | Google Scholar

5. Sun JM, Liu YX, Liu YD, Ho CK, Tsai YT, Wen DS, et al. Salvianolic acid B protects against UVB-induced skin aging via activation of NRF2. Phytomedicine. (2024) 130:155676. doi: 10.1016/j.phymed.2024.155676

PubMed Abstract | Crossref Full Text | Google Scholar

6. Xiang J, Zhang C, Di T, Chen L, Zhao W, Wei L, et al. Salvianolic acid B alleviates diabetic endothelial and mitochondrial dysfunction by down-regulating apoptosis and mitophagy of endothelial cells. Bioengineered. (2022) 13:3486–502. doi: 10.1080/21655979.2022.2026552

PubMed Abstract | Crossref Full Text | Google Scholar

7. Liu J, Zhang Y, Qu D, Zhang H, Wang L, Lau CW, et al. Salvianolic acid B ameliorates vascular endothelial dysfunction through influencing a bone morphogenetic protein 4-ROS cycle in diabetic mice. Life Sci. (2021) 286:120039. doi: 10.1016/j.lfs.2021.120039

PubMed Abstract | Crossref Full Text | Google Scholar

8. Yang MC, You FL, Wang Z, Liu XN, and Wang YF. Salvianolic acid B improves the disruption of high glucose-mediated brain microvascular endothelial cells via the ROS/HIF-1alpha/VEGF and miR-200b/VEGF signaling pathways. Neurosci Lett. (2016) 630:233–40. doi: 10.1016/j.neulet.2016.08.005

PubMed Abstract | Crossref Full Text | Google Scholar

9. Wang QQ, Wang M, Li Y, Liu YH, and Sun LQ. Attenuation of oxidative stress-induced cell apoptosis and pyroptosis in RSC96 cells by salvianolic acid B. Chin J Integr Med. (2022) 28:243–8. doi: 10.1007/s11655-021-3507-2

PubMed Abstract | Crossref Full Text | Google Scholar

10. Li M, Zhao C, Wong RN, Goto S, Wang Z, and Liao F. Inhibition of shear-induced platelet aggregation in rat by tetramethylpyrazine and salvianolic acid B. Clin Hemorheol Microcirc. (2004) 31:97–103.

PubMed Abstract | Google Scholar

11. Neves MAD, Ni TT, Mackeigan DT, Shoara AA, Lei X, Slavkovic S, et al. Salvianolic acid B inhibits thrombosis and directly blocks the thrombin catalytic site. Res Pract Thromb Haemost. (2024) 8:102443. doi: 10.1016/j.rpth.2024.102443

PubMed Abstract | Crossref Full Text | Google Scholar

12. Liu L, Li J, Zhang Y, Zhang S, Ye J, Wen Z, et al. Salvianolic acid B inhibits platelets as a P2Y12 antagonist and PDE inhibitor: evidence from clinic to laboratory. Thromb Res. (2014) 134:866–76. doi: 10.1016/j.thromres.2014.07.019

PubMed Abstract | Crossref Full Text | Google Scholar

13. Qin T, Rasul A, Sarfraz A, Sarfraz I, Hussain G, Anwar H, et al. Salvianolic acid A & B: potential cytotoxic polyphenols in battle against cancer via targeting multiple signaling pathways. Int J Biol Sci. (2019) 15:2256–64. doi: 10.7150/ijbs.37467

PubMed Abstract | Crossref Full Text | Google Scholar

14. Liang Q, Liu X, Peng X, Luo T, Su Y, Xu X, et al. Salvianolic acid B in fibrosis treatment: a comprehensive review. Front Pharmacol. (2024) 15:1442181. doi: 10.3389/fphar.2024.1442181

PubMed Abstract | Crossref Full Text | Google Scholar

15. Li J, Li R, Wu X, Zheng C, Shiu PH, Rangsinth P, et al. An update on the potential application of herbal medicine in promoting angiogenesis. Front Pharmacol. (2022) 13:928817. doi: 10.3389/fphar.2022.928817

PubMed Abstract | Crossref Full Text | Google Scholar

16. Guo SS and Wang ZG. Salvianolic acid B from Salvia miltiorrhiza bunge: A potential antitumor agent. Front Pharmacol. (2022) 13:1042745. doi: 10.3389/fphar.2022.1042745

PubMed Abstract | Crossref Full Text | Google Scholar

17. Zhang Y, Zhai W, Fan M, Wu J, and Wang C. Salvianolic Acid B Significantly Suppresses the Migration of Melanoma Cells via Direct Interaction with beta-Actin. Molecules. (2024) 29. doi: 10.3390/molecules29040906

PubMed Abstract | Crossref Full Text | Google Scholar

18. Tian LL, Wang XJ, Sun YN, Li CR, Xing YL, Zhao HB, et al. Salvianolic acid B, an antioxidant from Salvia miltiorrhiza, prevents 6-hydroxydopamine induced apoptosis in SH-SY5Y cells. Int J Biochem Cell Biol. (2008) 40:409–22. doi: 10.1016/j.biocel.2007.08.005

PubMed Abstract | Crossref Full Text | Google Scholar

19. Kan S, Li J, Huang W, Shao L, and Chen D. Microsphere resin chromatography combined with microbial biotransformation for the separation and purification of salvianolic acid B in aqueous extract of roots of Salvia multiorrihza Bunge. J Chromatogr A. (2009) 1216:3881–6. doi: 10.1016/j.chroma.2009.02.084

PubMed Abstract | Crossref Full Text | Google Scholar

20. Li C, Ma Y, Xu Y, Qiu R, Shen X, Huang L, et al. Ultrasonic-assisted nanofiltration separation recovering salvianolic acid B from ethanol wastewater. Ultrason Sonochem. (2024) 108:106967. doi: 10.1016/j.ultsonch.2024.106967

PubMed Abstract | Crossref Full Text | Google Scholar

21. Zhang M, Ignatova S, Liang Q, Wu Jun F, Sutherland I, Wang Y, et al. Rapid and high-throughput purification of salvianolic acid B from Salvia miltiorrhiza Bunge by high-performance counter-current chromatography. J Chromatogr A. (2009) 1216:3869–73. doi: 10.1016/j.chroma.2009.02.067

PubMed Abstract | Crossref Full Text | Google Scholar

22. Zhang X, Zou L, Li J, Xu B, Wu T, Fan H, et al. Salvianolic acid B and danshensu induce osteogenic differentiation of rat bone marrow stromal stem cells by upregulating the nitric oxide pathway. Exp Ther Med. (2017) 14:2779–88. doi: 10.3892/etm.2017.4914

PubMed Abstract | Crossref Full Text | Google Scholar

23. Lu P, Xing Y, Xue Z, Ma Z, Zhang B, Peng H, et al. Pharmacokinetics of salvianolic acid B, rosmarinic acid and Danshensu in rat after pulmonary administration of Salvia miltiorrhiza polyphenolic acid solution. BioMed Chromatogr. (2019) 33:e4561. doi: 10.1002/bmc.4561

PubMed Abstract | Crossref Full Text | Google Scholar

24. Habtemariam S. Molecular pharmacology of rosmarinic and salvianolic acids: potential seeds for alzheimer's and vascular dementia drugs. Int J Mol Sci. (2018) 19:458. doi: 10.3390/ijms19020458

PubMed Abstract | Crossref Full Text | Google Scholar

25. He G, Chen G, Liu W, Ye D, Liu X, Liang X, et al. Salvianolic acid B: A review of pharmacological effects, safety, combination therapy, new dosage forms, and novel drug delivery routes. Pharmaceutics. (2023) 15:2235. doi: 10.3390/pharmaceutics15092235

PubMed Abstract | Crossref Full Text | Google Scholar

26. Wu YT, Chen YF, Hsieh YJ, Jaw I, Shiao MS, and Tsai TH. Bioavailability of salvianolic acid B in conscious and freely moving rats. Int J Pharm. (2006) 326:25–31. doi: 10.1016/j.ijpharm.2006.07.003

PubMed Abstract | Crossref Full Text | Google Scholar

27. Gao DY, Han LM, Zhang LH, Fang XL, and Wang JX. Bioavailability of salvianolic acid B and effect on blood viscosities after oral administration of salvianolic acids in beagle dogs. Arch Pharm Res. (2009) 32:773–9. doi: 10.1007/s12272-009-1517-2

