Next Article in Journal
Residual Humidity in Paraffin-Embedded Tissue Reduces Nucleic Acid Stability
Previous Article in Journal
Shiga Toxin, Stx2e, Influences the Activity of Porcine Lymphocytes In Vitro
Previous Article in Special Issue
Nature-Inspired Surface Structures Design for Antimicrobial Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ferulic Acid: A Review of Pharmacology, Toxicology, and Therapeutic Effects on Pulmonary Diseases

Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 8011; https://doi.org/10.3390/ijms24098011
Submission received: 13 March 2023 / Revised: 14 April 2023 / Accepted: 27 April 2023 / Published: 28 April 2023
(This article belongs to the Collection Advances in Medicinal Chemistry)

Abstract

:
Ferulic acid (FA), a prevalent dietary phytochemical, has many pharmacological effects, including anti-oxidation and anti-inflammation effects, and has been widely used in the pharmaceutical, food, and cosmetics industries. Many studies have shown that FA can significantly downregulate the expression of reactive oxygen species and activate nuclear factor erythroid-2-related factor-2/heme oxygenase-1 signaling, exerting anti-oxidative effects. The anti-inflammatory effect of FA is mainly related to the p38 mitogen-activated protein kinase and nuclear factor-kappaB signaling pathways. FA has demonstrated potential clinical applications in the treatment of pulmonary diseases. The transforming growth factor-β1/small mothers against decapentaplegic 3 signaling pathway can be blocked by FA, thereby alleviating pulmonary fibrosis. Moreover, in the context of asthma, the T helper cell 1/2 imbalance is restored by FA. Furthermore, FA ameliorates acute lung injury by inhibiting nuclear factor-kappaB and mitogen-activated protein kinase pathways via toll-like receptor 4, consequently decreasing the expression of downstream inflammatory mediators. Additionally, there is a moderate neuraminidase inhibitory activity showing a tendency to reduce the interleukin-8 level in response to influenza virus infections. Although the application of FA has broad prospects, more preclinical mechanism-based research should be carried out to test these applications in clinical settings. This review not only covers the literature on the pharmacological effects and mechanisms of FA, but also discusses the therapeutic role and toxicology of FA in several pulmonary diseases.

1. Introduction

Natural products have long been investigated and exploited for the development of new drugs [1]. Ferulic acid (4-hydroxy-3-methoxycinnamic acid, FA) belongs to the phenolic acids commonly found in medical plants (Figure 1). The key physico-chemical properties of FA are shown in Table 1. FA mainly exists in plant cell walls and contributes to the structural integrity and rigidity by being covalently linked to polysaccharides, such as arabinoxylans, which is also a precursor of lignin, a complex polymer that provides mechanical strength and resistance to biodegradation in plant tissues [2,3,4,5]. So far, it has been proven that FA and its derivatives have a variety of pharmacological effects, especially anti-oxidative, anti-inflammatory, anti-allergic, anti-cancer, and anti-fibrotic effects [6,7,8,9,10,11].
FA has been proven to be effective in many disease models through various mechanisms [12,13,14,15,16,17,18]. For example, the nuclear factor-kappaB (NF-κB) and nuclear factor erythroid-2-related factor-2/heme oxygenase-1 (Nrf2/HO-1) signaling pathways are regulated by FA to resist oxidative damage and restrain inflammatory responses, thereby exerting cardioprotective effects [19,20]. Chronic pulmonary diseases are the leading causes of morbidity and mortality worldwide, and the treatment has received increasing attention in recent years. Accumulating evidence suggests that FA improves lung function and survival in pulmonary diseases, including idiopathic pulmonary fibrosis (IPF), acute lung injury/acute respiratory distress syndrome (ALI/ARDS), lung cancer, etc. However, low bioavailability and the limited number of clinical studies have restricted the use of FA in medicine. This review summarizes the main pharmacological effects and mechanisms of FA and discusses its potential role in the treatment of pulmonary diseases.

2. Pharmacological Effects

Many researchers have found that FA possesses distinct pharmacological effects, such as anti-oxidative, anti-inflammatory, anti-fibrotic, and anti-cancer effects, as well as complex mechanisms both in vitro and in vivo.

2.1. Anti-Oxidative Effects

FA is considered to be a superior antioxidant, which easily forms resonance-stabilized phenoxy radicals due to its phenolic nucleus and an extended side chain conjugation, thus acting as a free radical scavenger [21]. FA can attenuate oxidative stress damage, and the effect is much stronger than vanillic, coumaric, and cinnamic acid [22]. Studies have shown that FA can inhibit the production of reactive oxygen species (ROS), scavenge oxidative free radicals, and participate in various signaling pathways to exert its anti-oxidative effects.

2.1.1. ROS

Oxidative stress arises from impaired endogenous antioxidative defense and/or an overwhelming presence of ROS, which is an important molecular mechanism in various organ pathologies [23,24]. FA treatment can inhibit the production and activity of ROS inducer markers, such as advanced glycation end products and xanthine oxidase [25]. The content of advanced glycation end products and xanthine oxidase activity are evaluated by measuring the immunoblotting data and enzymatic oxidation of xanthine, respectively. In addition, FA can significantly downregulate ROS levels with an ROS-sensitive probe, inhibiting pathological angiogenesis and reducing cell damage [26,27]. In lung cancer, pretreatment with FA initially decreases ROS levels and reduces oxidative damage [28].

2.1.2. Free Radical Scavenging

Previous studies have reported that FA is an antioxidant that neutralizes free radicals such as superoxide, nitric oxide and hydroxyl radicals that may cause oxidative damage to cell membranes and DNA [22]. Due to its structural properties, FA is a direct scavenger of free radicals such as hydroxyl radicals, superoxide, hydrogen peroxide, and nitrogen dioxide radicals [29]. FA has been tested for the radical scavenging property using the 2,2-diphenyl 1-picryl hydrazyl scavenging assay [30]. In a concentration-dependent manner, FA is able to significantly scavenge 2,2-diphenyl 1-picryl hydrazyl free radical with a half-maximal inhibitory concentration (IC50) of approximately 33 μM [25]. The generation and detection of hydroxyl radicals are carried out according to a Fenton reaction [31]. Nitric oxide radical scavenging assay has been performed as described by Sumanont et al. [32], with minor modifications. Park and co-worker originally described the procedure used to assess superoxide anion radical production, which was later modified [33]. Catalase activity has been determined essentially using the method described by Aebi [34,35]. Among several tested polyphenols, including ellagic acid, tannic acid, caffeic acid, and FA, the latter is able to suppress the formation of superoxide anion radicals induced by tumor promoters to the greatest extent [36].
The anti-oxidative effect of FA is closely related to the 3-methoxy and 4-hydroxyl groups on the benzene ring, which can stabilize the resulting phenoxy radical intermediates and even terminate the radical chain reaction. The carboxylic acid group with adjacent immature carbon-carbon double bonds can further promote the resonance stabilization of the phenoxy radical intermediates or provide additional attack sites for the radicals [37]. Through this scavenging effect, FA significantly attenuates peroxyl radical-induced cell death and reduces both hydroxyl radical-induced proteins and lipid oxidative damage in hippocampal synaptosomes in vitro [38]. FA efficiently suppresses lipid peroxidation triggered by peroxyl radicals. Additionally, the activity and function of enzymes responsible for scavenging free radicals, such as cardiac superoxide dismutase, glutathione peroxidase, and catalase, are increased by FA [35].

2.1.3. Nrf2/HO-1

Nrf2 is one of the main coordinating factors of the oxidative stress response and plays a protective role in many different organs, including the lungs [39], kidneys [40], liver [41], and colon [42]. Nrf2 knockout mice have been reported to be hypersensitive to ALI; their lungs become severely edematous, and microscopy reveals a loss of alveolar structure with pulmonary hemorrhage and infiltration [43]. Under stressed conditions, exposure to toxicants, ROS, genetic mutations, oncogenic signals, or autophagic disruption results in a temporary or constitutive increase in cellular Nrf2, which disrupts the Kelch-like-ECH-associated protein 1 (keap1)-Nrf2 complex, causing Nrf2 activation. Then, Nrf2 dissociates from Keap1 and translocates to the cell nucleus, where it regulates the transcription of target genes, which encode proteins involved in antioxidants, detoxification, anti-inflammation, and metabolism [44,45,46]. FA induces the translocation of Nrf2 from the cytoplasm to nucleus and promotes the expression of Nrf2 as well as downstream antioxidative proteins such as HO-1, playing a cytoprotective role (Figure 2A) [47,48].
FA remarkably prevents nephrotoxicity in rats through activating Nrf2/HO-1 signaling, exerting an anti-oxidative effect [49]. It has been confirmed that FA reverses methotrexate-induced reduction of Nrf2 and HO-1 mRNA in rats with liver injury [50]. Moreover, FA improves alveolar epithelial barrier dysfunction to ameliorate ALI via Nrf2/HO-1 signaling [51]. Similarly, the activation of Nrf2/HO-1 by FA can also have therapeutic potential in alleviating ionizing radiation-induced cataracts [52].

2.2. Anti-Inflammatory Effects

Inflammation is the defensive response of the human body when exposed to external stimuli; however, excessive inflammation can result in a variety of diseases [53]. Lipopolysaccharide (LPS) is commonly used in experiments to construct inflammatory pathological models and can be recognized by toll-like receptor 4 (TLR4), which is expressed on the cell surface [54]. The bond of LPS and TLR4 triggers signal transduction cascades in cells, resulting in the activation of NF-κB and mitogen-activated protein kinases (MAPKs), thereby stimulating secretions of inflammatory mediators such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) (Figure 2B) [55,56]. Meanwhile, pro-inflammatory cytokines, such as IL-6, IL-11, and IL-13, activate the Janus kinase/signal transducer and activator of transcription (JAK/STAT) molecular pathway to induce inflammation and regulate the immune response [57,58]. One of the most extensively researched inflammasomes, NOD-like receptor-family pyrin domain-containing 3 (NLRP3), can be regulated by NF-κB-induced transcription [59]. FA has shown anti-inflammatory activity through inhibiting these signaling pathways in vitro and in vivo, highlighting its potential as an anti-inflammatory drug.

2.2.1. p38 MAPK

MAPKs are major signal molecules in transduction, catalyzing the phosphorylation of appropriate protein substrates on serine or threonine residues, and playing an important role in the development and progression of inflammation [60,61]. As a member of the MAPKs family, activation of p38 MAPK plays a significant role in the production of pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α [62,63], and induction of enzymes like cyclooxygenase-2 to regulate connective tissue remodeling, as well as adherent proteins and other inflammatory-related molecules [64,65]. Inflammatory stimuli such as LPS, TNF, platelet activator, and IL can induce p38 activation in endogenous immune cells, such as monocytes, endothelial cells, and neutrophils [66]. The phosphorylation of p38 followed by LPS stimulation is restrained by FA, suggesting that FA may exert anti-inflammatory effects via the inhibition of the p38 MAPK pathway [56,67].
The gene expression of indoleamine 2,3-dioxygenase (IDO) is determined by p38 MAPK, and the activity is induced by cellular immune activation associated with inflammatory diseases [68]. LPS induces the expression of IDO, and FA can pass through the blood-brain barrier to reduce IDO by suppressing the phosphorylation of p38 MAPK, which may provide new ideas for the prevention and treatment of diseases [69]. Moreover, in testicular toxicity induced by cisplatin, a chemotherapeutic drug, pretreatment with FA significantly degrades the expression of p38 MAPK in rats, markedly alleviating cisplatin-induced testicular damage [70].

