
Exploring the potential of honokiol as a treatment for cardiovascular disease (Review)
- Authors:
- Published online on: June 10, 2025 https://doi.org/10.3892/br.2025.2012
- Article Number: 134
-
Copyright: © Zeng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
Cardiovascular disease (CVD) is the leading cause of global mortality, and its prevention and treatment thus represent a major public health challenge. The World Health Organization has estimated that ~17.9 million deaths worldwide were due to CVD in 2019, with 85% resulting from acute events such as myocardial infarction and stroke (1). Low- and middle-income countries account for three-quarters of global CVD-related mortality, with an estimated one-third of these deaths occurring in individuals #x003C;70 years of age (1). Pathologically, CVD encompasses a spectrum of noncommunicable diseases, including coronary artery disease, cerebrovascular disease, and peripheral arterial disease, which are driven primarily by modifiable risk factors such as unhealthy diet, a lack of physical activity, tobacco use, and obesity (1). Despite pharmacological advances, current interventions face significant limitations. For instance, while statins are effective for managing dyslipidemia, they carry risks of myotoxicity and rhabdomyolysis (2,3). Similarly, although dual antiplatelet therapy reduces mortality from myocardial infarction, it is associated with an increased risk of bleeding complications. These challenges are compounded by the multifactorial pathogenesis of CVD, necessitating therapeutic strategies that target multiple interconnected pathways, such as the use of anti-inflammatory, antioxidant, and cardioprotective agents (3). The effectiveness of conventional chemical agents is limited by their single-target mechanisms, potential for drug resistance, and risk of myotoxicity, while antiplatelet therapies are also linked to increased hemorrhagic risk (4). Within this context, natural products from traditional medicinal formulations have re-emerged as promising candidates for multimodal intervention.
Honokiol, a hydrophobic allyl biphenol compound isolated from the traditional Chinese herb Magnolia officinalis, represents a prime candidate in this quest (5). As one of the major bioactive components of Magnolia officinalis, its concentration serves as a key indicator for evaluating the quality of the herb (6). Beyond its established antimicrobial, antiviral, anticancer, anti-inflammatory, antioxidative, and anti-aging properties, honokiol has been found to have significant pharmacological effects on the cardiovascular system, including cardioprotection, enhancement of cardiac function, vasodilation, and antihypertensive effects, inhibition of platelet aggregation, and anti-atherosclerotic properties (7-9). These multifaceted properties and effects on cardiovascular health suggest the potential of developing honokiol as a novel therapeutic agent, opening new avenues in the treatment of CVD. The present review focused on the functions and associated mechanisms of honokiol in CVD, with an anlysis of its clinical applications and challenges. The aim was to provide new directions for research on the use of honokiol in clinical practice.
2. Structure and function of honokiol
Honokiol, chemically designated as 3',5-di-2-propenyl-1,1'-biphenyl-2,4'-diol, has a molecular formula of C18H18O2 and a molecular weight of 266.33 g/mol (Fig. 1). In appearance, this hydrophobic allyl biphenol compound is a white to off-white crystalline powder with a melting point of ~87.5˚C (8). While it has limited solubility in water, it is highly soluble in organic solvents such as ethanol, diethyl ether, and chloroform (8). The chemical structure of honokiol is characterized by the presence of two phenolic hydroxyls and two allyl groups. This distinctive configuration contributes to the variety of biological activities of the compound (8). The phenolic hydroxyl moieties act as hydrogen donors, conferring significant antioxidant activity and enabling the scavenging of excessive free oxygen radicals and thus reducing oxidative stress-mediated damage in cells and tissues (9). Concurrently, the allyl groups contribute to various chemical reactions involved in antibacterial, anti-inflammatory, and antitumor activities (9).
Pharmacological analyses of honokiol have demonstrated its wide range of biological activities (9). Honokiol has antimicrobial properties, inhibiting the growth of various pathogenic microorganisms, including Staphylococcus aureus, Escherichia coli, and Candida albicans. Its antimicrobial effects primarily involve the disruption of the cell membrane of the microorganism and interference with transport and energy functions, thereby reducing both antimicrobial and bactericidal/fungicidal effects (10). In terms of its anti-inflammatory properties, honokiol can inhibit the production of inflammatory mediators such as tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6(11). It also regulates inflammation-related signaling pathways, including that of nuclear factor kappa B (NF-κB) and reduces inflammatory cell infiltration, thus inducing anti-inflammatory effects (12). As an effective antioxidant, honokiol can scavenge and eliminate free radicals, such as superoxide anions and hydroxyl radicals, directly, and can also upregulate the activities of intracellular antioxidant enzymes, including superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), thereby enhancing the antioxidant defense mechanisms of the body (13). Furthermore, honokiol has a variety of other biological activities, including antitumor, antiviral, anti-anxiety, and sleep-promoting effects (8,9,14,15).
These biological activities are potentially linked to pharmacological effects on the cardiovascular system. Inflammation and oxidative stress play pivotal roles in the onset and progression of CVD (16,17). For instance, the formation of atherosclerotic plaques is closely associated with inflammatory cell infiltration and the production of oxidized low-density lipoproteins (18). The anti-inflammatory and antioxidant activities of honokiol can mitigate damage to vascular endothelial cells, suppress inflammatory responses, and reduce lipid oxidation, thereby exerting a protective effect on the cardiovascular system. Additionally, honokiol can block calcium channels, and can thus modulate calcium ion concentrations in cardiomyocytes (19) and vascular smooth muscle cells (20), influencing both the contractile and diastolic functions of the heart, as well as vascular tension. These findings provide a theoretical basis for its application in the treatment of CVD.
3. Biological activity of honokiol
Honokiol exhibits a broad spectrum of biological activities relevant to the therapeutic management of CVDs (21) (Fig. 2). These activities including antioxidant, anti-inflammatory, vasodilatory, anti-thrombotic, anti-platelet, and cardioprotective effects, highlight the value of studying honokiol to develop improved strategies for managing CVDs.