PubMed Abstract | Crossref Full Text | Google Scholar

28. Liu Q, Lu J, Lin J, Tang Y, Pu W, Shi X, et al. Salvianolic acid B attenuates experimental skin fibrosis of systemic sclerosis. BioMed Pharmacother. (2019) 110:546–53. doi: 10.1016/j.biopha.2018.12.016

PubMed Abstract | Crossref Full Text | Google Scholar

29. Wang R, Yu XY, Guo ZY, Wang YJ, Wu Y, and Yuan YF. Inhibitory effects of salvianolic acid B on CCl(4)-induced hepatic fibrosis through regulating NF-kappaB/IkappaBalpha signaling. J Ethnopharmacol. (2012) 144:592–8. doi: 10.1016/j.jep.2012.09.048

PubMed Abstract | Crossref Full Text | Google Scholar

30. Yu F, Lu Z, Chen B, Wu X, Dong P, and Zheng J. Salvianolic acid B-induced microRNA-152 inhibits liver fibrosis by attenuating DNMT1-mediated Patched1 methylation. J Cell Mol Med. (2015) 19:2617–32. doi: 10.1111/jcmm.12655

PubMed Abstract | Crossref Full Text | Google Scholar

31. Yan Y, Zhou M, Meng K, Zhou C, Jia X, Li X, et al. Salvianolic acid B attenuates inflammation and prevent pathologic fibrosis by inhibiting CD36-mediated activation of the PI3K-Akt signaling pathway in frozen shoulder. Front Pharmacol. (2023) 14:1230174. doi: 10.3389/fphar.2023.1230174

PubMed Abstract | Crossref Full Text | Google Scholar

32. Tian S, Chen M, Wang B, Han Y, Shang H, and Chen J. Salvianolic acid B blocks hepatic stellate cell activation via FGF19/FGFR4 signaling. Ann Hepatol. (2021) 20:100259. doi: 10.1016/j.aohep.2020.07.013

PubMed Abstract | Crossref Full Text | Google Scholar

33. Lin P, Qiu F, Wu M, Xu L, Huang D, Wang C, et al. Salvianolic acid B attenuates tubulointerstitial fibrosis by inhibiting EZH2 to regulate the PTEN/Akt pathway. Pharm Biol. (2023) 61:23–9. doi: 10.1080/13880209.2022.2148169

PubMed Abstract | Crossref Full Text | Google Scholar

34. He Y, Lu R, Wu J, Pang Y, Li J, Chen J, et al. Salvianolic acid B attenuates epithelial-mesenchymal transition in renal fibrosis rats through activating Sirt1-mediated autophagy. BioMed Pharmacother. (2020) 128:110241. doi: 10.1016/j.biopha.2020.110241

PubMed Abstract | Crossref Full Text | Google Scholar

35. Yao L, Zhao R, He S, Feng Q, Qiao Y, Wang P, et al. Effects of salvianolic acid A and salvianolic acid B in renal interstitial fibrosis via PDGF-C/PDGFR-alpha signaling pathway. Phytomedicine. (2022) 106:154414. doi: 10.1016/j.phymed.2022.154414

PubMed Abstract | Crossref Full Text | Google Scholar

36. Liu J, Sun Q, Sun X, Wang Q, Zou G, Wang D, et al. Therapeutic effects of salvianolic acid B on angiotensin II-induced atrial fibrosis by regulating atrium metabolism via targeting AMPK/foxO1/miR-148a-3p axis. J Cardiovasc Transl Res. (2023) 16:341–57. doi: 10.1007/s12265-022-10303-3

PubMed Abstract | Crossref Full Text | Google Scholar

37. Hu Y, Wang M, Pan Y, Li Q, and Xu L. Salvianolic acid B attenuates renal interstitial fibrosis by regulating the HPSE/SDC1 axis. Mol Med Rep. (2020) 22:1325–34. doi: 10.3892/mmr.2020.11229

PubMed Abstract | Crossref Full Text | Google Scholar

38. Lu B, Li J, Gui M, Yao L, Fan M, Zhou X, et al. Salvianolic acid B inhibits myocardial I/R-induced ROS generation and cell apoptosis by regulating the TRIM8/GPX1 pathway. Pharm Biol. (2022) 60:1458–68. doi: 10.1080/13880209.2022.2096644

PubMed Abstract | Crossref Full Text | Google Scholar

39. Wang D, Lu X, Wang E, Shi L, Ma C, and Tan X. Salvianolic acid B attenuates oxidative stress-induced injuries in enterocytes by activating Akt/GSK3beta signaling and preserving mitochondrial function. Eur J Pharmacol. (2021) 909:174408. doi: 10.1016/j.ejphar.2021.174408

PubMed Abstract | Crossref Full Text | Google Scholar

40. Xiao Z, Liu W, Mu YP, Zhang H, Wang XN, Zhao CQ, et al. Pharmacological effects of salvianolic acid B against oxidative damage. Front Pharmacol. (2020) 11:572373. doi: 10.3389/fphar.2020.572373

PubMed Abstract | Crossref Full Text | Google Scholar

41. Liu B, Cao B, Zhang D, Xiao N, Chen H, Li GQ, et al. Salvianolic acid B protects against paraquat-induced pulmonary injury by mediating Nrf2/Nox4 redox balance and TGF-beta1/Smad3 signaling. Toxicol Appl Pharmacol. (2016) 309:111–20. doi: 10.1016/j.taap.2016.08.004

PubMed Abstract | Crossref Full Text | Google Scholar

42. Wang M, Sun GB, Sun X, Wang HW, Meng XB, Qin M, et al. Cardioprotective effect of salvianolic acid B against arsenic trioxide-induced injury in cardiac H9c2 cells via the PI3K/Akt signal pathway. Toxicol Lett. (2013) 216:100–7. doi: 10.1016/j.toxlet.2012.11.023

PubMed Abstract | Crossref Full Text | Google Scholar

43. Xu X, Mao C, Zhang C, Zhang M, Gong J, and Wang X. Salvianolic Acid B Inhibits Ferroptosis and Apoptosis during Myocardial Ischemia/Reperfusion Injury via Decreasing the Ubiquitin-Proteasome Degradation of GPX4 and the ROS-JNK/MAPK Pathways. Molecules. (2023) 28:4117. doi: 10.3390/molecules28104117

PubMed Abstract | Crossref Full Text | Google Scholar

44. Tang Y, Jacobi A, Vater C, Zou X, and Stiehler M. Salvianolic acid B protects human endothelial progenitor cells against oxidative stress-mediated dysfunction by modulating Akt/mTOR/4EBP1, p38 MAPK/ATF2, and ERK1/2 signaling pathways. Biochem Pharmacol. (2014) 90:34–49. doi: 10.1016/j.bcp.2014.04.008

PubMed Abstract | Crossref Full Text | Google Scholar

45. Lu B, Ye Z, Deng Y, Wu H, and Feng J. MEK/ERK pathway mediates cytoprotection of salvianolic acid B against oxidative stress-induced apoptosis in rat bone marrow stem cells. Cell Biol Int. (2010) 34:1063–8. doi: 10.1042/CBI20090126

PubMed Abstract | Crossref Full Text | Google Scholar

46. Guo Y, Yang JH, Cao SD, Gao CX, He Y, Wang Y, et al. Effect of main ingredients of Danhong Injection against oxidative stress induced autophagy injury via miR-19a/SIRT1 pathway in endothelial cells. Phytomedicine. (2021) 83:153480. doi: 10.1016/j.phymed.2021.153480

PubMed Abstract | Crossref Full Text | Google Scholar

47. Wen F, Zhang S, Sun L, Qian M, and Xu H. Salvianolic acid B inhibits oxidative stress in glomerular mesangial cells alleviating diabetic nephropathy by regulating SIRT3/FOXO1 signaling. Kidney Blood Press Res. (2023) 48:738–51. doi: 10.1159/000534832

PubMed Abstract | Crossref Full Text | Google Scholar

48. Zhao Y, Shao C, Zhou H, Yu L, Bao Y, Mao Q, et al. Salvianolic acid B inhibits atherosclerosis and TNF-alpha-induced inflammation by regulating NF-kappaB/NLRP3 signaling pathway. Phytomedicine. (2023) 119:155002. doi: 10.1016/j.phymed.2023.155002