2.2.2. NF-κB

The NF-κB family of transcription factors contains five members: NF-κB1 (p105/p50), NF-κB2 (p100/p52), RelA (p65), RelB, and c-Rel, among which p65 is the most extensively studied subunit that contains transcriptional activation domains [71,72]. As the major receptor for LPS, TLR4 exists in a complex with co-receptor myeloid differentiation protein-2 [73,74]. Upon binding to LPS, the TLR4-myeloid differentiation protein-2 complex dimerizes, which leads to the activation of downstream mediators, including NF-κB [75]. Stimulation triggers degradation of the inhibitor of NF-κB (IκB) protein and release of NF-κB homo- or heterodimers, which subsequently translocate to the nucleus, then bind to specific DNA sequences and promote the transcription of pro-inflammatory genes [76]. Treatment with FA significantly inhibits the expression of LPS-induced TLR4, degradation of IκB, and phosphorylation of p65. Docking results have shown that FA targets the key binding site of TLR4 and disrupts the formation of the TLR4-myeloid differentiation factor 2 complex, which provides a new strategy for the treatment of inflammation [77]. In addition, researchers have found that FA markedly prevents IκB phosphorylation and subsequent nuclear translocation of NF-κB [78,79]. FA treatment has the same effect as TLR4 inhibitor (TSK242) and NF-κB inhibitor (SP600125), giving it the potential to act as an effective inhibitor of inflammation [77].

2.2.3. JAK/STAT

Various cytokines and growth factors can bind to their respective receptors on the cell surface and lead to the phosphorylation and activation of JAK kinases and STATs sequentially [80]. The latter translocate to the nucleus and regulate the expression of genes involved in cell proliferation, differentiation, survival, and inflammation (Figure 2B) [81]. Accordingly, the JAK/STAT signaling system may be a useful indicator of a strong immune response, inhibiting of which could help to reduce hyperinflammatory conditions [82]. FA has been found to have promising JAK2 inhibition through molecular docking with a score of −6.7, which is comparable to that of ruxolitinib, a standard JAK2 inhibitor [83].

2.2.4. NLRP3

The NLRP3 inflammasome is a cytosolic protein complex that senses cellular stress or damage and initiates inflammatory responses, which has been implicated in various pulmonary diseases, such as asthma and chronic obstructive pulmonary disease (COPD) [84,85]. Therefore, inhibiting the activation of NLRP3 inflammasome is a potential strategy for preventing or treating inflammation-related diseases [86]. Studies have shown that FA can exhibit anti-inflammatory effects by blocking the activation of NLRP3 and reducing the secretion of TNF-α, IL-1β, and IL-6 [87,88].

2.3. Anti-Fibrotic Effects

Fibrosis is a repair or reactive process characterized primarily by the formation of fibrous connective tissue, resulting in progressive structural remodeling of almost all tissues and organs [89]. For example, the pathogenesis of pulmonary fibrosis (PF) arises from repeated damage to the alveolar epithelium or endothelium, triggering the immune system to recover the tissue structure of the injured tissue. Inflammatory mediators, such as transforming growth factor-β (TGF-β), are able to activate angiogenesis and myofibroblasts, which promote the generation of extracellular matrix (ECM) constituents [90,91]. Excessive accumulation of ECM and promotion of fibrosis through endogenous and exogenous stimuli that induce elevation of TGF-β1 exacerbate the imbalance between matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) [92,93]. When ECM is overproduced and deposited in organ tissues, substantial scar formation and destruction of normal organ architecture occurs [94]. Almost all lung diseases end in fibrosis, but an effective cure for fibrosis has yet to be found. In some early reports, FA was found to be helpful in suppressing fibrosis [95,96].

2.3.1. TGF-β/Small Mothers against Decapentaplegic

TGF-β is a multifunctional regulatory cytokine known to regulate various cellular processes, such as proliferation, differentiation, apoptosis, adhesion, and the pathogenesis of fibrosis, in which TGF-β1 is a key mediator of fibrosis development, exerting biological effects through activation of downstream mediators [97]. After the downstream small mothers against decapentaplegic (Smad)2 and Smad3 are activated, they form a complex with Smad4 that translocates to the nucleus, binds consensus sequences, and regulates gene transcription, which induces ECM deposition to promote fibrosis (Figure 2C) [98,99,100].
FA can block the activation of the Smad2/3 signal, reverse Smad4 nuclear translocation, inhibit the epithelial–mesenchymal transition process, which is driven by TGF-β1, and resist the occurrence of fibrosis [101,102]. In addition, it has been found that FA can decrease Smad3 and Smad4 by inhibiting the expression of TGF-β and its receptor and can cooperate with astragaloside IV to alleviate fibrosis in rats [103]. Based on an in vitro study, TGF-β signal transduction can be blocked by FA, which significantly reduces Smad signal transduction to inhibit the activation of hepatic stellate cells [95].

2.3.2. MMPs/TIMPs

The MMPs/TIMPs system has been reported to be regulated by FA [104]. MMP2 and MMP9 have three fibronectin type II structural domains repeatedly inserted into the catalytic structural domain, which are closely associated with the development of fibrosis [105]. MMP1 is involved in the diminution of normal and hypertrophic scars [106]. TIMPs play essential roles in the activation or elimination of MMPs from the extracellular environment, which determines the effects of ECM on cytokines, chemokines, cell adhesion molecules, and growth factors [107]. TIMP1 is secreted by most cells and inhibits all types of MMPs, among which TIMP1 binds particularly strongly to MMP9 [108]. FA has displayed reductions in MMP2 and MMP9, and an increase in TIMP1 expression [109]. One study has shown that FA may lead to a significant reduction in MMP2 and MMP9 levels via the proteasome pathway [110].

2.4. Anti-Cancer Effects

Cancer is a serious disease that causes deaths all over the world, and the incidence and mortality rates are increasing rapidly [111]. Evading apoptosis has been established as one of the key characteristic features of cancer cells [112]. Cancer proliferation can be inhibited by FA in a variety of ways, including by altering the cancer cell cycle, inducing apoptosis, and regulating protein production [113].
FA prevents migration in breast cancer cells and so suppresses breast cancer cell proliferation and induces apoptosis [114]. In addition, many studies have shown that FA has anti-cancer effects against cervical cancer [115], colon cancer [116], liver cancer [28], and lung cancer [117]. ElKhazendar et al., have studied the therapeutic effect of FA on liver cancer and found that FA (100 and 200 μg/mL) has cytotoxic effects on HepG2 cells with IC50 values of 150.7, 81.38, and 210.4 μg/mL at 24, 48, and 72 h, respectively [118]. They have reported similar findings after analyzing the anti-cancer potential of FA (100 and 200 μg/mL) on MCF-7 breast cancer cells in vitro and found IC50 values of cell proliferation for MCF-7 cells of 143.8, 75.4, and 85.6 μg/mL at 24, 48, and 72 h, respectively [118]. It is reported that FA can take on an anti-cancer role by mediating different targets.

2.4.1. p53

Genes involved in cancer development can be divided into oncogenes and tumor suppressor genes [119]. As one of the most important tumor suppressor genes involved in cell cycle control and induction of apoptosis following DNA damage and oncogene activation, p53 is activated, stabilized, and accumulated by post-translational modifications in the cell, which reduces the risk of tumorigenesis [120]. Many different cell biological responses are induced by p53, such as G1 arrest, senescence, and apoptosis [121]. p53 has been shown to promote apoptosis, whereas overexpression of cyclin D1 leads to a shorter duration of the G1 phase and accelerates cancer progression [122]. The protein level of cyclin D1 is decreased and p53 is upregulated after FA treatment, indicating that FA arrests the G0/G1 phase in human cervical cancer cells [115,123]. Additionally, FA can ameliorate placental apoptosis in a preeclampsia rat model by facilitating B cell lymphoma-2 (an anti-apoptotic protein) expression and decreasing the expression of Bcl2-associated X protein (Bax), which is a pro-apoptotic effector (Figure 2D) [124]. FA derivatives also inhibit cell proliferation, and also induce cell cycle changes and apoptosis. Hexyl ferulate acts mainly through a mitochondrial pathway involving p53 and Bax, resulting in increased cell death and restrained development of cancer [125].

2.4.2. Extracellular Signal-Regulated Kinase

Extracellular signal-regulated kinase (ERK) targets different molecules to stimulate cell proliferation and plays a crucial role in regulating physiological processes such as cell growth, proliferation, and apoptosis. The inactivation of ERK can upregulate or downregulate expression of pro-apoptotic proteins and survival proteins, respectively [119,126]. Sustained activation of ERK can promote proliferation and migration of tumor cells. FA is capable of inhibiting the overexpression of p-ERK1/2 and ERK1/2 proteins, thus exerting a proliferation-inhibiting effect (Figure 2D) [127].
FA inactivates ERK1/2 and c-Jun N-terminal kinase (JNK), so as to inhibit angiotensin II-induced proliferation of vascular smooth muscle cells, thereby reducing the expression of cell cyclin D1 and regulating the process of cells from the G1 to S phase [128,129]. Furthermore, a FA derivative activates the JNK signaling pathway, while inhibiting the ERK signaling pathway, and induces apoptosis in lung cancer cells [130].

2.4.3. Protein Kinase B

Protein kinase B (AKT) is a serine threonine kinase that mediates various biological functions such as cell proliferation, survival, glucose metabolism, protein synthesis, genome stabilization, and inhibition of apoptosis in response to different growth factors and extracellular stimuli. Many studies have shown that one of the corporate molecular features of human malignancies is excessive activation of AKT, leading to tumor aggressiveness and drug resistance [131,132]. Treatment with FA inhibits the proliferation of osteosarcoma cells (IC50 = 59.88 μM) and promotes the apoptosis by downregulating the expression and activation of AKT (Figure 2D) [133,134]. Additionally, inhibitory effects of FA on angiogenesis and cell proliferation have been demonstrated in vitro and in vivo, with significant inhibition of FGFR1-mediated AKT phosphorylation by FA [135,136]. FGFR1 kinase activity has been directly inhibited by FA in a dose-dependent manner with an IC50 of approximately 3.78 μM.

2.4.4. Programmed Cell Death

Programmed cell death (PCD) is a process that regulates the elimination of unwanted or damaged cells in a controlled manner, which can be classified into apoptosis, necrosis, autophagy, and ferroptosis, etc. [137,138]. PCD plays an important role in maintaining tissue homeostasis and preventing tumorigenesis, but nonetheless results in various pathological conditions [139,140]. One of the PCD types associated with pulmonary diseases is ferroptosis, which is caused by an increase in iron-dependent ROS due to intracellular iron overload, leading to lipid peroxidation and cell membrane damage [141]. Ferroptosis has been shown to be involved in the pathogenesis and progression of ALI/ARDS, lung cancer, PF, and asthma [142,143,144,145]. FA has been shown to inhibit ferroptosis by modulating several key factors involved in this process, such as glutathione peroxidase 4, Nrf2, and adenosine monophosphate-activated protein kinase [146]. Another type of PCD related to pulmonary diseases is apoptosis mediated by programmed cell death 4 (PDCD4), which encodes a tumor suppressor protein that inhibits translation initiation and promotes apoptosis [147,148]. PDCD4 is frequently downregulated in lung cancer, and the expression is associated with tumor progression and prognosis [149]. FA may inhibit the degradation of PDCD4 protein by preventing its phosphorylation through the mechanistic targeting of rapamycin/ribosomal protein S6 kinase 1 [150,151].

3. Therapeutic Effects on Pulmonary Diseases

Pulmonary diseases, including IPF, asthma, lung cancer, ALI/ARDS, influenza, etc., rank amongst the most common causes of death globally [152]. Both the incidence and mortality rate of lung cancer are at the top of the list among all malignant tumors [153]. The prevalence of IPF and asthma is increasing, constituting a significant threat to public health. Available therapeutic approaches for pulmonary diseases focus on relieving symptom severity and enhancing quality of life; existing treatments are unable to achieve complete recovery of lung function [154]. As a possible treatment for end-stage pulmonary diseases, lung transplantation has a high mortality rate; thus, novel therapies are urgently needed [155,156,157]. In recent years, there has been an increasing number of studies on the therapeutic effects of FA in pulmonary diseases (Figure 3).