Antioxidant activity
A key attribute of honokiol is its potent antioxidant capacity. Oxidative stress, caused by the loss of appropriate balance between reactive oxygen species (ROS) and antioxidant defense systems, is a major contributor to the onset of CVDs such as atherosclerosis (22), myocardial infarction (23), and heart failure (24). Honokiol exhibits potent antioxidant activity through its ability to scavenge both ROS and reactive nitrogen species (RNS), including superoxide anions (O2-), hydroxyl radicals (·OH), and peroxynitrites (ONOO-). Its unique ortho-dihydroxy structural motif enables the efficient capture of free radicals (2.5 radicals per molecule), thereby disrupting lipid peroxidation. In a linoleic acid emulsion model, honokiol was found to block lipid peroxidation 15-20% more effectively than under control conditions, attributed to its ability to stabilize lipid membranes and suppress free radical propagation. This mechanism reduces the amount of oxidative damage to cellular organelles, extracellular matrices, and genomic DNA, as evidenced by decreased levels of malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine adducts in treated samples (25). It can help to scavenge free radicals, including superoxide anions and hydroxyl radicals, and it suppresses the biogenesis of ROS within cells (22). Moreover, honokiol was found to preserve mitochondrial function by inhibiting Fe(III)-adenosine diphosphate (ADP)/NADPH- and Fe(III)-ADP/NADH-induced lipid peroxidation, thereby sustaining the activities of respiratory enzymes and maintaining mitochondrial redox homeostasis. Honokiol treatment was observed to significantly reduce MDA levels in myocardial mitochondria in spontaneously hypertensive rat models while simultaneously increasing plasma nitrite/nitrate (NO2-/NO3-) levels, ultimately improving nitric oxide (NO) metabolism (13). In a model of sodium fluoride (NaF)-induced neurotoxicity, activation of the AMPK/PGC-1α/SIRT3 signaling axis restored the activity of SOD2, mitochondrial DNA transcription, and ATP synthesis, thereby reversing synaptic damage and cognitive deficits (26); these effects were mediated by honokiol activation of the AMPK/NFE2L2/SIRT3 signaling pathway, promoting AMPK phosphorylation and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) (22). This process led to an upregulation of sirtuin 3 (SIRT3) expression, thereby enhancing the activities of mitochondrial manganese superoxide dismutase (MnSOD) and catalase (CAT) and thus reducing ROS accumulation. Concurrently, the Keap1/Nrf2/ARE axis was found to promote the expression of the antioxidant enzyme glutathione (GSH), resulting in further reductions in both ROS accumulation and apoptosis (27). These data collectively demonstrate that honokiol enhances the activity of antioxidant enzymes such as SOD and CAT, strengthening the ability of the body to defend against oxidative stress. Consistent with these findings, it is reasonable to infer that these properties are particularly valuable in combating oxidative damage associated with endothelial dysfunction, plaque formation, and myocardial injury.
Anti-inflammatory effects
Chronic inflammation plays a central role in various cardiovascular conditions, including atherosclerosis, hypertension, and heart failure (28-30). Honokiol has been demonstrated to effectively inhibit pro-inflammatory signaling pathways such as the NF-κB (31), mitogen-activated protein kinase (MAPK) (32), and silent information regulator SIRT3(33) pathways. By suppressing NF-κB activation, honokiol can reduce the expression of inflammatory cytokines and mediators such as TNF-α, IL-6, IL-1β, inducible NO synthase (iNOS), cyclooxygenase-2 (COX2), vascular cell adhesion molecule 1, and intercellular adhesion molecule-1, all of which are pivotal in vascular inflammation and atherosclerosis (34). Additionally, honokiol has been shown to inhibit inflammatory enzymes, including COX2 and NOS, further mitigating inflammatory responses (35).
Anti-thrombotic and anti-platelet effects
Platelet aggregation and thrombus formation are both central to the pathophysiology of stroke, myocardial infarction, and other facets of ischemic heart disease. Honokiol has been demonstrated to inhibit both of these processes (36). It is capable of suppressing platelet aggregation and the formation of thromboxane A2, which mediates vasoconstriction and platelet function (37). Honokiol was observed to selectively inhibit platelet aggregation and calcium mobilization induced by collagen or convulxin [a glycoprotein VI (GPVI) agonist], preventing the phosphorylation of Lyn, phosholipase C γ2, protein kinase C, MAPKs, and AKT following convulxin stimulation, reducing binding of an anti-GPVI antibody (FITC-JAQ1) to human platelets, and significantly prolonging closure time in human whole-blood assays while increasing the occlusion time in murine models of thrombotic platelet thrombosis. These findings demonstrate the potential of honokiol as a novel anti-platelet and anti-thrombotic agent (38). Experimental studies have shown that honokiol can prevent platelet aggregation by reducing numerous agonists, such as ADP and collagen, emphasizing its potential utility as a therapeutic agent for limiting the incidence of thrombosis-related cardiovascular episodes (36).
Vasodilatory effects and protection of endothelial function
The maintenance of optimal blood pressure and organ perfusion hinges on the effective regulation of vascular tonus (39). Honokiol has been been shown to enhance vascular function through a variety of mechanisms, suggesting a promising avenue for the management of hypertension and ischemic heart disease. By stimulating the release of NO from endothelial cells, honokiol promotes vasodilation, as NO is a potent vasodilator that increases cyclic guanosine monophosphate levels and blocks L-type calcium channels, inhibiting calcium ion influx and relaxing vascular smooth muscle, thereby reducing vascular resistance and lowering blood pressure (40). Furthermore, honokiol has been demonstrated to enhance endothelial relaxation by activation of SIRT3, particularly under conditions of severe inflammation. As a specific activator of SIRT3, honokiol was shown to promote the SIRT3-dependent AMPK/eNOS pathway, restoring phosphorylation of eNOS at the Ser1177 site (41). This process not only induced NO generation but also improved endothelium-dependent vasodilation, ultimately restoring microcirculatory homeostasis (41).
In addition, honokiol mitigated reduced vasodilation in the aorta and mesenteric arteries, thereby improving endothelium-dependent vasodilation in a rat model of type 2 diabetes. Specifically, honokiol maintained the ratio of phosphorylated to total eNOS and increased the expression of CD31, which is typically reduced in diabetic rats. These findings suggest that honokiol may prevent diabetes-induced atherosclerosis by protecting the vascular endothelium and activating SIRT3, thereby enhancing the overall health and function of the endothelium (42). Honokiol has also been found to play a significant role in the regulation of vascular tonus, improvements in endothelial function, and the prevention of vascular dysfunction through multiple mechanisms. Activation of SIRT3 was shown to promote the generation of NO, and this, together with the inhibition of inflammation and oxidative stress, collectively improve vascular function and endothelial health (42). This multifaceted approach offers new strategies and hope for the treatment of CVD, highlighting the potential of honokiol as a valuable therapeutic agent.