PubMed Abstract | Crossref Full Text | Google Scholar

49. Guan Y, Li L, Kan L, and Xie Q. Inhalation of salvianolic acid B prevents fine particulate matter-induced acute airway inflammation and oxidative stress by downregulating the LTR4/myD88/NLRP3 pathway. Oxid Med Cell Longev. (2022) 2022:5044356. doi: 10.1155/2022/5044356

PubMed Abstract | Crossref Full Text | Google Scholar

50. Zhang Y, Feng X, Du M, Ding J, and Liu P. Salvianolic acid B attenuates the inflammatory response in atherosclerosis by regulating MAPKs/ NF-kappaB signaling pathways in LDLR-/- mice and RAW264.7 cells. Int J Immunopathol Pharmacol. (2022) 36:3946320221079468. doi: 10.1177/03946320221079468

PubMed Abstract | Crossref Full Text | Google Scholar

51. Wang T, Wang J, Xu H, Yan H, Liu Y, Zhang N, et al. Salvianolic acid B alleviates autoimmunity in Treg-deficient mice via inhibiting IL2-STAT5 signaling. Phytother Res. (2024) 38:3825–36. doi: 10.1002/ptr.8222

PubMed Abstract | Crossref Full Text | Google Scholar

52. Wang Y, Chen G, Yu X, Li Y, Zhang L, He Z, et al. Salvianolic acid B ameliorates cerebral ischemia/reperfusion injury through inhibiting TLR4/myD88 signaling pathway. Inflammation. (2016) 39:1503–13. doi: 10.1007/s10753-016-0384-5

PubMed Abstract | Crossref Full Text | Google Scholar

53. Zhang ML, Zhang MN, Chen H, Wang X, Zhao K, Li X, et al. Salvianolic acid B alleviates high glucose-induced vascular smooth muscle cell inflammation by upregulating the miR-486a-5p expression. Mediators Inflammation. (2024) 2024:4121166. doi: 10.1155/2024/4121166

PubMed Abstract | Crossref Full Text | Google Scholar

54. Zou T, Gao S, Yu Z, Zhang F, Yao L, Xu M, et al. Salvianolic acid B inhibits RAW264.7 cell polarization towards the M1 phenotype by inhibiting NF-kappaB and Akt/mTOR pathway activation. Sci Rep. (2022) 12:13857. doi: 10.1038/s41598-022-18246-0

PubMed Abstract | Crossref Full Text | Google Scholar

55. Wang J, Ma Y, Guo M, Yang H, and Guan X. Salvianolic acid B suppresses EMT and apoptosis to lessen drug resistance through AKT/mTOR in gastric cancer cells. Cytotechnology. (2021) 73:49–61. doi: 10.1007/s10616-020-00441-4

PubMed Abstract | Crossref Full Text | Google Scholar

56. Xu W, Shi Z, Yu X, Xu Y, Chen Y, He Y, et al. Salvianolic acid B exerts an anti-hepatocellular carcinoma effect by regulating the Hippo/YAP pathway and promoting pSmad3L to pSmad3C simultaneously. Eur J Pharmacol. (2023) 939:175423. doi: 10.1016/j.ejphar.2022.175423

PubMed Abstract | Crossref Full Text | Google Scholar

57. Wei J, Wu J, Xu W, Nie H, Zhou R, Wang R, et al. Salvianolic acid B inhibits glycolysis in oral squamous cell carcinoma via targeting PI3K/AKT/HIF-1alpha signaling pathway. Cell Death Dis. (2018) 9:599. doi: 10.1038/s41419-018-0623-9

PubMed Abstract | Crossref Full Text | Google Scholar

58. Chen Y, Hu M, Wang S, Wang Q, Lu H, Wang F, et al. Nano-delivery of salvianolic acid B induces the quiescence of tumor-associated fibroblasts via interfering with TGF-beta1/Smad signaling to facilitate chemo- and immunotherapy in desmoplastic tumor. Int J Pharm. (2022) 623:121953. doi: 10.1016/j.ijpharm.2022.121953

PubMed Abstract | Crossref Full Text | Google Scholar

59. Teng M, Hu C, Yang B, Xiao W, Zhou Q, Li Y, et al. Salvianolic acid B targets mortalin and inhibits the migration and invasion of hepatocellular carcinoma via the RECK/STAT3 pathway. Cancer Cell Int. (2021) 21:654. doi: 10.1186/s12935-021-02367-z

PubMed Abstract | Crossref Full Text | Google Scholar

60. Yang Y, Huang L, Gao J, and Qian B. Salvianolic acid B inhibits the growth and metastasis of A549 lung cancer cells through the NDRG2/PTEN pathway by inducing oxidative stress. Med Oncol. (2024) 41:170. doi: 10.1007/s12032-024-02413-6

PubMed Abstract | Crossref Full Text | Google Scholar

61. Xu S, Zhong A, Ma H, Li D, Hu Y, Xu Y, et al. Neuroprotective effect of salvianolic acid B against cerebral ischemic injury in rats via the CD40/NF-kappaB pathway associated with suppression of platelets activation and neuroinflammation. Brain Res. (2017) 1661:37–48. doi: 10.1016/j.brainres.2017.02.011

PubMed Abstract | Crossref Full Text | Google Scholar

62. Ma X, Xu W, Zhang Z, Liu N, Yang J, Wang M, et al. Salvianolic acid B ameliorates cognitive deficits through IGF-1/akt pathway in rats with vascular dementia. Cell Physiol Biochem. (2017) 43:1381–91. doi: 10.1159/000481849

PubMed Abstract | Crossref Full Text | Google Scholar

63. Liu X, Hou Z, Han M, Chen K, Wang Y, Qing J, et al. Salvianolic acid B alleviates comorbid pain in depression induced by chronic restraint stress through inhibiting GABAergic neuron excitation via an ERK-CREB-BDNF axis-dependent mechanism. J Psychiatr Res. (2022) 151:205–16. doi: 10.1016/j.jpsychires.2022.04.014

PubMed Abstract | Crossref Full Text | Google Scholar

64. Xiong Y, Wang J, Chu H, Chen D, and Guo H. Salvianolic Acid B Restored Impaired Barrier Function via Downregulation of MLCK by microRNA-1 in Rat Colitis Model. Front Pharmacol. (2016) 7:134. doi: 10.3389/fphar.2016.00134

PubMed Abstract | Crossref Full Text | Google Scholar

65. Liu J, Dong W, Gao C, and Meng Y. Salvianolic acid B protects cardiomyocytes from ischemia/reperfusion injury by mediating circTRRAP/miR-214-3p/SOX6 axis. Int Heart J. (2022) 63:1176–86. doi: 10.1536/ihj.22-102

PubMed Abstract | Crossref Full Text | Google Scholar

66. Yu J, Chen R, Tan Y, Wu J, Qi J, Zhang M, et al. Salvianolic acid B alleviates heart failure by inactivating ERK1/2/GATA4 signaling pathway after pressure overload in mice. PloS One. (2016) 11:e0166560. doi: 10.1371/journal.pone.0166560

PubMed Abstract | Crossref Full Text | Google Scholar

67. Wu Y, Lin Z, Bao Y, Liu Y, and Zhang X. Salvianolic acid B ameliorated chemotherapeutic injury of cardiac myocytes through the nrf2/ARE signaling pathway. Discov Med. (2024) 36:415–23. doi: 10.24976/Discov.Med.202436181.39

PubMed Abstract | Crossref Full Text | Google Scholar

68. Liu H, Liu W, Qiu H, Zou D, Cai H, Chen Q, et al. Salvianolic acid B protects against myocardial ischaemia-reperfusion injury in rats via inhibiting high mobility group box 1 protein expression through the PI3K/Akt signalling pathway. Naunyn Schmiedebergs Arch Pharmacol. (2020) 393:1527–39. doi: 10.1007/s00210-019-01755-7

PubMed Abstract | Crossref Full Text | Google Scholar

69. Wang J, Zhang Y, Guo LL, Wu GJ, and Liu RH. Salvianolic acid B inhibits the TLR4-NFkappaB-TNFalpha pathway and attenuates neonatal rat cardiomyocyte injury induced by lipopolysaccharide. Chin J Integr Med. (2011) 17:775–9. doi: 10.1007/s11655-011-0877-x

PubMed Abstract | Crossref Full Text | Google Scholar

70. Wang YM, Chu TJ, Wan RT, Niu WP, Bian YF, and Li J. Quercetin ameliorates atherosclerosis by inhibiting inflammation of vascular endothelial cells via Piezo1 channels. Phytomedicine. (2024) 132:155865. doi: 10.1016/j.phymed.2024.155865