3.1. IPF

As a fatal and incurable lung disease with increasing incidence, IPF can be restricted by FA, which reduces the migration of inflammatory cells, deposition of excessive ECM components, and secretion of pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α [96,101,158].
Several fibrotic proteins are known to exacerbate PF, among which TGF-β enhances ECM deposition, promotes epithelial–mesenchymal transition, and induces fibroblast differentiation [159,160,161]. FA can inhibit the TGF-β1/Smad3 signaling pathway by downregulating the phosphorylation of Smad2/Smad3 and can block the TGF-β mediated downstream regulation of epithelial marker E-cadherin, so as to alleviate PF [96,162]. Of note, FA alleviates TGF-β induced ECM production through the Smad3-dependent/non-dependent pathway, i.e., MMPs (Figure 4) [163,164]. As a component of the Yangfei Huoxue Decoction, FA restrains vascularized vascular endothelial growth factor and IL-1β expression, indicating a possible protective effect in PF treatment [165,166].

3.2. Asthma

Asthma is a chronic inflammatory disorder of the respiratory tract, characterized by mucus hypersecretion, airflow limitation, bronchial hyperresponsiveness, and airway inflammation [167,168]. FA can relieve several allergic complications by exerting immunomodulatory effects, such as cutaneous anaphylaxis in an allergic mouse model, suggesting that FA is a promising candidate for the effective control of asthma [8,169]. It has also been demonstrated that FA decreases the expression of P-selectin on the platelet surface and reduces airway inflammation, which can inhibit endothelial cell adhesion and improve lung function in asthma [166,170]. FA can reduce immunoglobulin E and activate dendritic cells via enhancing the expression of CD40, then restoring the T helper cell (Th)1/Th2 imbalance [8,171]. Recently, in order to better exert the anti-asthma effect, FA is packaged into chitosan-based nanocarriers to ensure drug delivery to epithelial cells [172].

3.3. Lung Cancer

Globally, lung cancer is the most common cancer and the leading cause of cancer deaths, of which approximately 80% are non-small cell lung cancer (NSCLC) [111,173]. FA has been used to enhance the sensitivity of cancer cells to radiation, with low systemic toxicity [174,175]. FA treatment given along with radiation is able to arrest the cell cycle, increase the expression of the pro-apoptotic proteins p53 and Bax, and inhibit the anti-apoptotic capacity of A549 and NCI-H460 cells [28,176]. FA derivatives can limit the proliferation and metastasis of lung cancer by reducing the phosphorylated expression of ERK, AKT, and MAPK kinases, which have been shown to be involved in cell invasion and are associated with reduced survival rates in a variety of human malignancies [130]. Trans-FA inhibits the proliferation of H1299 lung cancer cells and induces a moderate increase in the apoptotic population by promoting phosphorylation of β-catenin at residues Thr41 and Ser45 and causing proteasomal degradation [117,177]. Intriguingly, FA is capable of inhibiting the proliferation and migration of lung cancer cells by eliminating intracellular ROS production in tumor cells and of slowing tumor progression by suppressing the adhesion and migration of A549 lung cancer cells [117,178].

3.4. ALI/ARDS

ALI and ARDS are successive lung changes arising from multifarious lung injuries with significant morbidity and mortality, which are characterized by bursts of inflammation and damaged alveolar-capillary structures [179,180]. FA can ameliorate ALI by inhibiting the NF-κB and MAPK pathways via TLR4 and consequently decreasing the expression of downstream inflammatory mediators, including TNF-α, IL-1β, IL-6, and IL-8 (Figure 5) [77,181,182]. FA also downregulates the activity of myeloperoxidase, an indicator of neutrophil infiltration [181]. As a derivative of FA, ethyl ferulate inhibits the production of inflammatory mediators in LPS-stimulated macrophages, which also block the translocation of NF-κB p65 to the nucleus and significantly reduce intracellular ROS levels [183]. The activity of superoxidase dismutase, which is an anti-oxidative enzyme that scavenges superoxide radicals, is significantly enhanced in the treatment of sodium ferulate [184]. Furthermore, FA has been found to prevent ARDS by inhibiting the expression of MAPK signaling pathway-related proteins, including p-p38, p-ERK1/2, and p-JNK (Figure 5) [185]. Growing evidence indicates that lung inflammation and injury are regulated by adenosine monophosphate-activated protein kinase [186], which can be activated by ethyl ferulate in a Nrf2/HO-1 dependent manner [183,187].

3.5. Influenza

Influenza is an acute viral respiratory infection with a high morbidity rate [188]. Current strategies for treating influenza focus on inhibiting the function of neuraminidase (NA), one of the surface proteins of the virion, which supports the release of progeny virions from the host cells and their movement to target cells [189]. FA has moderate NA inhibitory activity and shows a tendency to reduce the downstream IL-8 level in response to influenza virus infections [190]. The ring structure of FA is similar to that of the NA inhibitor oseltamivir, such as in the C1 and C5 positions [190,191]. In contrast, at the C3 position, there is no semblable amino group in FA. Therefore, slight structural modifications of FA, such as the introduction of an amino substituent into the guanidine group, could improve FA activity, which would eventually increase NA inhibition in vitro. A virus inhibition experiment shows that the FA derivative MY15 has good activity, with a median effective concentration of about 0.95 μM [190]. Beyond that, the protective immune response to influenza is controlled by the TLR7/TLR9-myeloid differentiation primary response 88 pathway, which is enhanced by sodium ferulate in mice [192].

3.6. Other Pulmonary Diseases

COPD is a progressive lung disorder characterized by oxidative stress, inflammation, endothelial dysfunction, fibrosis, and apoptosis [193,194]. As one of the bioactive components of Bu-Zhong-Yi-Qi-Tang, FA has been shown to reduce the levels of TNF-α and IL-6, as well as prevent neutrophil and macrophage infiltration by downregulating cell-adhesion molecules, such as P-selectin, which may help to relieve the symptoms of COPD [195].
Pneumoconiosis is a group of serious occupational diseases which are associated with the inhalation of mine dust and the corresponding reaction of the lung tissue [196,197]. Sodium ferulate can inhibit the activation of the TGF-β1/neutrophilic alkaline phosphatase 3/α-smooth muscle actin pathway, which provides a potential therapeutic strategy for silicosis-associated PF [197].
As one of the major components of Rhodiola algida, which prevents high latitude sickness clinically, FA is effective in hypoxia-induced pulmonary arterial hypertension animals [198]. Similarly, sodium ferulate has been used clinically in the treatment of pulmonary hypertension with satisfactory results [199].
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) caused the outbreak of coronavirus disease 2019, against which the therapeutic target is the SARS-CoV-2 main protease [200]. Some FA derivatives, such as FA rutinoside and raffinose ferulate, show comparable or better binding affinities for the main protease of SARS-CoV-2 as confirmed inhibitors, and might have antiviral potential against coronavirus disease 2019 [201].

4. Toxicological Effects

At a dose of 300 μg/mL, FA has no effect on the cell count and viability of platelets, leukocytes, and erythrocytes. There is hardly any toxicity of FA to NIH-3T3 and 3T3-L1 cells at the concentration of 500 μg/mL [202]. Nevertheless, studies have demonstrated the renal-damaging effect of FA when used for the 28-week cure of chronic kidney diseases [203]. Intriguingly, the toxicity of FA is seasonally dependent, being more toxic in May, June, and September, which may be related to abiotic factors, including carbon dioxide, temperature, and pH [204].

5. Conclusions and Future Directions

FA and its derivatives are currently being used with breakthrough results in various fields. Modern pharmacological studies have proven that FA has a variety of effects, such as anti-oxidative, anti-inflammatory, anti-fibrotic and anti-cancer effects. Firstly, as a free radical scavenger, FA significantly downregulates ROS expression and activates Nrf2/HO-1 signaling, exerting anti-oxidative effects. Secondly, FA acts as an anti-inflammatory agent by inhibiting the p38 MAPK, NF-κB, and JAK/STAT pathways. The TGF-β/Smad signaling pathway can be blocked by FA, which plays an anti-fibrotic role. The MMPs/TIMPs system can be regulated by FA to inhibit the expression of MMP2 and MMP9, thereby enhancing the anti-fibrotic effect. Ultimately, the anti-cancer effect of FA is closely connected with p53 upregulation, Bax downregulation, and inactivation of ERK and AKT.
In addition to providing a summary of the pharmacological mechanisms of FA, we also consider the therapeutic advances in pulmonary diseases. FA has shown significant promise in the field of lung disease treatment. Initially, by blocking the TGF-β1/Smad3 signaling pathway and inhibiting MMPs expression, FA plays a role in improving IPF. Furthermore, FA reduces the expression of P-selectin and restores the Th1/Th2 imbalance, exerting an anti-asthma effect. With regards to lung cancer, the expression of p53 and ROS production are regulated by FA. Thirdly, FA has prevented ARDS through inhibiting the expression of MAPK signaling pathway-related proteins, including p-p38, p-ERK1/2, and p-JNK. Additionally, FA has a moderate NA inhibitory activity, which shows a tendency to reduce downstream IL-8 levels in response to influenza virus infections.
One of the main limitations of the clinical application of FA to date has been low bioavailability, and most of the technological strategies used to improve the oral bioavailability of FA are based on lipid delivery systems [205]. Both nanostructured lipid carriers and solid lipid nanoparticles can enhance the oral bioavailability of FA [206]. In addition, a major issue in the drug-discovery process is toxicity. Thus, FA, with its low toxicity properties, is a very valuable natural compound with potential for the treatment of pulmonary diseases.
This review discusses the pharmacological effects, applications in pulmonary diseases, and toxicology of FA to heighten our understanding of existing research. More in-depth research is needed to explore the molecular mechanisms of FA, in order to provide an efficient scientific basis for enlarging the scope of clinical treatment and exploiting the potential of FA in drug applications.

Author Contributions

Y.Z. wrote the review; T.W. and Y.F. designed the framework; T.Y. and Y.D. designed the figures; H.N. revised the draft of manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The study is financially supported by the National Natural Science Foundation of China (82170093).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts 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.