Cardioprotective effects
Honokiol exhibits significant cardioprotective effects, particularly in ischemic heart diseases such as myocardial infarction and reperfusion injury (43). Its multifaceted mechanisms, including the reduction of oxidative stress, inflammation, and apoptosis, contribute to its efficacy in minimizing myocardial damage (44,45).
Experimental studies have demonstrated that honokiol reduces infarct size, prevents cardiomyocyte apoptosis, and improves cardiac function post-ischemia (44,45). These effects are mediated primarily through the activation of survival signaling pathways, inhibition of apoptotic cascades, and enhancement of mitochondrial function. Honokiol achieves these benefits by boosting autophagic flux and modulating the PI3K/AKT signaling pathway (44). Additionally, it has been shown to reduce the levels of cardiac fibrosis following ischemia, thereby preserving myocardial structure and function (45).
The cardioprotective effects of honokiol are further substantiated by its ability to promote the expression of the cardiac protective protein uncoupling protein 3 (UCP3), as well as its maintenance of the mitochondrial membrane potential. These effects reduce the production and accumulation of ROS following myocardial infarction, thereby mitigating myocardial fibrosis and improving heart failure outcomes (24). Honokiol was also found to block agonist-induced and pressure overload-mediated cardiac hypertrophy in mice, and these anti-hypertrophic effects were observed to be dependent on the activation of SIRT3(46). By enhancing the expression and activity of SIRT3, honokiol treatment resulted in reduced acetylation of SIRT3 substrates in the mitochondria, such as MnSOD and oligomycin sensitivity conferring protein (47). This resulted in enhanced mitochondrial respiration and reduced ROS synthesis in wild-type, but not in SIRT3-knockout, cells. Moreover, honokiol inhibited the proliferation of cardiac fibroblasts and their differentiation into myofibroblasts in a SIRT3-dependent manner (47).
In summary, the comprehensive cardioprotective functions of honokiol suggest its potential as a promising therapeutic candidate for ischemic heart diseases. Its ability to reduce oxidative stress, inflammation, and apoptosis, while enhancing mitochondrial function and autophagy, underscores its potential in mitigating myocardial damage and improving cardiac outcomes. The activation of SIRT3 and modulation of the PI3K/AKT pathway further consolidate its role in preventing cardiac hypertrophy and fibrosis.
4. Research progress of honokiol in cardiovascular diseases
Honokiol has garnered increasing attention in cardiovascular medicine due to its diverse pharmacological properties. This section offers an overview of the most up-to-date findings discussing the benefits of honokiol in different cardiovascular conditions (Fig. 3).
Honokiol in atherosclerosis
Atherosclerosis is characterized by chronic inflammation, oxidative stress, and endothelial dysfunction, with honokiol providing a means of targeting all of these deleterious features (48). Research indicates that honokiol can limit plaque formation, reduce arterial inflammation, and enhance endothelial function (11). Zhu et al observed that honokiol regulated proliferation and migration in rat aortic smooth muscle cells induced by TNF-α. This involved blocking the activation of NF-κB through suppression of the ERK signaling pathway (34). Preclinical investigations using animal models (such as ApoE-/- mice) have demonstrated marked reductions in atherosclerotic plaque size together with improved arterial function, attributed to the antioxidant and anti-inflammatory properties of honokiol (22).
Honokiol in hypertension
Hypertension management is vital for reducing cardiovascular morbidity and mortality. Preclinical studies have shown that honokiol effectively decreases both systolic and diastolic blood pressure (39,40). In spontaneously hypertensive rats, oral administration of honokiol (30 mg/kg/day) significantly reduced systolic blood pressure by 28% over 4 weeks by the reprogramming of arachidonic acid (AA) metabolism. This involved the downregulation of COX-2 and 5-lipoxygenase (LOX), critical enzymes in pro-inflammatory eicosanoid biosynthesis, while upregulating the expression of 12/15-LOX leading to increased production of the vasoprotective metabolite lipoxin A4. Concurrently, honokiol suppressed leukotriene B4 generation, thereby reducing inflammatory vasoconstriction (39).
In a complementary study using angiotensin II (Ang II)-induced hypertensive mice, honokiol (10 mg/kg/day) was observed to attenuate systolic hypertension by 22% through a novel epigenetic mechanism. Honokiol was found to inhibit histone deacetylase 6 (HDAC6)-mediated ubiquitination and proteasomal degradation of cystathionine γ-lyase (CSE), rate-limiting enzymes in the biosynthesis of hydrogen sulfide (H2S). Increased CSE activity restored H2S bioavailability, which in turn preserved the phosphorylation of eNOS and reduced oxidative stress, thereby ameliorating endothelial dysfunction (40).
Collectively, these findings demonstrate that honokiol exerts antihypertensive effects through several pathways, namely, the modulation of AA metabolism to promote vasodilatory mediators and epigenetic regulation of the HDAC6/CSE/H2S axis to protect endothelial function. These complementary mechanisms highlight the potential of honokiol as a multi-target therapeutic agent for hypertension, with implications for both blood pressure control and vascular protection.
Honokiol in myocardial infarction and ischemic heart disease
The cardioprotective properties of honokiol have been extensively explored in both acute and chronic myocardial infarction contexts. A study by Liu et al (24) investigated the therapeutic potential of honokiol in ameliorating heart failure following myocardial infarction. Using a mouse model of myocardial infarction, it was demonstrated that honokiol treatment led to a marked improvement in cardiac function, as evidenced by an increased ejection fraction and reduced left ventricular dilation. Mechanistically, honokiol upregulated the expression of UCP3, leading to reduced ROS generation and thus preventing oxidative stress (24). This effect was associated with preserved mitochondrial integrity and enhanced antioxidant capacity in cardiomyocytes. Furthermore, honokiol treatment reduced the levels of pro-inflammatory cytokines (such as TNF-α and IL-6) and inhibited apoptosis-related pathways, contributing to reduced myocardial fibrosis and cardiomyocyte death (24). The study highlighted UCP3-mediated ROS inhibition as a critical mechanism underlying the cardioprotective effects of honokiol, suggesting its potential as a therapeutic agent for heart failure after myocardial infarction.