PubMed Abstract | Crossref Full Text | Google Scholar

71. Jia M, Li Q, Guo J, Shi W, Zhu L, Huang Y, et al. Deletion of BACH1 attenuates atherosclerosis by reducing endothelial inflammation. Circ Res. (2022) 130:1038–55. doi: 10.1161/CIRCRESAHA.121.319540

PubMed Abstract | Crossref Full Text | Google Scholar

72. Bu LL, Yuan HH, Xie LL, Guo MH, Liao DF, and Zheng XL. New dawn for atherosclerosis: vascular endothelial cell senescence and death. Int J Mol Sci. (2023) 24:15160. doi: 10.3390/ijms242015160

PubMed Abstract | Crossref Full Text | Google Scholar

73. Chen YH, Lin SJ, Ku HH, Shiao MS, Lin FY, Chen JW, et al. Salvianolic acid B attenuates VCAM-1 and ICAM-1 expression in TNF-alpha-treated human aortic endothelial cells. J Cell Biochem. (2001) 82:512–21. doi: 10.1002/jcb.1176

PubMed Abstract | Crossref Full Text | Google Scholar

74. Zhou Z, Liu Y, Miao AD, and Wang SQ. Salvianolic acid B attenuates plasminogen activator inhibitor type 1 production in TNF-alpha treated human umbilical vein endothelial cells. J Cell Biochem. (2005) 96:109–16. doi: 10.1002/jcb.20567

PubMed Abstract | Crossref Full Text | Google Scholar

75. Xu S, Zhong A, Bu X, Ma H, Li W, Xu X, et al. Salvianolic acid B inhibits platelets-mediated inflammatory response in vascular endothelial cells. Thromb Res. (2015) 135:137–45. doi: 10.1016/j.thromres.2014.10.034

PubMed Abstract | Crossref Full Text | Google Scholar

76. Qui Y, Rui YC, Zhang L, Li TJ, and Zhang WD. VEGF induced hyperpermeability in bovine aortic endothelial cell and inhibitory effect of salvianolic acid B. Acta Pharmacol Sin. (2001) 22:117–20.

PubMed Abstract | Google Scholar

77. Ding M, Ye TX, Zhao GR, Yuan YJ, and Guo ZX. Aqueous extract of Salvia miltiorrhiza attenuates increased endothelial permeability induced by tumor necrosis factor-alpha. Int Immunopharmacol. (2005) 5:1641–51. doi: 10.1016/j.intimp.2005.05.005

PubMed Abstract | Crossref Full Text | Google Scholar

78. Ba J, Peng H, Chen Y, and Gao Y. Effects and mechanism analysis of vascular endothelial growth factor and salvianolic acid B on 125I-low density lipoprotein permeability of the rabbit aortary endothelial cells. Cell Biochem Biophys. (2014) 70:1533–8. doi: 10.1007/s12013-014-0089-z

PubMed Abstract | Crossref Full Text | Google Scholar

79. Joe Y, Zheng M, Kim HJ, Kim S, Uddin MJ, Park C, et al. Salvianolic acid B exerts vasoprotective effects through the modulation of heme oxygenase-1 and arginase activities. J Pharmacol Exp Ther. (2012) 341:850–8. doi: 10.1124/jpet.111.190736

PubMed Abstract | Crossref Full Text | Google Scholar

80. Pan X, Wan R, Wang Y, Liu S, He Y, Deng B, et al. Inhibition of chemically and mechanically activated Piezo1 channels as a mechanism for ameliorating atherosclerosis with salvianolic acid B. Br J Pharmacol. (2022) 179:3778–814. doi: 10.1111/bph.15826

PubMed Abstract | Crossref Full Text | Google Scholar

81. Tang Y, Wa Q, Peng L, Zheng Y, Chen J, Chen X, et al. Salvianolic acid B suppresses ER stress-induced NLRP3 inflammasome and pyroptosis via the AMPK/foxO4 and syndecan-4/rac1 signaling pathways in human endothelial progenitor cells. Oxid Med Cell Longev. (2022) 2022:8332825. doi: 10.1155/2022/8332825

PubMed Abstract | Crossref Full Text | Google Scholar

82. Chen YL, Hu CS, Lin FY, Chen YH, Sheu LM, Ku HH, et al. Salvianolic acid B attenuates cyclooxygenase-2 expression in vitro in LPS-treated human aortic smooth muscle cells and in vivo in the apolipoprotein-E-deficient mouse aorta. J Cell Biochem. (2006) 98:618–31. doi: 10.1002/jcb.20793

PubMed Abstract | Crossref Full Text | Google Scholar

83. Yang TL, Lin FY, Chen YH, Chiu JJ, Shiao MS, Tsai CS, et al. Salvianolic acid B inhibits low-density lipoprotein oxidation and neointimal hyperplasia in endothelium-denuded hypercholesterolaemic rabbits. J Sci Food Agric. (2011) 91:134–41. doi: 10.1002/jsfa.4163

PubMed Abstract | Crossref Full Text | Google Scholar

84. Chen SC, Lin YL, Huang B, Wang DL, and Cheng JJ. Salvianolic acid B suppresses IFN-gamma-induced JAK/STAT1 activation in endothelial cells. Thromb Res. (2011) 128:560–4. doi: 10.1016/j.thromres.2011.08.032

PubMed Abstract | Crossref Full Text | Google Scholar

85. Demers A, Samami S, Lauzier B, Des Rosiers C, Ngo Sock ET, Ong H, et al. PCSK9 induces CD36 degradation and affects long-chain fatty acid uptake and triglyceride metabolism in adipocytes and in mouse liver. Arterioscler Thromb Vasc Biol. (2015) 35:2517–25. doi: 10.1161/ATVBAHA.115.306032

PubMed Abstract | Crossref Full Text | Google Scholar

86. Li Z, Zou X, Lu R, Wan X, Sun S, Wang S, et al. Arsenic trioxide alleviates atherosclerosis by inhibiting CD36-induced endocytosis and TLR4/NF-kappaB-induced inflammation in macrophage and ApoE(-/-) mice. Int Immunopharmacol. (2024) 128:111452. doi: 10.1016/j.intimp.2023.111452

PubMed Abstract | Crossref Full Text | Google Scholar

87. Silverstein RL. Inflammation, atherosclerosis, and arterial thrombosis: role of the scavenger receptor CD36. Cleve Clin J Med. (2009) 76:S27–30. doi: 10.3949/ccjm.76.s2.06

PubMed Abstract | Crossref Full Text | Google Scholar

88. Wang L, Bao Y, Yang Y, Wu Y, Chen X, Si S, et al. Discovery of antagonists for human scavenger receptor CD36 via an ELISA-like high-throughput screening assay. J Biomol Screen. (2010) 15:239–50. doi: 10.1177/1087057109359686

PubMed Abstract | Crossref Full Text | Google Scholar

89. Bao Y, Wang L, Xu Y, Yang Y, Wang L, Si S, et al. Salvianolic acid B inhibits macrophage uptake of modified low density lipoprotein (mLDL) in a scavenger receptor CD36-dependent manner. Atherosclerosis. (2012) 223:152–9. doi: 10.1016/j.atherosclerosis.2012.05.006

PubMed Abstract | Crossref Full Text | Google Scholar

90. Sun A, Liu H, Wang S, Shi D, Xu L, Cheng Y, et al. Salvianolic acid B suppresses maturation of human monocyte-derived dendritic cells by activating PPARgamma. Br J Pharmacol. (2011) 164:2042–53. doi: 10.1111/j.1476-5381.2011.01518.x

PubMed Abstract | Crossref Full Text | Google Scholar

91. Sun M, Ye Y, Huang Y, Yin W, Yu Z, and Wang S. Salvianolic acid B improves autophagic dysfunction and decreases the apoptosis of cholesterol crystal−induced macrophages via inhibiting the Akt/mTOR signaling pathway. Mol Med Rep. (2021) 24:763. doi: 10.3892/mmr.2021.12403

PubMed Abstract | Crossref Full Text | Google Scholar

92. Yang Y, Pei K, Zhang Q, Wang D, Feng H, Du Z, et al. Salvianolic acid B ameliorates atherosclerosis via inhibiting YAP/TAZ/JNK signaling pathway in endothelial cells and pericytes. Biochim Biophys Acta Mol Cell Biol Lipids. (2020) 1865:158779. doi: 10.1016/j.bbalip.2020.158779

PubMed Abstract | Crossref Full Text | Google Scholar

93. He H, Shi M, Yang X, Zeng X, Wu L, and Li L. Comparison of cardioprotective effects using salvianolic acid B and benazepril for the treatment of chronic myocardial infarction in rats. Naunyn Schmiedebergs Arch Pharmacol. (2008) 378:311–22. doi: 10.1007/s00210-008-0287-6

PubMed Abstract | Crossref Full Text | Google Scholar

94. Tan JZ, Li QW, and Nan YY. Effects of salvianolic acid B preconditioning endothelial progenitor cells on expressions of myocardial genes in bone mesenchymal stem cells at the early cell differentiation stage of rats. Zhongguo Zhong Xi Yi Jie He Za Zhi. (2009) 29:529–32.