References

  1. Wakefield, J.; Hassan, H.M.; Jaspars, M.; Ebel, R.; Rateb, M.E. Dual Induction of New Microbial Secondary Metabolites by Fungal Bacterial Co-cultivation. Front. Microbiol. 2017, 8, 1284. [Google Scholar] [CrossRef] [PubMed]
  2. Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. Ferulic Acid from Plant Biomass: A Phytochemical with Promising Antiviral Properties. Front. Nutr. 2021, 8, 777576. [Google Scholar] [CrossRef] [PubMed]
  3. Bento-Silva, A.; Patto, M.C.V.; do Rosario Bronze, M. Relevance, structure and analysis of ferulic acid in maize cell walls. Food Chem. 2018, 246, 360–378. [Google Scholar] [CrossRef] [PubMed]
  4. Zhao, Z.; Moghadasian, M.H. Chemistry, natural sources, dietary intake and pharmacokinetic properties of ferulic acid: A review. Food Chem. 2008, 109, 691–702. [Google Scholar] [CrossRef]
  5. Smith, B.G.; Harris, P.J. Ferulic acid is esterified to glucuronoarabinoxylans in pineapple cell walls. Phytochemistry 2001, 56, 513–519. [Google Scholar] [CrossRef]
  6. Kumar, N.; Pruthi, V. Potential applications of ferulic acid from natural sources. Biotechnol. Rep. 2014, 4, 86–93. [Google Scholar] [CrossRef]
  7. Wang, Y.; Chen, X.; Huang, Z.; Chen, D.; Yu, B.; Yu, J.; Chen, H.; He, J.; Luo, Y.; Zheng, P. Dietary ferulic acid supplementation improves antioxidant capacity and lipid metabolism in weaned piglets. Nutrients 2020, 12, 3811. [Google Scholar] [CrossRef]
  8. Lee, C.C.; Wang, C.C.; Huang, H.M.; Lin, C.L.; Leu, S.J.; Lee, Y.L. Ferulic acid induces Th1 responses by modulating the function of dendritic cells and ameliorates Th2-mediated allergic airway inflammation in mice. Evid. Based Complement. Alternat. Med. 2015, 2015, 678487. [Google Scholar] [CrossRef]
  9. Ibitoye, O.B.; Ajiboye, T.O. Ferulic acid potentiates the antibacterial activity of quinolone-based antibiotics against Acinetobacter baumannii. Microb. Pathog. 2019, 126, 393–398. [Google Scholar] [CrossRef]
  10. Pellerito, C.; Emanuele, S.; Ferrante, F.; Celesia, A.; Giuliano, M.; Fiore, T. Tributyltin(IV) ferulate, a novel synthetic ferulic acid derivative, induces autophagic cell death in colon cancer cells: From chemical synthesis to biochemical effects. J. Inorg. Biochem. 2020, 205, 110999. [Google Scholar] [CrossRef]
  11. Kuang, J.; Wei, X.L.; Xie, M. The effect of sodium ferulate in experimental pulmonary fibrosis via NALP3 inflammasome. Sichuan Da Xue Xue Bao Yi Xue Ban 2017, 48, 503–508. [Google Scholar]
  12. Zeni, A.L.B.; Camargo, A.; Dalmagro, A.P. Ferulic acid reverses depression-like behavior and oxidative stress induced by chronic corticosterone treatment in mice. Steroids 2017, 125, 131–136. [Google Scholar] [CrossRef] [PubMed]
  13. Panneerselvam, L.; Subbiah, K.; Arumugam, A.; Senapathy, J.G. Ferulic acid modulates fluoride-induced oxidative hepatotoxicity in male Wistar rats. Biol. Trace Elem. Res. 2013, 151, 85–91. [Google Scholar] [CrossRef]
  14. Sadar, S.S.; Vyawahare, N.S.; Bodhankar, S.L. Ferulic acid ameliorates TNBS-induced ulcerative colitis through modulation of cytokines, oxidative stress, iNOs, COX-2, and apoptosis in laboratory rats. EXCLI J. 2016, 15, 482–499. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, E.J.; Wu, M.Y.; Lu, J.H. Ferulic Acid in Animal Models of Alzheimer’s Disease: A Systematic Review of Preclinical Studies. Cells 2021, 10, 2653. [Google Scholar] [CrossRef]
  16. El-Bassossy, H.; Badawy, D.; Neamatallah, T.; Fahmy, A. Ferulic acid, a natural polyphenol, alleviates insulin resistance and hypertension in fructose fed rats: Effect on endothelial-dependent relaxation. Chem-Biol. Interact. 2016, 254, 191–197. [Google Scholar] [CrossRef]
  17. Gu, Y.Y.; Zhang, Y.X.; Li, M.; Huang, Z.Y.; Jiang, J.; Chen, Y.H.; Chen, J.Q.; Jia, Y.H.; Zhang, L.H.; Zhou, F.H. Ferulic Acid Ameliorates Atherosclerotic Injury by Modulating Gut Microbiota and Lipid Metabolism. Front. Pharmacol. 2021, 12, 621339. [Google Scholar] [CrossRef]
  18. Zhang, S.H.; Liu, D.H.; Hu, Q.Y.; Zhu, J.L.; Wang, S.Q.; Zhou, S.B. Ferulic acid ameliorates pentylenetetrazol-induced seizures by reducing neuron cell death. Epilepsy Res. 2019, 156, 106183. [Google Scholar] [CrossRef]
  19. Zhang, X.J.; Cui, Z.H.; Zhao, Y.X.; He, T.T.; Wang, L.; Liang, X.W. Ferulic acid ameliorates isoproterenol-induced heart failure by decreasing oxidative stress and inhibiting cardiocyte apoptosis via activating Nrf2 signaling pathway in rats. Biol. Pharm. Bull. 2021, 44, 396–403. [Google Scholar] [CrossRef]
  20. Song, Y.; Zhang, C.; Wang, C.; Zhao, L.; Wang, Z.; Dai, Z.; Lin, S.; Kang, H.; Ma, X. Ferulic acid against cyclophosphamide-induced heart toxicity in mice by inhibiting NF-kappaB pathway. Evid. Based Complement. Alternat. Med. 2016, 2016, 1261270. [Google Scholar] [CrossRef]
  21. Roghani, M.; Kalantari, H.; Khodayar, M.J.; Khorsandi, L.; Kalantar, M.; Goudarzi, M.; Kalantar, H. Alleviation of Liver Dysfunction, Oxidative Stress and Inflammation Underlies the Protective Effect of Ferulic Acid in Methotrexate-Induced Hepatotoxicity. Drug. Des. Devel Ther. 2020, 14, 1933–1941. [Google Scholar] [CrossRef] [PubMed]
  22. Kanski, J.; Aksenova, M.; Stoyanova, A.; Butterfield, D.A. Ferulic acid antioxidant protection against hydroxyl and peroxyl radical oxidation in synaptosomal and neuronal cell culture systems in vitro: Structure-activity studies. J. Nutr. Biochem. 2002, 13, 273–281. [Google Scholar] [CrossRef] [PubMed]
  23. McGuinness, A.J.; Sapey, E. Oxidative Stress in COPD: Sources, Markers, and Potential Mechanisms. J. Clin. Med. 2017, 6, 21. [Google Scholar] [CrossRef]
  24. Barnes, P.J. Oxidative stress-based therapeutics in COPD. Redox Biol. 2020, 33, 101544. [Google Scholar] [CrossRef] [PubMed]
  25. Chowdhury, S.; Ghosh, S.; Das, A.K.; Sil, P.C. Ferulic Acid Protects Hyperglycemia-Induced Kidney Damage by Regulating Oxidative Insult, Inflammation and Autophagy. Front. Pharmacol. 2019, 10, 27. [Google Scholar] [CrossRef] [PubMed]
  26. Sun, X.; Ma, L.; Li, X.; Wang, J.; Li, Y.; Huang, Z. Ferulic acid alleviates retinal neovascularization by modulating microglia/macrophage polarization through the ROS/NF-kappaB axis. Front. Immunol. 2022, 13, 976729. [Google Scholar] [CrossRef]
  27. Choi, R.; Kim, B.H.; Naowaboot, J.; Lee, M.Y.; Hyun, M.R.; Cho, E.J.; Lee, E.S.; Lee, E.Y.; Yang, Y.C.; Chung, C.H. Effects of ferulic acid on diabetic nephropathy in a rat model of type 2 diabetes. Exp. Mol. Med. 2011, 43, 676–683. [Google Scholar] [CrossRef]
  28. Das, U.; Manna, K.; Adhikary, A.; Mishra, S.; Das Saha, K.; Sharma, R.D.; Majumder, B.; Dey, S. Ferulic acid enhances the radiation sensitivity of lung and liver carcinoma cells by collapsing redox homeostasis: Mechanistic involvement of Akt/p38 MAPK signalling pathway. Free. Radic. Res. 2019, 53, 944–967. [Google Scholar] [CrossRef]
  29. Alam, M.A. Anti-hypertensive Effect of Cereal Antioxidant Ferulic Acid and Its Mechanism of Action. Front. Nutr. 2019, 6, 121. [Google Scholar] [CrossRef] [PubMed]
  30. Maurya, D.K.; Devasagayam, T.P. Antioxidant and prooxidant nature of hydroxycinnamic acid derivatives ferulic and caffeic acids. Food Chem. Toxicol. 2010, 48, 3369–3373. [Google Scholar] [CrossRef]
  31. Mimura, T.; Yazaki, K.; Sawaki, K.; Ozawa, T.; Kawaguchi, M. Hydroxyl radical scavenging effects of guaiacol used in traditional dental pulp sedation: Reaction kinetic study. Biomed. Res 2005, 26, 139–145. [Google Scholar] [CrossRef]
  32. Sumanont, Y.; Murakami, Y.; Tohda, M.; Vajragupta, O.; Matsumoto, K.; Watanabe, H. Evaluation of the nitric oxide radical scavenging activity of manganese complexes of curcumin and its derivative. Biol. Pharm. Bull. 2004, 27, 170–173. [Google Scholar] [CrossRef]
  33. Park, B.H.; Fikrig, S.M.; Smithwick, E.M. Infection and nitroblue-tetrazolium reduction by neutrophils. A diagnostic acid. Lancet 1968, 2, 532–534. [Google Scholar] [CrossRef] [PubMed]
  34. Aebi, H. Catalase in vitro. Methods Enzymol. 1984, 105, 121–126. [Google Scholar] [CrossRef]
  35. Yeh, C.T.; Ching, L.C.; Yen, G.C. Inducing gene expression of cardiac antioxidant enzymes by dietary phenolic acids in rats. J. Nutr. Biochem. 2009, 20, 163–171. [Google Scholar] [CrossRef] [PubMed]
  36. Kaul, A.; Khanduja, K.L. Plant polyphenols inhibit benzoyl peroxide-induced superoxide anion radical production and diacylglyceride formation in murine peritoneal macrophages. Nutr. Cancer 1999, 35, 207–211. [Google Scholar] [CrossRef] [PubMed]
  37. Khanduja, K.L.; Avti, P.K.; Kumar, S.; Mittal, N.; Sohi, K.K.; Pathak, C.M. Anti-apoptotic activity of caffeic acid, ellagic acid and ferulic acid in normal human peripheral blood mononuclear cells: A Bcl-2 independent mechanism. Biochim. Biophys. Acta 2006, 1760, 283–289. [Google Scholar] [CrossRef]
  38. Trombino, S.; Cassano, R.; Ferrarelli, T.; Barone, E.; Picci, N.; Mancuso, C. Trans-ferulic acid-based solid lipid nanoparticles and their antioxidant effect in rat brain microsomes. Colloids Surf. B Biointerfaces 2013, 109, 273–279. [Google Scholar] [CrossRef]
  39. Fan, J.; Lv, H.; Li, J.; Che, Y.; Xu, B.; Tao, Z.; Jiang, W. Roles of Nrf2/HO-1 and HIF-1alpha/VEGF in lung tissue injury and repair following cerebral ischemia/reperfusion injury. J. Cell. Physiol. 2019, 234, 7695–7707. [Google Scholar] [CrossRef]
  40. Zhang, Q.; Liu, J.; Duan, H.X.Y.; Li, R.L.; Peng, W.; Wu, C.J. Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef]