In addition to its role in myocardial infarction, honokiol also has cardioprotective effects in ischemic heart disease. Tan et al (43) reported that honokiol treatment ameliorated myocardial ischemia/reperfusion injury by increasing autophagic flux and reducing ROS production. These protective effects were linked to the suppression of oxidative stress-related pathways (including NF-κB), coupled with the preservation of mitochondrial function via inhibition of mitochondrial permeability transition pore (mPTP) opening. Furthermore, honokiol alleviated myocardial ischemia/reperfusion injury by activating the PI3K/AKT signaling pathway, which inhibits mitochondrial apoptosis. Specifically, phosphorylation of downstream targets (such as GSK-3β) suppressed mPTP opening, thereby reducing cardiomyocyte apoptosis and improving the recovery of cardiac function (44). Moreover, Wang et al (45) described the dual actions of honokiol in cardioprotection as i) antioxidant effects induced by reducing lipid peroxidation markers (such as MDA) and ROS generation, and ii) anti-inflammatory effects induced by downregulation of pro-inflammatory cytokines (including TNF-α and IL-6). Additionally, honokiol may enhance myocardial tolerance to hypoxia by improving both microcirculation and energy metabolism (45). Collectively, these studies underscore the multifaceted role of honokiol in myocardial ischemia/reperfusion injury through the modulation of autophagy, mitochondrial apoptosis, oxidative stress, and inflammation, offering a promising therapeutic avenue for ischemic heart disease.
Honokiol in myocardial hypertrophy
Recent research has demonstrated that honokiol can effectively mitigate Ang II-induced cardiac hypertrophy by disrupting the nuclear receptor 77 (Nur77)-liver kinase B1 (LKB1) complex and activating the AMPK pathway (49). Under Ang II stimulation, Nur77 binds to LKB1, leading to its sequestration in the nucleus and impairment of its cytoplasmic role as an upstream activator of AMPKα, a critical regulator of energy homeostasis and suppressor of hypertrophy. Honokiol promotes the dissociation of the Nur77-LKB1 complex, facilitating LKB1 translocation to the cytoplasm, where it phosphorylates AMPKα at Thr172(49). Activated AMPKα inhibits mTOR/p70S6K signaling, which is a key driver of protein synthesis and cardiomyocyte growth, while restoring metabolic balance by enhancing fatty acid oxidation and suppressing glycolysis.
Furthermore, honokiol has also been found to exhibit multi-target anti-hypertrophic effects by attenuating Ang II-induced activation of the NF-κB and MAPK pathways, reducing the levels of inflammatory cytokines (such as TNF-α) and oxidative stress (40,50). It also downregulates the levels of angiotensin-converting enzyme and the Ang II receptor type 1 receptor, thereby suppressing overactivation of the renin-angiotensin system. In vivo studies using Ang II-infused mice revealed that honokiol could significantly reduce the thickness of the left ventricular wall, the cross-sectional area of cardiomyocytes, and the levels of fibrosis markers such as collagen deposition. In vitro experiments with neonatal cardiomyocytes confirmed that honokiol reversed Ang II-induced hypertrophy, an effect abolished by AMPK inhibition or LKB1 knockdown, underscoring the centrality of this pathway (49).
These findings highlight the potential of honokiol as a dual-target agent that disrupts pathological protein interactions (Nur77-LKB1) and restores AMPK-mediated metabolic regulation. In contrast to conventional therapies that primarily lower blood pressure, honokiol specifically addresses maladaptive cardiac remodeling without compromising hemodynamics, offering a safer profile. These findings indicate the advantages of the clinical translation of natural compounds targeting energy-sensitive pathways in CVD.
5. Prospects of honokiol in cardiovascular diseases
Honokiol has emerged as a promising therapeutic agent for CVD due to its diverse pharmacological properties. Its ability to exert anti-inflammatory, antioxidant, and cytoprotective effects makes it a versatile candidate for the management of various CVD-related pathologies. A key advantage of honokiol is its cardioprotective ability, particularly in the alleviation of myocardial ischemia/reperfusion injury. By activating the Nrf2/HO-1 pathway, honokiol promotes cellular resistance to oxidative stress, a critical contributor to both atherosclerosis and heart failure (26,27). Additionally, its anti-atherogenic effects, such as the alleviation of endothelial dysfunction, inhibition of low-density lipoprotein (LDL) oxidation, and the prevention of foam cell formation, highlight its potential in slowing plaque progression (11,48).
Honokiol also has vasoprotective benefits, primarily through its upregulation of eNOS, which promotes vasodilation and improves blood pressure regulation (39-42). This mechanism is particularly relevant for patients with hypertension and those with metabolic disorders, where endothelial dysfunction is prevalent. Furthermore, honokiol exhibits anti-arrhythmic properties through its ability to modulate cardiac ion channels, thereby stabilizing electrical activity and reducing the risk of ventricular arrhythmias (45). Its anti-fibrotic effects further aid in the prevention of adverse myocardial remodeling, making it a potential therapeutic option for atrial fibrillation and post-infarction complications. Another notable feature is the anti-thrombotic activity of honokiol, associated with suppression of platelet aggregation without increasing the risk of bleeding, a significant advantage over traditional antiplatelet drugs (36-38). Its dual role in mitigating inflammation (via NF-κB and NLRP3 inflammasome inhibition) (51,52) and enhancing metabolic homeostasis (via AMPK/SIRT1 activation) further underscores its potential in treating complex CVDs (13). Given its multi-targeted mechanisms and natural origin, honokiol holds promise as a complementary or alternative therapy for CVDs, particularly in patients with multifactorial conditions.