PubMed Abstract | Google Scholar

95. Jiang B, Chen J, Xu L, Gao Z, Deng Y, Wang Y, et al. Salvianolic acid B functioned as a competitive inhibitor of matrix metalloproteinase-9 and efficiently prevented cardiac remodeling. BMC Pharmacol. (2010) 10:10. doi: 10.1186/1471-2210-10-10

PubMed Abstract | Crossref Full Text | Google Scholar

96. Zhao GF, Fan YC, and Jiang XJ. Effects of the proliferation state of the endothelial progenitor cells preconditioned with salvianolic acid B and bone marrow mesenchymal stem cells transplanted in acute myocardial infarction rats. Zhongguo Zhong Xi Yi Jie He Za Zhi. (2012) 32:671–5.

PubMed Abstract | Google Scholar

97. Guo HD, Cui GH, Tian JX, Lu PP, Zhu QC, Lv R, et al. Transplantation of salvianolic acid B pretreated mesenchymal stem cells improves cardiac function in rats with myocardial infarction through angiogenesis and paracrine mechanisms. Int J Cardiol. (2014) 177:538–42. doi: 10.1016/j.ijcard.2014.08.104

PubMed Abstract | Crossref Full Text | Google Scholar

98. Pan C, Lou L, Huo Y, Singh G, Chen M, Zhang D, et al. Salvianolic acid B and tanshinone IIA attenuate myocardial ischemia injury in mice by NO production through multiple pathways. Ther Adv Cardiovasc Dis. (2011) 5:99–111. doi: 10.1177/1753944710396538

PubMed Abstract | Crossref Full Text | Google Scholar

99. Lin C, Liu Z, Lu Y, Yao Y, Zhang Y, Ma Z, et al. Cardioprotective effect of Salvianolic acid B on acute myocardial infarction by promoting autophagy and neovascularization and inhibiting apoptosis. J Pharm Pharmacol. (2016) 68:941–52. doi: 10.1111/jphp.12567

PubMed Abstract | Crossref Full Text | Google Scholar

100. Xu L, Deng Y, Feng L, Li D, Chen X, Ma C, et al. Cardio-protection of salvianolic acid B through inhibition of apoptosis network. PloS One. (2011) 6:e24036. doi: 10.1371/journal.pone.0024036

PubMed Abstract | Crossref Full Text | Google Scholar

101. Chen Q, Xu Q, Zhu H, Wang J, Sun N, Bian H, et al. Salvianolic acid B promotes angiogenesis and inhibits cardiomyocyte apoptosis by regulating autophagy in myocardial ischemia. Chin Med. (2023) 18:155. doi: 10.1186/s13020-023-00859-w

PubMed Abstract | Crossref Full Text | Google Scholar

102. Li Q, Zuo Z, Pan Y, Zhang Q, Xu L, and Jiang B. Salvianolic acid B alleviates myocardial ischemia injury by suppressing NLRP3 inflammasome activation via SIRT1-AMPK-PGC-1alpha signaling pathway. Cardiovasc Toxicol. (2022) 22:842–57. doi: 10.1007/s12012-022-09760-8

PubMed Abstract | Crossref Full Text | Google Scholar

103. Shen Y, Shen X, Wang S, Zhang Y, Wang Y, Ding Y, et al. Protective effects of Salvianolic acid B on rat ferroptosis in myocardial infarction through upregulating the Nrf2 signaling pathway. Int Immunopharmacol. (2022) 112:109257. doi: 10.1016/j.intimp.2022.109257

PubMed Abstract | Crossref Full Text | Google Scholar

104. Deng Y, Zhang T, Teng F, Li D, Xu F, Cho K, et al. Ginsenoside Rg1 and Rb1, in combination with salvianolic acid B, play different roles in myocardial infarction in rats. J Chin Med Assoc. (2015) 78:114–20. doi: 10.1016/j.jcma.2014.10.001

PubMed Abstract | Crossref Full Text | Google Scholar

105. Qiu J, Cai G, Liu X, and Ma D. alpha(v)beta(3) integrin receptor specific peptide modified, salvianolic acid B and panax notoginsenoside loaded nanomedicine for the combination therapy of acute myocardial ischemia. BioMed Pharmacother. (2017) 96:1418–26. doi: 10.1016/j.biopha.2017.10.086

PubMed Abstract | Crossref Full Text | Google Scholar

106. Shoba E, Lakra R, Kiran MS, and Korrapati PS. Strategic design of cardiac mimetic core-shell nanofibrous scaffold impregnated with Salvianolic acid B and Magnesium l-ascorbic acid 2 phosphate for myoblast differentiation. Mater Sci Eng C Mater Biol Appl. (2018) 90:131–47. doi: 10.1016/j.msec.2018.04.056

PubMed Abstract | Crossref Full Text | Google Scholar

107. Chen R, Zhu C, Xu L, Gu Y, Ren S, Bai H, et al. An injectable peptide hydrogel with excellent self-healing ability to continuously release salvianolic acid B for myocardial infarction. Biomaterials. (2021) 274:120855. doi: 10.1016/j.biomaterials.2021.120855

PubMed Abstract | Crossref Full Text | Google Scholar

108. Qiao Z, Ma J, and Liu H. Evaluation of the antioxidant potential of Salvia miltiorrhiza ethanol extract in a rat model of ischemia-reperfusion injury. Molecules. (2011) 16:10002–12. doi: 10.3390/molecules161210002

PubMed Abstract | Crossref Full Text | Google Scholar

109. Qiao Z and Xu Y. Salvianolic acid B alleviating myocardium injury in ischemia reperfusion rats. Afr J Tradit Complement Altern Med. (2016) 13:157–61. doi: 10.21010/ajtcam.v13i4.20

PubMed Abstract | Crossref Full Text | Google Scholar

110. Gao F, Sun G, Ren X, Nie Y, Sun J, Qin M, et al. Protective effect of salvianolic acid B on isolated heart ischemia/reperfusion injury in rats. Zhongguo Zhong Yao Za Zhi. (2012) 37:358–61.

PubMed Abstract | Google Scholar

111. Xue L, Wu Z, Ji XP, Gao XQ, and Guo YH. Effect and mechanism of salvianolic acid B on the myocardial ischemia-reperfusion injury in rats. Asian Pac J Trop Med. (2014) 7:280–4. doi: 10.1016/S1995-7645(14)60038-9

PubMed Abstract | Crossref Full Text | Google Scholar

112. Zhao M, Li F, Jian Y, Wang X, Yang H, Wang J, et al. Salvianolic acid B regulates macrophage polarization in ischemic/reperfused hearts by inhibiting mTORC1-induced glycolysis. Eur J Pharmacol. (2020) 871:172916. doi: 10.1016/j.ejphar.2020.172916

PubMed Abstract | Crossref Full Text | Google Scholar

113. Li D, Wang J, Hou J, Fu J, Liu J, and Lin R. Salvianolic acid B induced upregulation of miR-30a protects cardiac myocytes from ischemia/reperfusion injury. BMC Complement Altern Med. (2016) 16:336. doi: 10.1186/s12906-016-1275-x

PubMed Abstract | Crossref Full Text | Google Scholar

114. Mao Q, Shao C, Zhou H, Yu L, Bao Y, Zhao Y, et al. Exploring the mechanism of salvianolic acid B against myocardial ischemia-reperfusion injury based on network pharmacology. Pharm (Basel). (2024) 17:309. doi: 10.3390/ph17030309