  41. Feng, Y.; Cui, R.X.; Li, Z.Y.; Zhang, X.; Jia, Y.F.; Zhang, X.; Shi, J.H.; Qu, K.; Liu, C.; Zhang, J.Y. Methane Alleviates Acetaminophen-Induced Liver Injury by Inhibiting Inflammation, Oxidative Stress, Endoplasmic Reticulum Stress, and Apoptosis through the Nrf2/HO-1/NQO1 Signaling Pathway. Oxidative Med. Cell. Longev. 2019, 2019, 7067619. [Google Scholar] [CrossRef] [PubMed]
  42. Arab, H.H.; Al-Shorbagy, M.Y.; Saad, M.A. Activation of autophagy and suppression of apoptosis by dapagliflozin attenuates experimental inflammatory bowel disease in rats: Targeting AMPK/mTOR, HMGB1/RAGE and Nrf2/HO-1 pathways. Chem.-Biol. Interact. 2021, 335, 109368. [Google Scholar] [CrossRef]
  43. Chan, K.; Kan, Y.W. Nrf2 is essential for protection against acute pulmonary injury in mice. Proc. Natl. Acad. Sci. USA 1999, 96, 12731–12736. [Google Scholar] [CrossRef] [PubMed]
  44. He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef]
  45. Chen, Q.M.; Maltagliati, A.J. Nrf2 at the heart of oxidative stress and cardiac protection. Physiol. Genom. 2018, 50, 77–97. [Google Scholar] [CrossRef]
  46. Ma, Z.C.; Hong, Q.; Wang, Y.G.; Liang, Q.D.; Tan, H.L.; Xiao, C.R.; Tang, X.L.; Shao, S.; Zhou, S.S.; Gao, Y. Ferulic acid induces heme oxygenase-1 via activation of ERK and Nrf2. Drug Discov. Ther. 2011, 5, 299–305. [Google Scholar] [CrossRef]
  47. He, S.; Guo, Y.; Zhao, J.; Xu, X.; Song, J.; Wang, N.; Liu, Q. Ferulic acid protects against heat stress-induced intestinal epithelial barrier dysfunction in IEC-6 cells via the PI3K/Akt-mediated Nrf2/HO-1 signaling pathway. Int. J. Hyperth. 2019, 35, 112–121. [Google Scholar] [CrossRef] [PubMed]
  48. Kose, T.; Sharp, P.A.; Latunde-Dada, G.O. Upregulation of Nrf2 Signalling and the Inhibition of Erastin-Induced Ferroptosis by Ferulic Acid in MIN6 Cells. Int. J. Mol. Sci. 2022, 23, 15886. [Google Scholar] [CrossRef]
  49. Nouri, A.; Ghatreh-Samani, K.; Amini-Khoei, H.; Mohammadi, A.; Heidarian, E.; Najafi, M. Ferulic acid prevents cyclosporine-induced nephrotoxicity in rats through exerting anti-oxidant and anti-inflammatory effects via activation of Nrf2/HO-1 signaling and suppression of NF-kappaB/TNF-alpha axis. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2022, 395, 387–395. [Google Scholar] [CrossRef]
  50. Mahmoud, A.M.; Hussein, O.E.; Hozayen, W.G.; Bin-Jumah, M.; Abd El-Twab, S.M. Ferulic acid prevents oxidative stress, inflammation, and liver injury via upregulation of Nrf2/HO-1 signaling in methotrexate-induced rats. Environ. Sci. Pollut. Res. 2020, 27, 7910–7921. [Google Scholar] [CrossRef]
  51. Tang, X.; Liu, J.; Yao, S.; Zheng, J.; Gong, X.; Xiao, B. Ferulic acid alleviates alveolar epithelial barrier dysfunction in sepsis-induced acute lung injury by activating the Nrf2/HO-1 pathway and inhibiting ferroptosis. Pharm. Biol. 2022, 60, 2286–2294. [Google Scholar] [CrossRef]
  52. Chen, Y.; Zhu, L.; Meng, H.; Sun, X.; Xue, C. Ferulic acid protects human lens epithelial cells against ionizing radiation-induced oxidative damage by activating Nrf2/HO-1 signal pathway. Oxid. Med. Cell. Longev. 2022, 2022, 6932188. [Google Scholar] [CrossRef]
  53. Liang, L.; Li, C.; Wang, Y.; Yue, Y.; Zhang, H.; Yang, M.; Cao, X.; Zhao, M.; Du, J.; Peng, M.; et al. Physalis alkekengi L. var. franchetii (Mast.) Makino: A review of the pharmacognosy, chemical constituents, pharmacological effects, quality control, and applications. Phytomedicine 2022, 105, 154328. [Google Scholar] [CrossRef] [PubMed]
  54. Afroz, R.; Tanvir, E.M.; Tania, M.; Fu, J.; Kamal, M.A.; Khan, M.A. LPS/TLR4 pathways in breast cancer: Insights into cell signalling. Curr. Med. Chem. 2022, 29, 2274–2289. [Google Scholar] [CrossRef]
  55. Liu, Y.M.; Shen, J.D.; Xu, L.P.; Li, H.B.; Li, Y.C.; Yi, L.T. Ferulic acid inhibits neuro-inflammation in mice exposed to chronic unpredictable mild stress. Int. Immunopharmacol. 2017, 45, 128–134. [Google Scholar] [CrossRef]
  56. Yin, P.; Zhang, Z.; Li, J.; Shi, Y.; Jin, N.; Zou, W.; Gao, Q.; Wang, W.; Liu, F. Ferulic acid inhibits bovine endometrial epithelial cells against LPS-induced inflammation via suppressing NK-kappaB and MAPK pathway. Res. Vet. Sci. 2019, 126, 164–169. [Google Scholar] [CrossRef] [PubMed]
  57. Morris, R.; Kershaw, N.J.; Babon, J.J. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018, 27, 1984–2009. [Google Scholar] [CrossRef] [PubMed]
  58. Montero, P.; Milara, J.; Roger, I.; Cortijo, J. Role of JAK/STAT in Interstitial Lung Diseases; Molecular and Cellular Mechanisms. Int. J. Mol. Sci. 2021, 22, 6211. [Google Scholar] [CrossRef]
  59. Guarda, G.; So, A. Regulation of inflammasome activity. Immunology 2010, 130, 329–336. [Google Scholar] [CrossRef]
  60. Jagodzik, P.; Tajdel-Zielinska, M.; Ciesla, A.; Marczak, M.; Ludwikow, A. Mitogen-Activated Protein Kinase Cascades in Plant Hormone Signaling. Front. Plant Sci. 2018, 9, 1387. [Google Scholar] [CrossRef]
  61. Yong, H.Y.; Koh, M.S.; Moon, A. The p38 MAPK inhibitors for the treatment of inflammatory diseases and cancer. Expert Opin. Investig. Drugs 2009, 18, 1893–1905. [Google Scholar] [CrossRef] [PubMed]
  62. Guan, Z.; Buckman, S.Y.; Pentland, A.P.; Templeton, D.J.; Morrison, A.R. Induction of cyclooxygenase-2 by the activated MEKK1 → SEK1/MKK4 → p38 mitogen-activated protein kinase pathway. J. Biol. Chem. 1998, 273, 12901–12908. [Google Scholar] [CrossRef] [PubMed]
  63. Yeung, Y.T.; Aziz, F.; Guerrero-Castilla, A.; Arguelles, S. Signaling Pathways in Inflammation and Anti-inflammatory Therapies. Curr. Pharm. Des. 2018, 24, 1449–1484. [Google Scholar] [CrossRef]
  64. Pietersma, A.; Tilly, B.C.; Gaestel, M.; de Jong, N.; Lee, J.C.; Koster, J.F.; Sluiter, W. p38 mitogen activated protein kinase regulates endothelial VCAM-1 expression at the post-transcriptional level. Biochem. Biophys. Res. Commun. 1997, 230, 44–48. [Google Scholar] [CrossRef]
  65. Badger, A.M.; Cook, M.N.; Lark, M.W.; Newman-Tarr, T.M.; Swift, B.A.; Nelson, A.H.; Barone, F.C.; Kumar, S. SB 203580 inhibits p38 mitogen-activated protein kinase, nitric oxide production, and inducible nitric oxide synthase in bovine cartilage-derived chondrocytes. J. Immunol. 1998, 161, 467–473. [Google Scholar] [CrossRef]
  66. Ono, K.; Han, J. The p38 signal transduction pathway: Activation and function. Cell. Signal. 2000, 12, 1–13. [Google Scholar] [CrossRef]
  67. Zhang, D.; Jing, B.; Chen, Z.; Li, X.; Shi, H.; Zheng, Y.; Chang, S.; Zhao, G. Ferulic acid alleviates sciatica by inhibiting peripheral sensitization through the RhoA/p38MAPK signalling pathway. Phytomedicine 2022, 106, 154420. [Google Scholar] [CrossRef]
  68. Fellendorf, F.T.; Bonkat, N.; Dalkner, N.; Schonthaler, E.M.D.; Manchia, M.; Fuchs, D.; Reininghaus, E.Z. Indoleamine 2,3-dioxygenase (IDO)-activity in Severe Psychiatric Disorders: A Systemic Review. Curr. Top. Med. Chem. 2022, 22, 2107–2118. [Google Scholar] [CrossRef]
  69. Koshiguchi, M.; Komazaki, H.; Hirai, S.; Egashira, Y. Ferulic acid suppresses expression of tryptophan metabolic key enzyme indoleamine 2, 3-dioxygenase via NFkappaB and p38 MAPK in lipopolysaccharide-stimulated microglial cells. Biosci. Biotechnol. Biochem. 2017, 81, 966–971. [Google Scholar] [CrossRef]
  70. Hassanein, E.H.M.; Abdel-Wahab, B.A.; Ali, F.E.M.; Abd El-Ghafar, O.A.M.; Kozman, M.R.; Sharkawi, S.M.Z. Trans-ferulic acid ameliorates cisplatin-induced testicular damage via suppression of TLR4, P38-MAPK, and ERK1/2 signaling pathways. Environ. Sci. Pollut. Res. Int. 2021, 28, 41948–41964. [Google Scholar] [CrossRef]
  71. Caamano, J.; Hunter, C.A. NF-kappaB family of transcription factors: Central regulators of innate and adaptive immune functions. Clin. Microbiol. Rev. 2002, 15, 414–429. [Google Scholar] [CrossRef]
  72. Patel, S.; Santani, D. Role of NF-kappa B in the pathogenesis of diabetes and its associated complications. Pharmacol. Rep. 2009, 61, 595–603. [Google Scholar] [CrossRef]
  73. Shimazu, R.; Akashi, S.; Ogata, H.; Nagai, Y.; Fukudome, K.; Miyake, K.; Kimoto, M. MD-2, a molecule that confers lipopolysaccharide responsiveness on Toll-like receptor 4. J. Exp. Med. 1999, 189, 1777–1782. [Google Scholar] [CrossRef] [PubMed]
  74. Ohto, U.; Fukase, K.; Miyake, K.; Satow, Y. Crystal structures of human MD-2 and its complex with antiendotoxic lipid IVa. Science 2007, 316, 1632–1634. [Google Scholar] [CrossRef]
  75. Park, B.S.; Song, D.H.; Kim, H.M.; Choi, B.S.; Lee, H.; Lee, J.O. The structural basis of lipopolysaccharide recognition by the TLR4-MD-2 complex. Nature 2009, 458, 1191–1195. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, M.; Zhang, C.; Xu, X.; Zhao, X.; Han, Z.; Liu, D.; Bo, R.; Li, J.; Liu, Z. Ferulic acid inhibits LPS-induced apoptosis in bovine mammary epithelial cells by regulating the NF-kappaB and Nrf2 signalling pathways to restore mitochondrial dynamics and ROS generation. Vet. Res. 2021, 52, 104. [Google Scholar] [CrossRef]
  77. Rehman, S.U.; Ali, T.; Alam, S.I.; Ullah, R.; Zeb, A.; Lee, K.W.; Rutten, B.P.F.; Kim, M.O. Ferulic acid rescues LPS-induced neurotoxicity via modulation of the TLR4 receptor in the mouse hippocampus. Mol. Neurobiol. 2019, 56, 2774–2790. [Google Scholar] [CrossRef] [PubMed]
  78. Mir, S.M.; Ravuri, H.G.; Pradhan, R.K.; Narra, S.; Kumar, J.M.; Kuncha, M.; Kanjilal, S.; Sistla, R. Ferulic acid protects lipopolysaccharide-induced acute kidney injury by suppressing inflammatory events and upregulating antioxidant defenses in Balb/c mice. Biomed. Pharmacother. 2018, 100, 304–315. [Google Scholar] [CrossRef] [PubMed]