A 2022 patent (patent no. CN114588136A) described the preparation of honokiol-based medications for the treatment of myocardial infarction (53). In mice, honokiol use was found to significantly improve cardiac function for 4 consecutive weeks (Table I) (53). When honokiol was used in ischemic models, it was found that the levels of lactate dehydrogenase increased while apoptosis decreased. This suggests that honokiol may be effective for treating ischemia-related injury (Table I) (53). Moreover, a traditional Chinese medicinal formulation consisting of honokiol, pachymic acid, and 10-dehydroxygingerdione has been widely utilized in the treatment of cardiovascular and cerebrovascular diseases (Table I) (54). Additionally, Li et al (55) synthesized a novel compound through the cyclization of metformin with honokiol (Table I), which showed significant efficacy in combating CVD. Notably, it exhibited superior anti-inflammatory effects in mouse models of early atherosclerosis when compared to honokiol or metformin individually, indicating enhanced efficacy and reduced toxicity (55). One patent describes a class of honokiol derivatives or salts containing C2 and C4'-phenol hydroxy substitutions for the prevention and/or treatment of CVD (Table I) (56). In rat models, these derivatives demonstrated significant cardioprotective effects against stress-induced cardiomyopathy-related behaviors (Table I) (56). Recently, derivatives of honokiol that incorporate amine groups on the aromatic ring have been utilized in the formulation of drugs related to resistance to myocardial ischemia, as well as in the composition of myocardial protective medications (Table I) (57). In rat models of myocardial ischemia-reperfusion, the administration of compound 16 (1,500 µg/kg, delivered in two intravenous doses) notably decreased the size of the myocardial infarcts and lowered the levels of serum creatine kinase muscle-brain fraction and lactate dehydrogenase (Table I) (57). In conclusion, honokiol holds promising prospects for application in the prevention and treatment of CVD.
![]() | Table IPatents related to enhancing the therapeutic efficacy of honokiol in the treatment of cardiovascular diseases. |
6. Challenges in the clinical application of honokiol
Despite the therapeutic promise of honokiol, there are several barriers that impede the clinical adoption of honokiol in the treatment of CVD.
Limited bioavailability and pharmacokinetic constraints
One of the primary obstacles is the poor water solubility of honokiol, which restricts its oral absorption and systemic bioavailability. In CVDs, where sustained therapeutic levels are crucial, the rapid metabolism and short half-life of honokiol further diminish its efficacy. Research has indicated that honokiol undergoes extensive hepatic glucuronidation, leading to rapid clearance and suboptimal plasma concentrations (52). While the use of nanoparticle-based delivery systems and lipid formulations has been explored to enhance bioavailability, their scalability, stability, and cost-effectiveness remain unresolved issues for clinical use.
Insufficient clinical evidence in human CVD
Despite compelling preclinical data showing the ability of honokiol to reduce oxidative stress, inhibit vascular smooth muscle proliferation, and improve endothelial function, there are few clinical trials on its use in humans. Most studies have been conducted in cultured cells or animal models, which may not accurately reflect human pathophysiology (22-24). For instance, while honokiol has shown potential in reducing myocardial infarction size in rodents, its effects in human ischemic heart disease remain unverified. Additionally, there have been no systematic evaluations of optimal dosing, long-term safety, and potential interactions with standard cardiovascular medications (such as statins and antiplatelet drugs).
Mechanistic complexity and off-target effects
Honokiol modulates multiple signaling pathways relevant to CVDs, including the NF-κB, SIRT3, and PI3K/Akt pathways, which contribute to its anti-inflammatory and vasoprotective effects (42,58). However, this pleiotropic activity raises concerns about potential unintended effects, such as excessive modulation of blood pressure or interference with coagulation pathways. For example, the antiplatelet properties of honokiol could theoretically increase the risk of bleeding when used in combination with aspirin or clopidogrel. Further research is required to delineate its precise mechanisms and identify potential adverse interactions in patients with complex comorbidities.
Standardization and formulation challenges
The lack of standardized honokiol extracts complicates the reproducibility of clinical studies. Variations in purity, extraction methods, and chemical composition in preparations from different sources can lead to inconsistent therapeutic outcomes. Moreover, the development of a stable, scalable, and patient-friendly formulation (such as oral tablets or injectables) for CVD management remains a hurdle. While intravenous administration may bypass absorption issues, it introduces practical limitations for chronic use in outpatient settings.
Regulatory and commercial hurdles
As a natural product, honokiol faces regulatory ambiguities in terms of intellectual property and drug approval. Unlike synthetic drugs, its natural origin complicates its protection by patents, discouraging pharmaceutical investment. Additionally, competition with unregulated dietary supplements, which often lack rigorous quality control, undermines incentives for its clinical development. To overcome these barriers, collaborative efforts between academia, industry, and regulatory agencies are essential to establish standardized protocols and secure funding for large-scale trials.
7. Conclusion and outlook
Honokiol has demonstrated significant therapeutic potential in the treatment of CVD through its multifaceted mechanisms. It exerts cardioprotective effects by modulating calcium channels, suppressing Ang II-induced myocardial hypertrophy, and reducing ventricular remodeling. Its anti-inflammatory and antioxidant properties are mediated through inhibition of NF-κB signaling and enhancement of SOD/GSH-Px enzyme activities, effectively mitigating oxidative stress in atherosclerosis and myocardial ischemia. Furthermore, honokiol can ameliorate metabolic dysregulation by lowering blood glucose and lipid levels while inhibiting oxidized LDL formation, thereby preventing diabetic cardiovascular complications. Additional benefits include antiplatelet aggregation and vasodilation, highlighting its utility in thrombosis prevention. Despite these benefits, its translation to clinical use faces challenges. Specifically, its hydrophobicity limits oral bioavailability, necessitating advanced delivery systems such as lipid-based nanoparticles, its multi-target interactions with pathways, such as the Nrf2-SLC7A11-GSH and MAPK pathways, require precise mechanistic elucidation to minimize off-target effects, and the lack of robust clinical trials hinders validation of safety and efficacy in humans. Future efforts should prioritize the development of innovative formulations (such as polymer-stabilized nanosuspensions), mechanistic exploration using single-cell and organoid models to map spatiotemporal regulatory networks in myocardial fibrosis, and investigation of combination therapies with statins or immunomodulators to enhance synergistic effects. Expanding its application to pulmonary hypertension and arrhythmia could further broaden its therapeutic scope. With interdisciplinary collaboration and evidence-based validation, honokiol is poised to emerge as a precision treatment in CVD management, bridging the gap between traditional medicine and modern pharmacotherapy.
Acknowledgements
Not applicable.