PubMed Abstract | Crossref Full Text | Google Scholar

115. Deng Y, Yang M, Xu F, Zhang Q, Zhao Q, Yu H, et al. Combined salvianolic acid B and ginsenoside rg1 exerts cardioprotection against ischemia/reperfusion injury in rats. PloS One. (2015) 10:e0135435. doi: 10.1371/journal.pone.0135435

PubMed Abstract | Crossref Full Text | Google Scholar

116. Liu M, Ye J, Gao S, Fang W, Li H, Geng B, et al. Salvianolic acid B protects cardiomyocytes from angiotensin II-induced hypertrophy via inhibition of PARP-1. Biochem Biophys Res Commun. (2014) 444:346–53. doi: 10.1016/j.bbrc.2014.01.045

PubMed Abstract | Crossref Full Text | Google Scholar

117. Ma D, Mandour AS, Yoshida T, Matsuura K, Shimada K, Kitpipatkun P, et al. Intraventricular pressure gradients change during the development of left ventricular hypertrophy: Effect of salvianolic acid B and beta-blocker. Ultrasound. (2021) 29:229–40. doi: 10.1177/1742271X20987584

PubMed Abstract | Crossref Full Text | Google Scholar

118. Dillmann WH. Diabetic cardiomyopathy. Circ Res. (2019) 124:1160–2. doi: 10.1161/CIRCRESAHA.118.314665

PubMed Abstract | Crossref Full Text | Google Scholar

119. Zhao X, Liu S, Wang X, Chen Y, Pang P, Yang Q, et al. Diabetic cardiomyopathy: Clinical phenotype and practice. Front Endocrinol (Lausanne). (2022) 13:1032268. doi: 10.3389/fendo.2022.1032268

PubMed Abstract | Crossref Full Text | Google Scholar

120. Jia G, Whaley-Connell A, and Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia. (2018) 61:21–8. doi: 10.1007/s00125-017-4390-4

PubMed Abstract | Crossref Full Text | Google Scholar

121. Li CL, Liu B, Wang ZY, Xie F, Qiao W, Cheng J, et al. Salvianolic acid B improves myocardial function in diabetic cardiomyopathy by suppressing IGFBP3. J Mol Cell Cardiol. (2020) 139:98–112. doi: 10.1016/j.yjmcc.2020.01.009

PubMed Abstract | Crossref Full Text | Google Scholar

122. Luo H, Fu L, Wang X, Yini X, Ling T, and Shen X. Salvianolic acid B ameliorates myocardial fibrosis in diabetic cardiomyopathy by deubiquitinating Smad7. Chin Med. (2023) 18:161. doi: 10.1186/s13020-023-00868-9

PubMed Abstract | Crossref Full Text | Google Scholar

123. Beesley SJ, Weber G, Sarge T, Nikravan S, Grissom CK, Lanspa MJ, et al. Septic cardiomyopathy. Crit Care Med. (2018) 46:625–34. doi: 10.1097/CCM.0000000000002851

PubMed Abstract | Crossref Full Text | Google Scholar

124. Wang R, Xu Y, Fang Y, Wang C, Xue Y, Wang F, et al. Pathogenetic mechanisms of septic cardiomyopathy. J Cell Physiol. (2022) 237:49–58. doi: 10.1002/jcp.30527

PubMed Abstract | Crossref Full Text | Google Scholar

125. Hollenberg SM and Singer M. Pathophysiology of sepsis-induced cardiomyopathy. Nat Rev Cardiol. (2021) 18:424–34. doi: 10.1038/s41569-020-00492-2

PubMed Abstract | Crossref Full Text | Google Scholar

126. Chen R, Zheng A, Wang Y, Guo L, Dou H, Lu L, et al. Salvianolic acid B improves mitochondrial dysfunction of septic cardiomyopathy via enhancing ATF5-mediated mitochondrial unfolded protein response. Toxicol Appl Pharmacol. (2024) 491:117072. doi: 10.1016/j.taap.2024.117072

PubMed Abstract | Crossref Full Text | Google Scholar

127. D'Agostino M, Mauro D, Zicarelli M, Carullo N, Greco M, Andreucci M, et al. miRNAs in uremic cardiomyopathy: A comprehensive review. Int J Mol Sci. (2023) 24:5425. doi: 10.3390/ijms24065425

PubMed Abstract | Crossref Full Text | Google Scholar

128. Hiraiwa H, Kasugai D, Okumura T, and Murohara T. Implications of uremic cardiomyopathy for the practicing clinician: an educational review. Heart Fail Rev. (2023) 28:1129–39. doi: 10.1007/s10741-023-10318-1

PubMed Abstract | Crossref Full Text | Google Scholar

129. Nguyen TD and Schulze PC. Cardiac metabolism in heart failure and implications for uremic cardiomyopathy. Circ Res. (2023) 132:1034–49. doi: 10.1161/CIRCRESAHA.123.321759

PubMed Abstract | Crossref Full Text | Google Scholar

130. Ma D, Mandour AS, Elfadadny A, Hendawy H, Yoshida T, El-Husseiny HM, et al. Changes in cardiac function during the development of uremic cardiomyopathy and the effect of salvianolic acid B administration in a rat model. Front Vet Sci. (2022) 9:905759. doi: 10.3389/fvets.2022.905759

PubMed Abstract | Crossref Full Text | Google Scholar

131. Shi S, Chen Y, Luo Z, Nie G, and Dai Y. Role of oxidative stress and inflammation-related signaling pathways in doxorubicin-induced cardiomyopathy. Cell Commun Signal. (2023) 21:61. doi: 10.1186/s12964-023-01077-5

PubMed Abstract | Crossref Full Text | Google Scholar

132. Glembotski CC, Bagchi S, and Blackwood EA. ER-specific autophagy or ER-phagy in cardiac myocytes protects the heart against doxorubicin-induced cardiotoxicity. JACC CardioOncol. (2023) 5:671–3. doi: 10.1016/j.jaccao.2023.08.004

PubMed Abstract | Crossref Full Text | Google Scholar

133. Malik A, Bagchi AK, Jassal DS, and Singal PK. Interleukin-10 mitigates doxorubicin-induced endoplasmic reticulum stress as well as cardiomyopathy. Biomedicines. (2022) 10:890. doi: 10.3390/biomedicines10040890

PubMed Abstract | Crossref Full Text | Google Scholar

134. Chen R, Sun G, Yang L, Wang J, and Sun X. Salvianolic acid B protects against doxorubicin induced cardiac dysfunction via inhibition of ER stress mediated cardiomyocyte apoptosis. Toxicol Res (Camb). (2016) 5:1335–45. doi: 10.1039/c6tx00111d

PubMed Abstract | Crossref Full Text | Google Scholar

135. Chen RC, Sun GB, Ye JX, Wang J, Zhang MD, and Sun XB. Salvianolic acid B attenuates doxorubicin-induced ER stress by inhibiting TRPC3 and TRPC6 mediated Ca(2+) overload in rat cardiomyocytes. Toxicol Lett. (2017) 276:21–30. doi: 10.1016/j.toxlet.2017.04.010

PubMed Abstract | Crossref Full Text | Google Scholar

136. Jia Y, Guo H, Cheng X, Zhang Y, Si M, Shi J, et al. Hesperidin protects against cisplatin-induced cardiotoxicity in mice by regulating the p62-Keap1-Nrf2 pathway. Food Funct. (2022) 13:4205–15. doi: 10.1039/d2fo00298a

PubMed Abstract | Crossref Full Text | Google Scholar

137. Xia J, Hu JN, Zhang RB, Liu W, Zhang H, Wang Z, et al. Icariin exhibits protective effects on cisplatin-induced cardiotoxicity via ROS-mediated oxidative stress injury in vivo and in vitro. Phytomedicine. (2022) 104:154331. doi: 10.1016/j.phymed.2022.154331

PubMed Abstract | Crossref Full Text | Google Scholar

138. Xing JJ, Mi XJ, Hou JG, Cai EB, Zheng SW, Wang SH, et al. Maltol mitigates cisplatin-evoked cardiotoxicity via inhibiting the PI3K/Akt signaling pathway in rodents in vivo and in vitro. Phytother Res. (2022) 36:1724–35. doi: 10.1002/ptr.7405