  79. Das, U.; Manna, K.; Sinha, M.; Datta, S.; Das, D.K.; Chakraborty, A.; Ghosh, M.; Saha, K.D.; Dey, S. Role of ferulic acid in the amelioration of ionizing radiation induced inflammation: A murine model. PLoS ONE 2014, 9, e97599. [Google Scholar] [CrossRef]
  80. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  81. Xin, P.; Xu, X.; Deng, C.; Liu, S.; Wang, Y.; Zhou, X.; Ma, H.; Wei, D.; Sun, S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int. Immunopharmacol. 2020, 80, 106210. [Google Scholar] [CrossRef] [PubMed]
  82. Rojas, P.; Sarmiento, M. JAK/STAT Pathway Inhibition May Be a Promising Therapy for COVID-19-Related Hyperinflammation in Hematologic Patients. Acta Haematol. 2021, 144, 314–318. [Google Scholar] [CrossRef] [PubMed]
  83. Zhu, L.; Zhang, Z.; Xia, N.; Zhang, W.; Wei, Y.; Huang, J.; Ren, Z.; Meng, F.; Yang, L. Anti-arthritic activity of ferulic acid in complete Freund’s adjuvant (CFA)-induced arthritis in rats: JAK2 inhibition. Inflammopharmacology 2020, 28, 463–473. [Google Scholar] [CrossRef]
  84. Ma, M.; Li, G.; Qi, M.; Jiang, W.; Zhou, R. Inhibition of the Inflammasome Activity of NLRP3 Attenuates HDM-Induced Allergic Asthma. Front. Immunol. 2021, 12, 718779. [Google Scholar] [CrossRef]
  85. Zhang, M.Y.; Jiang, Y.X.; Yang, Y.C.; Liu, J.Y.; Huo, C.; Ji, X.L.; Qu, Y.Q. Cigarette smoke extract induces pyroptosis in human bronchial epithelial cells through the ROS/NLRP3/caspase-1 pathway. Life Sci. 2021, 269, 119090. [Google Scholar] [CrossRef] [PubMed]
  86. Zhao, C.; Zhao, W. NLRP3 Inflammasome-A Key Player in Antiviral Responses. Front. Immunol. 2020, 11, 211. [Google Scholar] [CrossRef]
  87. Yu, S.; Qian, H.; Zhang, D.; Jiang, Z. Ferulic acid relieved ulcerative colitis by inhibiting the TXNIP/NLRP3 pathway in rats. Cell Biol. Int. 2023, 47, 417–427. [Google Scholar] [CrossRef]
  88. Liu, Y.; Shi, L.; Qiu, W.; Shi, Y. Ferulic acid exhibits anti-inflammatory effects by inducing autophagy and blocking NLRP3 inflammasome activation. Mol. Cell. Toxicol. 2022, 18, 509–519. [Google Scholar] [CrossRef]
  89. Weiskirchen, R.; Weiskirchen, S.; Tacke, F. Organ and tissue fibrosis: Molecular signals, cellular mechanisms and translational implications. Mol. Asp. Med. 2019, 65, 2–15. [Google Scholar] [CrossRef]
  90. Verrecchia, F.; Mauviel, A. Transforming growth factor-beta and fibrosis. World J. Gastroenterol. 2007, 13, 3056–3062. [Google Scholar] [CrossRef]
  91. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef] [PubMed]
  92. Zhou, F.; Shi, L.B.; Zhang, S.Y. Ovarian Fibrosis: A Phenomenon of Concern. Chin. Med. J. 2017, 130, 365–371. [Google Scholar] [CrossRef] [PubMed]
  93. Zhao, X.Y.; Chen, J.Y.; Sun, H.X.; Zhang, Y.; Zou, D.W. New insights into fibrosis from the ECM degradation perspective: The macrophage-MMP-ECM interaction. Cell Biosci. 2022, 12, 117. [Google Scholar] [CrossRef] [PubMed]
  94. Pakshir, P.; Hinz, B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol. 2018, 68–69, 81–93. [Google Scholar] [CrossRef]
  95. Xu, T.; Pan, Z.; Dong, M.; Yu, C.; Niu, Y. Ferulic acid suppresses activation of hepatic stellate cells through ERK1/2 and Smad signaling pathways in vitro. Biochem. Pharmacol. 2015, 93, 49–58. [Google Scholar] [CrossRef]
  96. Ali, S.A.; Saifi, M.A.; Pulivendala, G.; Godugu, C.; Talla, V. Ferulic acid ameliorates the progression of pulmonary fibrosis via inhibition of TGF-beta/smad signalling. Food Chem. Toxicol. 2021, 149, 111980. [Google Scholar] [CrossRef]
  97. Meng, X.M.; Nikolic-Paterson, D.J.; Lan, H.Y. TGF-beta: The master regulator of fibrosis. Nat. Rev. Nephrol. 2016, 12, 325–338. [Google Scholar] [CrossRef]
  98. Hu, H.H.; Chen, D.Q.; Wang, Y.N.; Feng, Y.L.; Cao, G.; Vaziri, N.D.; Zhao, Y.Y. New insights into TGF-beta/Smad signaling in tissue fibrosis. Chem.-Biol. Interact. 2018, 292, 76–83. [Google Scholar] [CrossRef]
  99. Chen, L.; Yang, T.; Lu, D.W.; Zhao, H.; Feng, Y.L.; Chen, H.; Chen, D.Q.; Vaziri, N.D.; Zhao, Y.Y. Central role of dysregulation of TGF-beta/Smad in CKD progression and potential targets of its treatment. Biomed. Pharmacother. 2018, 101, 670–681. [Google Scholar] [CrossRef]
  100. Wang, Y.; Zhang, X.; Mao, Y.; Liang, L.; Liu, L.; Peng, W.; Liu, H.; Xiao, Y.; Zhang, Y.; Zhang, F.; et al. Smad2 and Smad3 play antagonistic roles in high glucose-induced renal tubular fibrosis via the regulation of SnoN. Exp. Mol. Pathol. 2020, 113, 104375. [Google Scholar] [CrossRef]
  101. Wei, M.G.; Sun, W.; He, W.M.; Ni, L.; Yang, Y.Y. Ferulic acid attenuates TGF-beta1-induced renal cellular fibrosis in NRK-52E cells by inhibiting Smad/ILK/Snail pathway. Evid. Based Complement. Alternat. Med. 2015, 2015, 619720. [Google Scholar] [CrossRef] [PubMed]
  102. Mu, M.; Zuo, S.; Wu, R.M.; Deng, K.S.; Lu, S.; Zhu, J.J.; Zou, G.L.; Yang, J.; Cheng, M.L.; Zhao, X.K. Ferulic acid attenuates liver fibrosis and hepatic stellate cell activation via inhibition of TGF-beta/Smad signaling pathway. Drug Des. Dev. Ther. 2018, 12, 4107–4115. [Google Scholar] [CrossRef]
  103. Zhao, X.M.; Zhang, J.; Liang, Y.N.; Niu, Y.C. Astragaloside IV synergizes with ferulic acid to alleviate hepatic fibrosis in bile duct-ligated cirrhotic rats. Dig. Dis. Sci. 2020, 65, 2925–2936. [Google Scholar] [CrossRef]
  104. Rukkumani, R.; Priyanka, A.; Sankar, P.; Menon, V.P. Ferulic acid influences hepatic expression pattern of matrix metalloproteinases during alcohol and PUFA induced toxicity. Eur. Rev. Med. Pharmacol. Sci. 2012, 16, 2147–2153. [Google Scholar] [PubMed]
  105. Craig, V.J.; Zhang, L.; Hagood, J.S.; Owen, C.A. Matrix metalloproteinases as therapeutic targets for idiopathic pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2015, 53, 585–600. [Google Scholar] [CrossRef] [PubMed]
  106. Rohani, M.G.; Parks, W.C. Matrix remodeling by MMPs during wound repair. Matrix Biol. 2015, 44–46, 113–121. [Google Scholar] [CrossRef]
  107. Cabral-Pacheco, G.A.; Garza-Veloz, I.; Castruita-De la Rosa, C.; Ramirez-Acuña, J.M.; Perez-Romero, B.A.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef]
  108. Vandenbroucke, R.E.; Libert, C. Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat. Rev. Drug Discov. 2014, 13, 904–927. [Google Scholar] [CrossRef]
  109. Tan, Y.; Zhang, C.L.; Zhang, Y.; Dai, X.S.; Wei, Q.H.; Wei, J.H.; Xu, P.L.; Chen, Y. Combination of ferulic acid, ligustrazine and tetrahydropalmatine inhibits invasion and metastasis through MMP/TIMP signaling in endometriosis. PeerJ 2021, 9, e11664. [Google Scholar] [CrossRef]
  110. Staniforth, V.; Huang, W.C.; Aravindaram, K.; Yang, N.S. Ferulic acid, a phenolic phytochemical, inhibits UVB-induced matrix metalloproteinases in mouse skin via posttranslational mechanisms. J. Nutr. Biochem. 2012, 23, 443–451. [Google Scholar] [CrossRef]
  111. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  112. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  113. Gupta, A.; Singh, A.K.; Loka, M.; Pandey, A.K.; Bishayee, A. Ferulic acid-mediated modulation of apoptotic signaling pathways in cancer. Adv. Protein Chem. Struct. Biol. 2021, 125, 215–257. [Google Scholar] [CrossRef]
  114. Zhang, X.; Lin, D.; Jiang, R.; Li, H.; Wan, J.; Li, H. Ferulic acid exerts antitumor activity and inhibits metastasis in breast cancer cells by regulating epithelial to mesenchymal transition. Oncol. Rep. 2016, 36, 271–278. [Google Scholar] [CrossRef] [PubMed]
  115. Gao, J.H.; Yu, H.; Guo, W.K.; Kong, Y.; Gu, L.N.; Li, Q.; Yang, S.S.; Zhang, Y.Y.; Wang, Y.X. The anticancer effects of ferulic acid is associated with induction of cell cycle arrest and autophagy in cervical cancer cells. Cancer Cell Int. 2018, 18, 102. [Google Scholar] [CrossRef]
  116. Janicke, B.; Hegardt, C.; Krogh, M.; Onning, G.; Akesson, B.; Cirenajwis, H.M.; Oredsson, S.M. The antiproliferative effect of dietary fiber phenolic compounds ferulic acid and p-coumaric acid on the cell cycle of Caco-2 cells. Nutr. Cancer 2011, 63, 611–622. [Google Scholar] [CrossRef] [PubMed]
  117. Fong, Y.; Tang, C.C.; Hu, H.T.; Fang, H.Y.; Chen, B.H.; Wu, C.Y.; Yuan, S.S.; Wang, H.D.; Chen, Y.C.; Teng, Y.N.; et al. Inhibitory effect of trans-ferulic acid on proliferation and migration of human lung cancer cells accompanied with increased endogenous reactive oxygen species and beta-catenin instability. Chin. Med. 2016, 11, 45. [Google Scholar] [CrossRef]
  118. ElKhazendar, M.; Chalak, J.; El-Huneidi, W.; Vinod, A.; Abdel-Rahman, W.M.; Abu-Gharbieh, E. Antiproliferative and proapoptotic activities of ferulic acid in breast and liver cancer cell lines. Trop. J. Pharm. Res. 2019, 18, 2571–2576. [Google Scholar] [CrossRef]
  119. Vaghari-Tabari, M.; Ferns, G.A.; Qujeq, D.; Andevari, A.N.; Sabahi, Z.; Moein, S. Signaling, metabolism, and cancer: An important relationship for therapeutic intervention. J. Cell. Physiol. 2021, 236, 5512–5532. [Google Scholar] [CrossRef]
  120. Stein, Y.; Rotter, V.; Aloni-Grinstein, R. Gain-of-Function Mutant p53: All the Roads Lead to Tumorigenesis. Int. J. Mol. Sci. 2019, 20, 6197. [Google Scholar] [CrossRef]
  121. Olivares-Illana, V.; Fahraeus, R. p53 isoforms gain functions. Oncogene 2010, 29, 5113–5119. [Google Scholar] [CrossRef] [PubMed]
  122. Tashiro, E.; Tsuchiya, A.; Imoto, M. Functions of cyclin D1 as an oncogene and regulation of cyclin D1 expression. Cancer Sci. 2007, 98, 629–635. [Google Scholar] [CrossRef]