Funding
Funding: Funding for the present study was received from the Natural Science Foundation of Guangdong Province (grant no. 2023A1515030033), and the Guangdong Basic and Applied Basic Research Foundation (grant no. 2022A1515140185).
Availability of data and materials
Not applicable.
Authors' contributions
YZ and ZL contributed to the conception and design of the review, and the literature collection. XL prepared the draft of the manuscript. All authors contributed to manuscript revision, and read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Podolec P and Matusik PT: New clinical classification of rare cardiovascular diseases and disorders: Relevance for cardiovascular research. Cardiovasc Res. 115:e77–e79. 2019.PubMed/NCBI View Article : Google Scholar | |
Kee PS, Chin PKL, Kennedy MA and Maggo SDS: Pharmacogenetics of statin-induced myotoxicity. Front Genet. 11(575678)2020.PubMed/NCBI View Article : Google Scholar | |
Vinci P, Panizon E, Tosoni LM, Cerrato C, Pellicori F, Mearelli F, Biasinutto C, Fiotti N, Di Girolamo FG and Biolo G: Statin-associated myopathy: Emphasis on mechanisms and targeted therapy. Int J Mol Sci. 22(11687)2021.PubMed/NCBI View Article : Google Scholar | |
Wang J, Zou J, Shi Y, Zeng N, Guo D, Wang H, Zhao C, Luan F, Zhang X and Sun J: Traditional Chinese medicine and mitophagy: A novel approach for cardiovascular disease management. Phytomedicine. 128(155472)2024.PubMed/NCBI View Article : Google Scholar | |
Chen C, Zhang QW, Ye Y and Lin LG: Honokiol: A naturally occurring lignan with pleiotropic bioactivities. Chin J Nat Med. 19:481–490. 2021.PubMed/NCBI View Article : Google Scholar | |
Rauf A, Olatunde A, Imran M, Alhumaydhi FA, Aljohani ASM, Khan SA, Uddin MS, Mitra S, Emran TB, Khayrullin M, et al: Honokiol: A review of its pharmacological potential and therapeutic insights. Phytomedicine. 90(153647)2021.PubMed/NCBI View Article : Google Scholar | |
Prasher P, Fatima R, Sharma M, Tynybekov B, Alshahrani AM, Ateşşahin DA, Sharifi-Rad J and Calina D: Honokiol and its analogues as anticancer compounds: Current mechanistic insights and structure-activity relationship. Chem Biol Interact. 386(110747)2023.PubMed/NCBI View Article : Google Scholar | |
Dai SY, Qin WX, Yu S, Li C, Yang YH and Pei YH: Honokiol and magnolol: A review of structure-activity relationships of their derivatives. Phytochemistry. 223(114132)2024.PubMed/NCBI View Article : Google Scholar | |
Li X, Yuan Z, Wang Y, Wang W and Shi J: Recent advances of honokiol: Pharmacological activities, manmade derivatives and structure-activity relationship. Eur J Med Chem. 272(116471)2024.PubMed/NCBI View Article : Google Scholar | |
Yang R, Cui L, Xu S, Zhong Y, Xu T, Liu J, Lan Z, Qin S and Guo Y: Membrane-targeting amphiphilic honokiol derivatives containing an oxazole moiety as potential antibacterials against methicillin-resistant Staphylococcus aureus. J Med Chem. 67:16858–16872. 2024.PubMed/NCBI View Article : Google Scholar | |
Liu A, Xun S, Zhou G, Zhang Y and Lin L: Honokiol alleviates sepsis-associated cardiac dysfunction via attenuating inflammation, apoptosis and oxidative stress. J Pharm Pharmacol. 75:397–406. 2023.PubMed/NCBI View Article : Google Scholar | |
Wang N, Kong R, Han W, Bao W, Shi Y, Ye L and Lu J: Honokiol alleviates ulcerative colitis by targeting PPAR-γ-TLR4-NF-κB signaling and suppressing gasdermin-D-mediated pyroptosis in vivo and in vitro. Int Immunopharmacol. 111(109058)2022.PubMed/NCBI View Article : Google Scholar | |
Zhou Y, Tang J, Lan J, Zhang Y, Wang H, Chen Q, Kang Y, Sun Y, Feng X, Wu L, et al: Honokiol alleviated neurodegeneration by reducing oxidative stress and improving mitochondrial function in mutant SOD1 cellular and mouse models of amyotrophic lateral sclerosis. Acta Pharm Sin B. 13:577–597. 2023.PubMed/NCBI View Article : Google Scholar | |
Kim H, Lim CY and Chung MS: Magnolia officinalis and its honokiol and magnolol constituents inhibit human norovirus surrogates. Foodborne Pathog Dis. 18:24–30. 2021.PubMed/NCBI View Article : Google Scholar | |
Qu WM, Yue XF, Sun Y, Fan K, Chen CR, Hou YP, Urade Y and Huang ZL: Honokiol promotes non-rapid eye movement sleep via the benzodiazepine site of the GABA(A) receptor in mice. Br J Pharmacol. 167:587–598. 2012.PubMed/NCBI View Article : Google Scholar | |
Ferrucci L and Fabbri E: Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 15:505–522. 2018.PubMed/NCBI View Article : Google Scholar | |
Shaito A, Aramouni K, Assaf R, Parenti A, Orekhov A, Yazbi AE, Pintus G and Eid AH: Oxidative stress-induced endothelial dysfunction in cardiovascular diseases. Front Biosci (Landmark Ed). 27(105)2022.PubMed/NCBI View Article : Google Scholar | |
Marchio P, Guerra-Ojeda S, Vila JM, Aldasoro M, Victor VM and Mauricio MD: Targeting early atherosclerosis: A focus on oxidative stress and inflammation. Oxid Med Cell Longev. 2019(8563845)2019.PubMed/NCBI View Article : Google Scholar | |
Aktay I, Bitirim CV, Olgar Y, Durak A, Tuncay E, Billur D, Akcali KC and Turan B: Cardioprotective role of a magnolol and honokiol complex in the prevention of doxorubicin-mediated cardiotoxicity in adult rats. Mol Cell Biochem. 479:337–350. 2024.PubMed/NCBI View Article : Google Scholar | |