PubMed Abstract | Crossref Full Text | Google Scholar

139. Lin Z, Bao Y, Hong B, Wang Y, Zhang X, and Wu Y. Salvianolic acid B attenuated cisplatin-induced cardiac injury and oxidative stress via modulating Nrf2 signal pathway. J Toxicol Sci. (2021) 46:199–207. doi: 10.2131/jts.46.199

PubMed Abstract | Crossref Full Text | Google Scholar

140. Akbar N, Anum H, Razzaq SS, Salim A, Usman S, and Haneef K. Ascorbic acid and salvianolic acid B enhance the valproic acid and 5-azacytidinemediated cardiac differentiation of mesenchymal stem cells. Mol Biol Rep. (2023) 50:7371–80. doi: 10.1007/s11033-023-08634-8

PubMed Abstract | Crossref Full Text | Google Scholar

141. Kong XL, Lyu Q, Zhang YQ, Kang DF, Li C, Zhang L, et al. Effect of astragaloside IV and salvianolic acid B on antioxidant stress and vascular endothelial protection in the treatment of atherosclerosis based on metabonomics. Chin J Nat Med. (2022) 20:601–13. doi: 10.1016/S1875-5364(22)60186-9

PubMed Abstract | Crossref Full Text | Google Scholar

142. Yang K, Luo Y, Lu S, Hu R, Du Y, Liao P, et al. Salvianolic acid B and ginsenoside re synergistically protect against ox-LDL-induced endothelial apoptosis through the antioxidative and antiinflammatory mechanisms. Front Pharmacol. (2018) 9:662. doi: 10.3389/fphar.2018.00662

PubMed Abstract | Crossref Full Text | Google Scholar

143. Li Y, Wang L, Dong Z, Wang S, Qi L, Cho K, et al. Cardioprotection of salvianolic acid B and ginsenoside Rg1 combination on subacute myocardial infarction and the underlying mechanism. Phytomedicine. (2019) 57:255–61. doi: 10.1016/j.phymed.2018.12.040

PubMed Abstract | Crossref Full Text | Google Scholar

144. Shen H, Zhang Y, Shao Y, Chen S, Yin P, Liu X, et al. Synergism of salvianolic acid B and ginsenoside Rg1 magnifies the therapeutic potency against ischemic stroke. Neuroreport. (2024) 35:1041–51. doi: 10.1097/WNR.0000000000002099

PubMed Abstract | Crossref Full Text | Google Scholar

145. Fu Y, Xing R, Wang L, Yang L, and Jiang B. Neurovascular protection of salvianolic acid B and ginsenoside Rg1 combination against acute ischemic stroke in rats. Neuroreport. (2021) 32:1140–6. doi: 10.1097/WNR.0000000000001706

PubMed Abstract | Crossref Full Text | Google Scholar

146. Zhao Q, Yang M, Deng Y, Yu H, Wang L, Teng F, et al. The safety evaluation of salvianolic acid B and ginsenoside rg1 combination on mice. Int J Mol Sci. (2015) 16:29345–56. doi: 10.3390/ijms161226176

PubMed Abstract | Crossref Full Text | Google Scholar

147. Zhang Q, Li Y, Wang S, Gu D, Zhang C, Xu S, et al. Chitosan-based oral nanoparticles as an efficient platform for kidney-targeted drug delivery in the treatment of renal fibrosis. Int J Biol Macromol. (2024) 256:128315. doi: 10.1016/j.ijbiomac.2023.128315

PubMed Abstract | Crossref Full Text | Google Scholar

148. Li J, Zhang C, He W, Qiao H, Chen J, Wang K, et al. Coordination-driven assembly of catechol-modified chitosan for the kidney-specific delivery of salvianolic acid B to treat renal fibrosis. Biomater Sci. (2017) 6:179–88. doi: 10.1039/c7bm00811b

PubMed Abstract | Crossref Full Text | Google Scholar

149. Grossi C, Guccione C, Isacchi B, Bergonzi MC, Luccarini I, Casamenti F, et al. Development of blood-brain barrier permeable nanoparticles as potential carriers for salvianolic acid B to CNS. Planta Med. (2017) 83:382–91. doi: 10.1055/s-0042-101945

PubMed Abstract | Crossref Full Text | Google Scholar

150. Isacchi B, Fabbri V, Galeotti N, Bergonzi MC, Karioti A, Ghelardini C, et al. Salvianolic acid B and its liposomal formulations: anti-hyperalgesic activity in the treatment of neuropathic pain. Eur J Pharm Sci. (2011) 44:552–8. doi: 10.1016/j.ejps.2011.09.019

PubMed Abstract | Crossref Full Text | Google Scholar

151. Shi J, Guo S, Wu Y, Chen G, Lai J, and Xu X. Behaviour of cell penetrating peptide TAT-modified liposomes loaded with salvianolic acid B on the migration, proliferation, and survival of human skin fibroblasts. J Liposome Res. (2020) 30:93–106. doi: 10.1080/08982104.2019.1593451

PubMed Abstract | Crossref Full Text | Google Scholar

152. Lihao Q, Tingting L, Jiawei Z, Yifei B, Zheyu T, Jingyan L, et al. 3D bioprinting of Salvianolic acid B-sodium alginate-gelatin skin scaffolds promotes diabetic wound repair via antioxidant, anti-inflammatory, and proangiogenic effects. BioMed Pharmacother. (2024) 171:116168. doi: 10.1016/j.biopha.2024.116168

PubMed Abstract | Crossref Full Text | Google Scholar

153. Li J, Wang Q, Zhi W, Wang J, Feng B, Qu S, et al. Immobilization of salvianolic acid B-loaded chitosan microspheres distributed three-dimensionally and homogeneously on the porous surface of hydroxyapatite scaffolds. BioMed Mater. (2016) 11:55014. doi: 10.1088/1748-6041/11/5/055014

PubMed Abstract | Crossref Full Text | Google Scholar

154. Liu S, Xu Z, Hu J, Wu Z, and Zheng Y. Preparation and sustained-release properties of poly(lactic acid)/graphene oxide porous biomimetic composite scaffolds loaded with salvianolic acid B. RSC Adv. (2022) 12:28867–77. doi: 10.1039/d2ra05371c

PubMed Abstract | Crossref Full Text | Google Scholar

155. Zhou G, Cao Y, Yan Y, Xu H, Zhang X, Yan T, et al. Injectable hydrogels based on hyaluronic acid and gelatin combined with salvianolic acid B and vascular endothelial growth factor for treatment of traumatic brain injury in mice. Molecules. (2024) 29:1705. doi: 10.3390/molecules29081705

PubMed Abstract | Crossref Full Text | Google Scholar

156. Wu L, Wei Z, He S, Bi Y, Cao Y, and Wang W. Mesoporous bioactive glass scaffold delivers salvianolic acid B to promote bone regeneration in a rat cranial defect model. Curr Drug Delivery. (2021) 18:323–33. doi: 10.2174/1567201817666200916091253

PubMed Abstract | Crossref Full Text | Google Scholar

157. Kan SL, Li J, Liu JP, and Zhao Y. Preparation and IVIVC evaluation of salvianolic acid B micro-porous osmotic pump pellets. Drug Dev Ind Pharm. (2015) 41:476–81. doi: 10.3109/03639045.2013.879722

PubMed Abstract | Crossref Full Text | Google Scholar

158. Meng LC, Zheng JY, Qiu YH, Zheng L, Zheng JY, Liu YQ, et al. Salvianolic acid B ameliorates non-alcoholic fatty liver disease by inhibiting hepatic lipid accumulation and NLRP3 inflammasome in ob/ob mice. Int Immunopharmacol. (2022) 111:109099. doi: 10.1016/j.intimp.2022.109099

PubMed Abstract | Crossref Full Text | Google Scholar

159. Zhang N, Hu Y, Ding C, Zeng W, Shan W, Fan H, et al. Salvianolic acid B protects against chronic alcoholic liver injury via SIRT1-mediated inhibition of CRP and ChREBP in rats. Toxicol Lett. (2017) 267:1–10. doi: 10.1016/j.toxlet.2016.12.010

PubMed Abstract | Crossref Full Text | Google Scholar

160. Zeng W, Shan W, Gao L, Gao D, Hu Y, Wang G, et al. Inhibition of HMGB1 release via salvianolic acid B-mediated SIRT1 up-regulation protects rats against non-alcoholic fatty liver disease. Sci Rep. (2015) 5:16013. doi: 10.1038/srep16013