  123. Saenglee, S.; Jogloy, S.; Patanothai, A.; Leid, M.; Senawong, T. Cytotoxic effects of peanut phenolics possessing histone deacetylase inhibitory activity in breast and cervical cancer cell lines. Pharmacol. Rep. 2016, 68, 1102–1110. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, Y.; Xue, F.; Han, C.; Yang, H.; Han, L.; Li, K.; Li, J.; Xu, Q.; Li, Z.; Yuan, B.; et al. Ferulic acid ameliorated placental inflammation and apoptosis in rat with preeclampsia. Clin. Exp. Hypertens. 2019, 41, 524–530. [Google Scholar] [CrossRef] [PubMed]
  125. Serafim, T.L.; Carvalho, F.S.; Marques, M.P.; Calheiros, R.; Silva, T.; Garrido, J.; Milhazes, N.; Borges, F.; Roleira, F.; Silva, E.T.; et al. Lipophilic caffeic and ferulic acid derivatives presenting cytotoxicity against human breast cancer cells. Chem. Res. Toxicol. 2011, 24, 763–774. [Google Scholar] [CrossRef]
  126. Balmanno, K.; Cook, S.J. Tumour cell survival signalling by the ERK1/2 pathway. Cell Death Differ. 2009, 16, 368–377. [Google Scholar] [CrossRef]
  127. Li, X.; Shi, G. Therapeutic effects and mechanism of ferulic acid and icariin in mammary gland hyperplasia model rats via regulation of the ERK pathway. Ann. Transl. Med. 2021, 9, 666. [Google Scholar] [CrossRef]
  128. Hou, Y.Z.; Yang, J.; Zhao, G.R.; Yuan, Y.J. Ferulic acid inhibits vascular smooth muscle cell proliferation induced by angiotensin II. Eur. J. Pharmacol. 2004, 499, 85–90. [Google Scholar] [CrossRef]
  129. Hou, Y.; Yang, J.; Zhao, G.; Yuan, Y. Ferulic acid inhibits endothelial cell proliferation through NO down-regulating ERK1/2 pathway. J. Cell. Biochem. 2004, 93, 1203–1209. [Google Scholar] [CrossRef]
  130. Yue, S.J.; Zhang, P.X.; Zhu, Y.; Li, N.G.; Chen, Y.Y.; Li, J.J.; Zhang, S.; Jin, R.Y.; Yan, H.; Shi, X.Q.; et al. A ferulic acid derivative FXS-3 inhibits proliferation and metastasis of human lung cancer A549 cells via positive JNK signaling pathway and negative ERK/p38, AKT/mTOR and MEK/ERK signaling pathways. Molecules 2019, 24, 2165. [Google Scholar] [CrossRef]
  131. Shariati, M.; Meric-Bernstam, F. Targeting AKT for cancer therapy. Expert Opin. Investig. Drugs 2019, 28, 977–988. [Google Scholar] [CrossRef]
  132. Song, M.; Bode, A.M.; Dong, Z.; Lee, M.H. AKT as a Therapeutic Target for Cancer. Cancer Res. 2019, 79, 1019–1031. [Google Scholar] [CrossRef]
  133. Wang, T.; Gong, X.; Jiang, R.; Li, H.; Du, W.; Kuang, G. Ferulic acid inhibits proliferation and promotes apoptosis via blockage of PI3K/Akt pathway in osteosarcoma cell. Am. J. Transl. Res. 2016, 8, 968–980. [Google Scholar] [PubMed]
  134. Luo, L.; Zhu, S.; Tong, Y.; Peng, S. Ferulic Acid Induces Apoptosis of HeLa and Caski Cervical Carcinoma Cells by Down-Regulating the Phosphatidylinositol 3-Kinase (PI3K)/Akt Signaling Pathway. Med. Sci. Monit. 2020, 26, e920095. [Google Scholar] [CrossRef]
  135. Lin, C.M.; Chiu, J.H.; Wu, I.H.; Wang, B.W.; Pan, C.M.; Chen, Y.H. Ferulic acid augments angiogenesis via VEGF, PDGF and HIF-1 alpha. J. Nutr. Biochem. 2010, 21, 627–633. [Google Scholar] [CrossRef] [PubMed]
  136. Yang, G.W.; Jiang, J.S.; Lu, W.Q. Ferulic acid exerts anti-angiogenic and anti-tumor activity by targeting fibroblast growth factor receptor 1-mediated angiogenesis. Int. J. Mol. Sci. 2015, 16, 24011–24031. [Google Scholar] [CrossRef] [PubMed]
  137. Moujalled, D.; Strasser, A.; Liddell, J.R. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ. 2021, 28, 2029–2044. [Google Scholar] [CrossRef]
  138. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
  139. Mao, S.; Lv, J.; Chen, M.; Guo, N.; Fang, Y.; Tong, J.; He, X.; Wu, G.; Wang, Z. Serinc2 deficiency causes susceptibility to sepsis-associated acute lung injury. J. Inflamm. 2022, 19, 9. [Google Scholar] [CrossRef]
  140. Bader, S.M.; Cooney, J.P.; Pellegrini, M.; Doerflinger, M. Programmed cell death: The pathways to severe COVID-19? Biochem. J. 2022, 479, 609–628. [Google Scholar] [CrossRef]
  141. Xu, W.; Deng, H.; Hu, S.; Zhang, Y.; Zheng, L.; Liu, M.; Chen, Y.; Wei, J.; Yang, H.; Lv, X. Role of Ferroptosis in Lung Diseases. J. Inflamm. Res. 2021, 14, 2079–2090. [Google Scholar] [CrossRef]
  142. Liu, P.; Feng, Y.; Li, H.; Chen, X.; Wang, G.; Xu, S.; Li, Y.; Zhao, L. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell. Mol. Biol. Lett. 2020, 25, 10. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, W.J.; Sun, Y.; Bai, L.; Zhi, L.L.; Yang, Y.; Zhao, Q.Z.; Chen, C.Q.; Qi, Y.F.; Gao, W.T.; He, W.X.; et al. RBMS1 regulates lung cancer ferroptosis through translational control of SLC7A11. J. Clin. Investig. 2021, 131, e152067. [Google Scholar] [CrossRef]
  144. He, J.; Li, X.; Yu, M. Bioinformatics Analysis Identifies Potential Ferroptosis Key Genes in the Pathogenesis of Pulmonary Fibrosis. Front. Genet. 2021, 12, 788417. [Google Scholar] [CrossRef] [PubMed]
  145. Lv, X.; Dong, M.; Tang, W.; Qin, J.; Wang, W.; Li, M.; Teng, F.; Yi, L.; Dong, J.; Wei, Y. Ferroptosis, novel therapeutics in asthma. Biomed. Pharmacother. 2022, 153, 113516. [Google Scholar] [CrossRef]
  146. Liu, X.L.; Qi, K.; Gong, Y.; Long, X.; Zhu, S.Q.; Lu, F.; Lin, K.; Xu, J.J. Ferulic Acid Alleviates Myocardial Ischemia Reperfusion Injury Via Upregulating AMPK alpha 2 Expression-Mediated Ferroptosis Depression. J. Cardiovasc. Pharm. 2022, 79, 489–500. [Google Scholar] [CrossRef]
  147. Palamarchuk, A.; Efanov, A.; Maximov, V.; Aqeilan, R.I.; Croce, C.M.; Pekarsky, Y. Akt phosphorylates and regulates Pdcd4 tumor suppressor protein. Cancer Res. 2005, 65, 11282–11286. [Google Scholar] [CrossRef]
  148. Lu, K.; Chen, Q.; Li, M.; He, L.; Riaz, F.; Zhang, T.; Li, D. Programmed cell death factor 4 (PDCD4), a novel therapy target for metabolic diseases besides cancer. Free Radic. Biol. Med. 2020, 159, 150–163. [Google Scholar] [CrossRef]
  149. Bitomsky, N.; Bohm, M.; Klempnauer, K.H. Transformation suppressor protein Pdcd4 interferes with JNK-mediated phosphorylation of c-Jun and recruitment of the coactivator p300 by c-Jun. Oncogene 2004, 23, 7484–7493. [Google Scholar] [CrossRef]
  150. Bianchi, S.; Giovannini, L. Inhibition of mTOR/S6K1/4E-BP1 Signaling by Nutraceutical SIRT1 Modulators. Nutr. Cancer 2018, 70, 490–501. [Google Scholar] [CrossRef]
  151. Dorrello, N.V.; Peschiaroli, A.; Guardavaccaro, D.; Colburn, N.H.; Sherman, N.E.; Pagano, M. S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth. Science 2006, 314, 467–471. [Google Scholar] [CrossRef]
  152. Mortality, G.B.D.; Causes of Death, C. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015, 385, 117–171. [Google Scholar] [CrossRef]
  153. Zeng, Y.; Wang, X.; Yin, B.; Xia, G.; Shen, Z.; Gu, W.; Wu, M. Role of the stromal cell derived factor-1/CXC chemokine receptor 4 axis in the invasion and metastasis of lung cancer and mechanism. J. Thorac. Dis. 2017, 9, 4947–4959. [Google Scholar] [CrossRef] [PubMed]
  154. Weiss, D.J. Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010-2012. Ann. Am. Thorac. Soc. 2013, 10, S45–S97. [Google Scholar] [CrossRef] [PubMed]
  155. Weiss, D.J. Concise review: Current status of stem cells and regenerative medicine in lung biology and diseases. Stem Cells 2014, 32, 16–25. [Google Scholar] [CrossRef]
  156. Bos, S.; Vos, R.; Van Raemdonck, D.E.; Verleden, G.M. Survival in adult lung transplantation: Where are we in 2020? Curr. Opin. Organ Transplant. 2020, 25, 268–273. [Google Scholar] [CrossRef] [PubMed]
  157. Cui, L.; Chen, S.Y.; Lerbs, T.; Lee, J.W.; Domizi, P.; Gordon, S.; Kim, Y.H.; Nolan, G.; Betancur, P.; Wernig, G. Activation of JUN in fibroblasts promotes pro-fibrotic programme and modulates protective immunity. Nat. Commun. 2020, 11, 2795. [Google Scholar] [CrossRef]
  158. George, P.M.; Patterson, C.M.; Reed, A.K.; Thillai, M. Lung transplantation for idiopathic pulmonary fibrosis. Lancet Respir. Med. 2019, 7, 271–282. [Google Scholar] [CrossRef]
  159. Wynn, T.A. Integrating mechanisms of pulmonary fibrosis. J. Exp. Med. 2011, 208, 1339–1350. [Google Scholar] [CrossRef]
  160. Inui, N.; Sakai, S.; Kitagawa, M. Molecular pathogenesis of pulmonary fibrosis, with focus on pathways related to TGF-beta and the ubiquitin-proteasome pathway. Int. J. Mol. Sci. 2021, 22, 6107. [Google Scholar] [CrossRef]
  161. George, P.M.; Wells, A.U.; Jenkins, R.G. Pulmonary fibrosis and COVID-19: The potential role for antifibrotic therapy. Lancet Respir. Med. 2020, 8, 807–815. [Google Scholar] [CrossRef]
  162. Tong, J.; Wu, Z.; Wang, Y.; Hao, Q.; Liu, H.; Cao, F.; Jiao, Y. Astragaloside IV synergizing with ferulic acid ameliorates pulmonary fibrosis by TGF-beta1/Smad3 signaling. Evid. Based Complement. Altern. Med. 2021, 2021, 8845798. [Google Scholar] [CrossRef]
  163. Samarakoon, R.; Overstreet, J.M.; Higgins, S.P.; Higgins, P.J. TGF-beta1 --> SMAD/p53/USF2 --> PAI-1 transcriptional axis in ureteral obstruction-induced renal fibrosis. Cell Tissue Res. 2012, 347, 117–128. [Google Scholar] [CrossRef] [PubMed]
  164. Meng, X.M.; Tang, P.M.; Li, J.; Lan, H.Y. TGF-beta/Smad signaling in renal fibrosis. Front. Physiol. 2015, 6, 82. [Google Scholar] [CrossRef]
  165. Chen, H.; Liu, X.; Su, Y.; Chang, K.; Wu, H.; Su, G.; Gong, J. Notch signaling pathway mediates the immunomodulatory mechanism of Yangfei Huoxue decoction alleviating bleomycin-induced pulmonary fibrosis in rats. J. Tradit. Chin. Med. 2020, 40, 204–211. [Google Scholar]