Fan S, Li X, Lin J, Chen S, Shan J and Qi G: Honokiol inhibits tumor necrosis factor-alpha-stimulated rat aortic smooth muscle cell proliferation via caspase- and mitochondrial-dependent apoptosis. Inflammation. 37:17–26. 2014.PubMed/NCBI View Article : Google Scholar | |
Yuan Y, Zhou X, Wang Y, Wang Y, Teng X and Wang S: Cardiovascular modulating effects of magnolol and honokiol, two polyphenolic compounds from traditional Chinese medicine-Magnolia officinalis. Curr Drug Target. 21:559–572. 2020.PubMed/NCBI View Article : Google Scholar | |
Liu Y, Cheng P and Wu AH: Honokiol inhibits carotid artery atherosclerotic plaque formation by suppressing inflammation and oxidative stress. Aging (Albany NY). 12:8016–8028. 2020.PubMed/NCBI View Article : Google Scholar | |
Liou KT, Lin SM, Huang SS, Chih CL and Tsai SK: Honokiol ameliorates cerebral infarction from ischemia-reperfusion injury in rats. Planta Med. 69:130–134. 2003.PubMed/NCBI View Article : Google Scholar | |
Liu J, Tang M, Li T, Su Z, Zhu Z, Dou C, Liu Y, Pei H, Yang J, Ye H and Chen L: Honokiol ameliorates post-myocardial infarction heart failure through Ucp3-mediated reactive oxygen species inhibition. Front Pharmacol. 13(811682)2022.PubMed/NCBI View Article : Google Scholar | |
Zhao C and Liu ZQ: Comparison of antioxidant abilities of magnolol and honokiol to scavenge radicals and to protect DNA. Biochimie. 93:1755–1760. 2011.PubMed/NCBI View Article : Google Scholar | |
Wang D, Cao L, Zhou X, Wang G, Ma Y, Hao X and Fan H: Mitigation of honokiol on fluoride-induced mitochondrial oxidative stress, mitochondrial dysfunction, and cognitive deficits through activating AMPK/PGC-1α/Sirt3. J Hazard Mater. 437(129381)2022.PubMed/NCBI View Article : Google Scholar | |
Caballero EP, Mariz-Ponte N, Rigazio CS, Santamaria MH and Corral RS: Honokiol attenuates oxidative stress-dependent heart dysfunction in chronic Chagas disease by targeting AMPK/NFE2L2/SIRT3 signaling pathway. Free Radical Biol Med. 156:113–124. 2020.PubMed/NCBI View Article : Google Scholar | |
Lin A, Miano JM, Fisher EA and Misra A: Chronic inflammation and vascular cell plasticity in atherosclerosis. Nat Cardiovasc Res. 3:1408–1423. 2024.PubMed/NCBI View Article : Google Scholar | |
Guzik TJ, Nosalski R, Maffia P and Drummond GR: Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol. 21:396–416. 2024.PubMed/NCBI View Article : Google Scholar | |
Boulet J, Sridhar VS, Bouabdallaoui N, Tardif JC and White M: Inflammation in heart failure: Pathophysiology and therapeutic strategies. Inflamm Res. 73:709–723. 2024.PubMed/NCBI View Article : Google Scholar | |
Lee J, Jung E, Park J, Jung K, Lee S, Hong S, Park J, Park E, Kim J, Park S and Park D: Anti-inflammatory effects of magnolol and honokiol are mediated through inhibition of the downstream pathway of MEKK-1 in NF-kappaB activation signaling. Planta Med. 71:338–343. 2005.PubMed/NCBI View Article : Google Scholar | |
Tang X, Yao K, Zhang L, Yang Y and Yao H: Honokiol inhibits H2O2-induced apoptosis in human lens epithelial cells via inhibition of the mitogen-activated protein kinase and Akt pathways. Eur J Pharmacol. 650:72–78. 2011.PubMed/NCBI View Article : Google Scholar | |
Ye JS, Chen L, Lu YY, Lei SQ, Peng M and Xia ZY: SIRT3 activator honokiol ameliorates surgery/anesthesia-induced cognitive decline in mice through anti-oxidative stress and anti-inflammatory in hippocampus. CNS Neurosci Ther. 25:355–366. 2019.PubMed/NCBI View Article : Google Scholar | |
Zhu X, Wang Z, Hu C, Li Z and Hu J: . Honokiol suppresses TNF-α-induced migration and matrix metalloproteinase expression by blocking NF-κB activation via the ERK signaling pathway in rat aortic smooth muscle cells. Acta Histochem. 116:588–595. 2014.PubMed/NCBI View Article : Google Scholar | |
Murakami Y, Kawata A, Seki Y, Koh T, Yuhara K, Maruyama T, Machino M, Ito S, Kadoma Y and Fujisawa S: Comparative inhibitory effects of magnolol, honokiol, eugenol and bis-eugenol on cyclooxygenase-2 expression and nuclear factor-kappa B activation in RAW264.7 macrophage-like cells stimulated with fimbriae of Porphyromonas gingivalis. In Vivo. 26:941–950. 2012.PubMed/NCBI | |
Montecino-Garrido H, Méndez D, Araya-Maturana R, Millas-Vargas JP, Wehinger S and Fuentes E: In vitro effect of mitochondria-targeted triphenylphosphonium-based compounds (Honokiol, Lonidamine, and Atovaquone) on the platelet function and cytotoxic activity. Front Pharmacol. 13(893873)2022.PubMed/NCBI View Article : Google Scholar | |
Seok YM, Cho HJ, Cha BY, Woo JT and Kim IK: Honokiol attenuates vascular contraction through the inhibition of the RhoA/Rho-kinase signalling pathway in rat aortic rings. J Pharm Pharmacol. 63:1244–1251. 2011.PubMed/NCBI View Article : Google Scholar | |
Onselaer MB, Nagy M, Pallini C, Pike JA, Perrella G, Quintanilla LG, Eble JA, Poulter NS, Heemskerk JWM and Watson SP: Comparison of the GPVI inhibitors losartan and honokiol. Platelets. 31:187–197. 2020.PubMed/NCBI View Article : Google Scholar | |