PubMed Abstract | Crossref Full Text | Google Scholar

161. Lv H, Wang L, Shen J, Hao S, Ming A, Wang X, et al. Salvianolic acid B attenuates apoptosis and inflammation via SIRT1 activation in experimental stroke rats. Brain Res Bull. (2015) 115:30–6. doi: 10.1016/j.brainresbull.2015.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

162. Chen T, Liu W, Chao X, Zhang L, Qu Y, Huo J, et al. Salvianolic acid B attenuates brain damage and inflammation after traumatic brain injury in mice. Brain Res Bull. (2011) 84:163–8. doi: 10.1016/j.brainresbull.2010.11.015

PubMed Abstract | Crossref Full Text | Google Scholar

163. Zhang X, Wu Q, Lu Y, Wan J, Dai H, Zhou X, et al. Cerebroprotection by salvianolic acid B after experimental subarachnoid hemorrhage occurs via Nrf2- and SIRT1-dependent pathways. Free Radic Biol Med. (2018) 124:504–16. doi: 10.1016/j.freeradbiomed.2018.06.035

PubMed Abstract | Crossref Full Text | Google Scholar

164. Tongqiang L, Shaopeng L, Xiaofang Y, Nana S, Xialian X, Jiachang H, et al. Salvianolic acid B prevents iodinated contrast media-induced acute renal injury in rats via the PI3K/akt/nrf2 pathway. Oxid Med Cell Longev. (2016) 2016:7079487. doi: 10.1155/2016/7079487

PubMed Abstract | Crossref Full Text | Google Scholar

165. Wang Y, Chang J, Qiao S, Yang Y, Yun C, Li Y, et al. Salvianolic acid B attenuates diabetic nephropathy through alleviating ADORA2B, NALP3 in flammasome, and NFkappaB activity. Can J Physiol Pharmacol. (2024) 102:318–30. doi: 10.1139/cjpp-2023-0089

PubMed Abstract | Crossref Full Text | Google Scholar

166. Chen J, Hu Q, Luo Y, Luo L, Lin H, Chen D, et al. Salvianolic acid B attenuates membranous nephropathy by activating renal autophagy via microRNA-145-5p/phosphatidylinositol 3-kinase/AKT pathway. Bioengineered. (2022) 13:13956–69. doi: 10.1080/21655979.2022.2083822

PubMed Abstract | Crossref Full Text | Google Scholar

167. Li Y, Chen R, Wu J, Xue X, Liu T, Peng G, et al. Salvianolic acid B protects against pulmonary fibrosis by attenuating stimulating protein 1-mediated macrophage and alveolar type 2 cell senescence. Phytother Res. (2024) 38:620–35. doi: 10.1002/ptr.8070

PubMed Abstract | Crossref Full Text | Google Scholar

168. Yang CW, Liu H, Li XD, Sui SG, and Liu YF. Salvianolic acid B protects against acute lung injury by decreasing TRPM6 and TRPM7 expressions in a rat model of sepsis. J Cell Biochem. (2018) 119:701–11. doi: 10.1002/jcb.26233

PubMed Abstract | Crossref Full Text | Google Scholar

169. Zhang T, Liu M, Gao Y, Li H, Song L, Hou H, et al. Salvianolic acid B inhalation solution enhances antifibrotic and anticoagulant effects in a rat model of pulmonary fibrosis. BioMed Pharmacother. (2021) 138:111475. doi: 10.1016/j.biopha.2021.111475

PubMed Abstract | Crossref Full Text | Google Scholar

170. Cheng J, Long J, Zhang J, Han L, Hu Y, Liu J, et al. Safety, tolerance, and pharmacokinetics of salvianolic acid B in healthy Chinese volunteers: A randomized, double-blind, placebo-controlled phase 1 clinical trial. Front Pharmacol. (2023) 14:1146309. doi: 10.3389/fphar.2023.1146309

PubMed Abstract | Crossref Full Text | Google Scholar

171. Jiang XW, Zhang Y, Yang SK, Zhang H, Lu K, and Sun GL. Efficacy of salvianolic acid B combined with triamcinolone acetonide in the treatment of oral submucous fibrosis. Oral Surg Oral Med Oral Pathol Oral Radiol. (2013) 115:339–44. doi: 10.1016/j.oooo.2012.10.006

PubMed Abstract | Crossref Full Text | Google Scholar

172. Liu P, Hu YY, Liu C, Zhu DY, Xue HM, Xu ZQ, et al. Clinical observation of salvianolic acid B in treatment of liver fibrosis in chronic hepatitis B. World J Gastroenterol. (2002) 8:679–85. doi: 10.3748/wjg.v8.i4.679

PubMed Abstract | Crossref Full Text | Google Scholar

173. Yang J, Zhang G, Tian J, Li C, Jiang W, Xing Y, et al. Cardioprotective effect of SMND-309, a novel derivate of salvianolic acid B on acute myocardial infarction in rats. Basic Clin Pharmacol Toxicol. (2010) 106:317–23. doi: 10.1111/j.1742-7843.2009.00490.x

PubMed Abstract | Crossref Full Text | Google Scholar

174. Mu L, Dong R, Li C, Chen J, Huang Y, Li T, et al. ROS responsive conductive microspheres loaded with salvianolic acid B as adipose derived stem cell carriers for acute myocardial infarction treatment. Biomaterials. (2025) 314:122849. doi: 10.1016/j.biomaterials.2024.122849

PubMed Abstract | Crossref Full Text | Google Scholar

175. Wei XH, Chen J, Wu XF, Zhang Q, Xia GY, Chu XY, et al. Salvianolic acid B alleviated myocardial ischemia-reperfusion injury via modulating SIRT3-mediated crosstalk between mitochondrial ROS and NLRP3. Phytomedicine. (2025) 136:156260. doi: 10.1016/j.phymed.2024.156260

PubMed Abstract | Crossref Full Text | Google Scholar

176. Ling WC, Liu J, Lau CW, Murugan DD, Mustafa MR, and Huang Y. Treatment with salvianolic acid B restores endothelial function in angiotensin II-induced hypertensive mice. Biochem Pharmacol. (2017) 136:76–85. doi: 10.1016/j.bcp.2017.04.007

PubMed Abstract | Crossref Full Text | Google Scholar

177. Zhao X, Jia H, Yang S, Liu Y, Deng B, Xu X, et al. Salvianolic Acid B reducing portal hypertension depends on macrophages in isolated portal perfused rat livers with chronic hepatitis. Evid Based Complement Alternat Med. (2012) 2012:786365. doi: 10.1155/2012/786365

PubMed Abstract | Crossref Full Text | Google Scholar

178. Xu H, Zhou Y, Lu C, Ping J, and Xu LM. Salvianolic acid B lowers portal pressure in cirrhotic rats and attenuates contraction of rat hepatic stellate cells by inhibiting RhoA signaling pathway. Lab Invest. (2012) 92:1738–48. doi: 10.1038/labinvest.2012.113

PubMed Abstract | Crossref Full Text | Google Scholar

179. Lin SJ, Lee IT, Chen YH, Lin FY, Sheu LM, Ku HH, et al. Salvianolic acid B attenuates MMP-2 and MMP-9 expression in vivo in apolipoprotein-E-deficient mouse aorta and in vitro in LPS-treated human aortic smooth muscle cells. J Cell Biochem. (2007) 100:372–84. doi: 10.1002/jcb.21042

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: polyphenols, salvianolic acid B, cardiovascular diseases, CVD, signaling pathways

Citation: Shao Y, Zhai L, Jiang S, Sun F and Liu X (2025) Salvianolic acid B: a promising cardioprotective agent. Front. Immunol. 16:1681358. doi: 10.3389/fimmu.2025.1681358

Received: 07 August 2025; Accepted: 05 November 2025; Revised: 04 November 2025;
Published: 21 November 2025.

Edited by:

Abdelhafid Nani, Université Ahmed Draia Adrar, Algeria

Reviewed by:

Youhua Wang, Shanghai University of Traditional Chinese Medicine, China
Baonian Liu, Shanghai University of Traditional Chinese Medicine, China

Copyright © 2025 Shao, Zhai, Jiang, Sun and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Fusheng Sun, NTY2NjY2NkAxMjYuY29t; Xuedong Liu, eHVlZG9uZ2xpdUAyNjMubmV0

These authors have contributed equally to this work

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.