Elbarbry F and Moshirian N: The modulation of arachidonic acid metabolism and blood pressure-lowering effect of honokiol in spontaneously hypertensive rats. Molecules. 27(3396)2022.PubMed/NCBI View Article : Google Scholar | |
Chi Z, Le TPH, Lee SK, Guo E, Kim D, Lee S, Seo SY and Lee SY, Kim JH and Lee SY: Honokiol ameliorates angiotensin II-induced hypertension and endothelial dysfunction by inhibiting HDAC6-mediated cystathionine γ-lyase degradation. J Cell Mol Med. 24:10663–10676. 2020.PubMed/NCBI View Article : Google Scholar | |
Lv D, Luo M, Yan J, Yang X and Luo S: Protective effect of sirtuin 3 on CLP-induced endothelial dysfunction of early sepsis by inhibiting NF-κB and NLRP3 signaling pathways. Inflammation. 44:1782–1792. 2021.PubMed/NCBI View Article : Google Scholar | |
He A, Yu H, Hu Y, Chen H, Li X, Shen J, Zhuang R, Chen Y, Sasmita BR, Luo M and Lv D: Honokiol improves endothelial function in type 2 diabetic rats via alleviating oxidative stress and insulin resistance. Biochem Biophys Res Commun. 600:109–116. 2022.PubMed/NCBI View Article : Google Scholar | |
Tan Z, Liu H, Song X, Ling Y, He S, Yan Y, Yan J, Wang S, Wang X and Chen A: Honokiol post-treatment ameliorates myocardial ischemia/reperfusion injury by enhancing autophagic flux and reducing intracellular ROS production. Chem Biol Interact. 307:82–90. 2019.PubMed/NCBI View Article : Google Scholar | |
Lv L, Kong Q, Li Z and Zhang Y, Chen B, Lv L and Zhang Y: Honokiol provides cardioprotection from myocardial ischemia/reperfusion injury (MI/RI) by inhibiting mitochondrial apoptosis via the PI3K/AKT signaling pathway. Cardiovasc Ther. 2022(1001692)2022.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Zhang ZZ, Wu Y, Zhan J, He XH and Wang YL: Honokiol protects rat hearts against myocardial ischemia reperfusion injury by reducing oxidative stress and inflammation. Exp Ther Med. 5:315–319. 2013.PubMed/NCBI View Article : Google Scholar | |
Pillai VB, Samant S, Sundaresan NR, Raghuraman H, Kim G, Bonner MY, Arbiser JL, Walker DI, Jones DP, Gius D and Gupta MP: Honokiol blocks and reverses cardiac hypertrophy in mice by activating mitochondrial Sirt3. Nat Commun. 6(6656)2015.PubMed/NCBI View Article : Google Scholar | |
Zhong C, Wang C, Li W, Li W, Chen X, Guo J, Feng Y and Wu X: A derivative of honokiol HM568 has an anti-neuroinflammatory effect in Parkinson's disease. Chem Biol Interact. 403(111212)2024.PubMed/NCBI View Article : Google Scholar | |
Qiu L, Xu R, Wang S, Li S, Sheng H, Wu J and Qu Y: Honokiol ameliorates endothelial dysfunction through suppression of PTX3 expression, a key mediator of IKK/IkappaB/NF-ĸB, in atherosclerotic cell model. Exp Mol Med. 47(e171)2015.PubMed/NCBI View Article : Google Scholar | |
Lin X, Zhang H, Chu Y, Zhang Y, Xu C, Xie H, Ruan Q, Lin J, Huang CK and Chai D: Honokiol ameliorates angiotensin II-induced cardiac hypertrophy by promoting dissociation of the Nur77-LKB1 complex and activating the AMPK pathway. J Cell Mol Med. 28(e18028)2024.PubMed/NCBI View Article : Google Scholar | |
Zhu J, Ning RB, Lin XY, Chai DJ, Xu CS, Xie H, Zeng JZ and Lin JX: Retinoid X receptor agonists inhibit hypertension-induced myocardial hypertrophy by modulating LKB1/AMPK/p70S6K signaling pathway. Am J Hypertens. 27:1112–1124. 2014.PubMed/NCBI View Article : Google Scholar | |
Cai X, Jiang X, Zhao M, Su K, Tang M, Hong F, Ye N, Zhang R, Li N, Wang L, et al: Identification of the target protein and molecular mechanism of honokiol in anti-inflammatory action. Phytomedicine. 109(154617)2023.PubMed/NCBI View Article : Google Scholar | |
Huang PP, Fu J, Liu LH, Wu KF, Liu HX, Qi BM, Liu Y and Qi BL: Honokiol antagonizes doxorubicin-induced cardiomyocyte senescence by inhibiting TXNIP-mediated NLRP3 inflammasome activation. Int J Mol Med. 45:186–194. 2020.PubMed/NCBI View Article : Google Scholar | |
Guo Y, Liu N, Zhang J, Yang J, Kang W, Lu T, Guo Y and Yao Y: Application of honokiol in preparation of medicine for treating myocardial infarction. China Patent CN114588136A. Filed April 7, 2022; issued June 7, 2022. | |
Miao GP, Han J, Zhang JF, Tong G and Wu Y: A kind of active ingredient of Chinese herbs composition for treating cardiovascular and cerebrovascular disease China Patent CN109045045A. Filed August 14, 2018; issued December 21, 2018. | |
Li WM, Feng YF, Zhu HN, Yu R and Li Y: A kind of compound and its preparation method and application. China Patent CN108033927A. Filed October 27, 2017; issued May 15, 2018. | |
Guang B, Yang T, Dong RH, Peng X, Liu I, Xie J, Qin L, Xu G and Liao X: The new application of honokiol derivative. China Patent CN109771431A. Filed January 31, 2019; issued May 21, 2019. | |
Zhang P, Liu Y, Zhang Y and Gu J: Application of magnolol or aromatic ring amino substituted derivative of honokiol in preparation of anti-myocardial ischemia drugs and pharmaceutical composition. China Patent CN115581690A. Filed July 5, 2021; issued January 10, 2023. | |
Xian X, Zhao X, Zhou X, Liu H, Li C, Wu X, Chen Y, Ye K, Yang H, Li M, et al: Honokiol attenuates oxidative stress and vascular calcification via the upregulation of heme oxygenase-1 in chronic kidney disease. Toxicol Appl Pharmacol. 499(117318)2025.PubMed/NCBI View Article : Google Scholar |