  166. Liu, X.; Chen, H.; Su, G.; Song, P.; Jiang, M.; Gong, J. An animal research and a chemical composition analysis of a Chinese prescription for pulmonary fibrosis: Yangfei Huoxue Decoction. J. Ethnopharmacol. 2019, 245, 112126. [Google Scholar] [CrossRef] [PubMed]
  167. Mims, J.W. Asthma: Definitions and pathophysiology. Int. Forum Allergy Rhinol. 2015, 5, S2–S6. [Google Scholar] [CrossRef] [PubMed]
  168. Pavord, I.D.; Beasley, R.; Agusti, A.; Anderson, G.P.; Bel, E.; Brusselle, G.; Cullinan, P.; Custovic, A.; Ducharme, F.M.; Fahy, J.V.; et al. After asthma: Redefining airways diseases. Lancet 2018, 391, 350–400. [Google Scholar] [CrossRef]
  169. Brugiolo, A.S.S.; Gouveia, A.C.C.; de Souza Alves, C.C.; de Castro, E.S.F.M.; de Oliveira, É.E.; Ferreira, A.P. Ferulic acid supresses Th2 immune response and prevents remodeling in ovalbumin-induced pulmonary allergy associated with inhibition of epithelial-derived cytokines. Pulm. Pharmacol. Ther. 2017, 45, 202–209. [Google Scholar] [CrossRef]
  170. Baeza, G.; Bachmair, E.M.; Wood, S.; Mateos, R.; Bravo, L.; de Roos, B. The colonic metabolites dihydrocaffeic acid and dihydroferulic acid are more effective inhibitors of in vitro platelet activation than their phenolic precursors. Food Funct. 2017, 8, 1333–1342. [Google Scholar] [CrossRef]
  171. Morianos, I.; Semitekolou, M. Dendritic cells: Critical regulators of allergic asthma. Int. J. Mol. Sci. 2020, 21, 7930. [Google Scholar] [CrossRef] [PubMed]
  172. Dhayanandamoorthy, Y.; Antoniraj, M.G.; Kandregula, C.A.B.; Kandasamy, R. Aerosolized hyaluronic acid decorated, ferulic acid loaded chitosan nanoparticle: A promising asthma control strategy. Int. J. Pharm. 2020, 591, 119958. [Google Scholar] [CrossRef]
  173. Mao, Y.; Yang, D.; He, J.; Krasna, M.J. Epidemiology of lung cancer. Surg. Oncol. Clin. N. Am. 2016, 25, 439–445. [Google Scholar] [CrossRef] [PubMed]
  174. Das, U.; Manna, K.; Khan, A.; Sinha, M.; Biswas, S.; Sengupta, A.; Chakraborty, A.; Dey, S. Ferulic acid (FA) abrogates gamma-radiation induced oxidative stress and DNA damage by up-regulating nuclear translocation of Nrf2 and activation of NHEJ pathway. Free Radic. Res. 2017, 51, 47–63. [Google Scholar] [CrossRef] [PubMed]
  175. El-Mesallamy, H.O.; Gawish, R.A.; Sallam, A.M.; Fahmy, H.A.; Nada, A.S. Ferulic acid protects against radiation-induced testicular damage in male rats: Impact on SIRT1 and PARP1. Environ. Sci. Pollut. Res. Int. 2018, 25, 6218–6227. [Google Scholar] [CrossRef]
  176. Bandugula, V.R.; Rajendra, P.N. 2-Deoxy-D-glucose and ferulic acid modulates radiation response signaling in non-small cell lung cancer cells. Tumour Biol. 2013, 34, 251–259. [Google Scholar] [CrossRef]
  177. Stewart, D.J. Wnt signaling pathway in non-small cell lung cancer. J. Natl. Cancer Inst. 2014, 106, djt356. [Google Scholar] [CrossRef]
  178. Bouzaiene, N.N.; Jaziri, S.K.; Kovacic, H.; Chekir-Ghedira, L.; Ghedira, K.; Luis, J. The effects of caffeic, coumaric and ferulic acids on proliferation, superoxide production, adhesion and migration of human tumor cells in vitro. Eur. J. Pharmacol. 2015, 766, 99–105. [Google Scholar] [CrossRef]
  179. Butt, Y.; Kurdowska, A.; Allen, T.C. Acute lung injury: A clinical and molecular review. Arch. Pathol. Lab. Med. 2016, 140, 345–350. [Google Scholar] [CrossRef]
  180. Kral-Pointner, J.B.; Schrottmaier, W.C.; Horvath, V.; Datler, H.; Hell, L.; Ay, C.; Niederreiter, B.; Jilma, B.; Schmid, J.A.; Assinger, A.; et al. Myeloid but not epithelial tissue factor exerts protective anti-inflammatory effects in acid aspiration-induced acute lung injury. J. Thromb. Haemost. 2017, 15, 1625–1639. [Google Scholar] [CrossRef]
  181. Wu, X.; Lin, L.; Wu, H. Ferulic acid alleviates lipopolysaccharide-induced acute lung injury through inhibiting TLR4/NF-kappaB signaling pathway. J. Biochem. Mol. Toxicol. 2021, 35, e22664. [Google Scholar] [CrossRef]
  182. Yang, Z.; Liu, S.; Wang, Y.; Chen, Y.; Zhang, P.; Liu, Y.; Zhang, H.; Zhang, P.; Tao, Z.; Xiong, K. High expression of KITLG is a new hallmark activating the MAPK pathway in type A and AB thymoma. Thorac. Cancer 2020, 11, 1944–1954. [Google Scholar] [CrossRef] [PubMed]
  183. Wang, Y.; Zhang, X.; Li, L.; Zhang, Z.; Wei, C.; Gong, G. Ethyl ferulate contributes to the inhibition of the inflammatory responses in murine RAW 264.7 macrophage cells and acute lung injury in mice. PLoS ONE 2021, 16, e0251578. [Google Scholar] [CrossRef] [PubMed]
  184. Yuan, X.; Wang, Y.; Du, D.; Hu, Z.; Xu, M.; Xu, M.; Liu, Z. The effects of the combination of sodium ferulate and oxymatrine on lipopolysaccharide-induced acute lung injury in mice. Inflammation 2012, 35, 1161–1168. [Google Scholar] [CrossRef]
  185. Zhang, S.; Wang, P.; Zhao, P.; Wang, D.; Zhang, Y.; Wang, J.; Chen, L.; Guo, W.; Gao, H.; Jiao, Y. Pretreatment of ferulic acid attenuates inflammation and oxidative stress in a rat model of lipopolysaccharide-induced acute respiratory distress syndrome. Int. J. Immunopathol. Pharmacol. 2018, 32, 394632017750518. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, Z.; Cheng, X.; Yue, L.; Cui, W.; Zhou, W.; Gao, J.; Yao, H. Molecular pathogenesis in chronic obstructive pulmonary disease and therapeutic potential by targeting AMP-activated protein kinase. J. Cell. Physiol. 2018, 233, 1999–2006. [Google Scholar] [CrossRef]
  187. Wu, Y.X.; Wang, Y.Y.; Gao, Z.Q.; Chen, D.; Liu, G.; Wan, B.B.; Jiang, F.J.; Wei, M.X.; Zuo, J.; Zhu, J.; et al. Ethyl ferulate protects against lipopolysaccharide-induced acute lung injury by activating AMPK/Nrf2 signaling pathway. Acta Pharmacol. Sin. 2021, 42, 2069–2081. [Google Scholar] [CrossRef]
  188. Gaitonde, D.Y.; Moore, F.C.; Morgan, M.K. Influenza: Diagnosis and treatment. Am. Fam. Physician 2019, 100, 751–758. [Google Scholar]
  189. Xiao, M.; Xu, L.; Lin, D.; Lian, W.; Cui, M.; Zhang, M.; Yan, X.; Li, S.; Zhao, J.; Ye, J.; et al. Design, synthesis, and bioassay of 4-thiazolinone derivatives as influenza neuraminidase inhibitors. Eur. J. Med. Chem. 2021, 213, 113161. [Google Scholar] [CrossRef]
  190. Hariono, M.; Abdullah, N.; Damodaran, K.V.; Kamarulzaman, E.E.; Mohamed, N.; Hassan, S.S.; Shamsuddin, S.; Wahab, H.A. Potential new H1N1 neuraminidase inhibitors from ferulic acid and vanillin: Molecular modelling, synthesis and in vitro assay. Sci. Rep. 2016, 6, 38692. [Google Scholar] [CrossRef]
  191. Anuwongcharoen, N.; Shoombuatong, W.; Tantimongcolwat, T.; Prachayasittikul, V.; Nantasenamat, C. Exploring the chemical space of influenza neuraminidase inhibitors. PeerJ 2016, 4, e1958. [Google Scholar] [CrossRef]
  192. Zhu, Y.H.; Shao, Y.; Qu, X.Y.; Guo, J.; Yang, J.; Zhou, Z.; Wang, S.Q. Sodium ferulate protects against influenza virus infection by activation of the TLR7/9-MyD88-IRF7 signaling pathway and inhibition of the NF-kappa B signaling pathway. Biochem. Biophys. Res. Commun. 2019, 512, 793–798. [Google Scholar] [CrossRef]
  193. Decramer, M.; Janssens, W.; Miravitlles, M. Chronic obstructive pulmonary disease. Lancet 2012, 379, 1341–1351. [Google Scholar] [CrossRef]
  194. Wiegman, C.H.; Li, F.; Ryffel, B.; Togbe, D.; Chung, K.F. Oxidative Stress in Ozone-Induced Chronic Lung Inflammation and Emphysema: A Facet of Chronic Obstructive Pulmonary Disease. Front. Immunol. 2020, 11, 1957. [Google Scholar] [CrossRef]
  195. Hu, L.; Yamamoto, M.; Chen, J.; Duan, H.; Du, J.; He, L.; Shi, D.; Yao, X.; Nagai, T.; Kiyohara, H.; et al. Integrating network pharmacology and experimental verification to decipher the immunomodulatory effect of Bu-Zhong-Yi-Qi-Tang against poly (I:C)-induced pulmonary inflammation. Front. Pharmacol. 2022, 13, 1015486. [Google Scholar] [CrossRef]
  196. Shi, P.; Xing, X.; Xi, S.; Jing, H.; Yuan, J.; Fu, Z.; Zhao, H. Trends in global, regional and national incidence of pneumoconiosis caused by different aetiologies: An analysis from the Global Burden of Disease Study 2017. Occup. Environ. Med. 2020, 77, 407–414. [Google Scholar] [CrossRef] [PubMed]
  197. Han, J.; Jia, Y.; Wang, S.; Gan, X. The improvement effect of sodium ferulate on the formation of pulmonary fibrosis in silicosis mice through the neutrophil alkaline phosphatase 3 (NALP3)/transforming growth factor-beta1 (TGF-beta1)/alpha-smooth muscle actin (alpha-SMA) pathway. Med. Sci. Monit. 2021, 27, e927978. [Google Scholar] [CrossRef]
  198. Nan, X.; Su, S.; Ma, K.; Ma, X.; Wang, X.; Zhaxi, D.; Ge, R.; Li, Z.; Lu, D. Bioactive fraction of Rhodiola algida against chronic hypoxia-induced pulmonary arterial hypertension and its anti-proliferation mechanism in rats. J. Ethnopharmacol. 2018, 216, 175–183. [Google Scholar] [CrossRef] [PubMed]
  199. Zhao, Y.; Wang, H.; Zhao, J.; Wang, X.; Wang, Y.; Li, W.; Song, T.; Hao, G.; Fu, X.; Gu, X. Renal protective effect of sodium ferulate on pulmonary hypertension patients undergoing computed tomography pulmonary angiography. Pulm. Circ. 2020, 10, 1–7. [Google Scholar] [CrossRef] [PubMed]
  200. Swiderek, K.; Moliner, V. Revealing the molecular mechanisms of proteolysis of SARS-CoV-2 M(pro) by QM/MM computational methods. Chem. Sci. 2020, 11, 10626–10630. [Google Scholar] [CrossRef]
  201. Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. The Inhibitory Potential of Ferulic Acid Derivatives against the SARS-CoV-2 Main Protease: Molecular Docking, Molecular Dynamics, and ADMET Evaluation. Biomedicines 2022, 10, 1787. [Google Scholar] [CrossRef] [PubMed]
  202. Choi, J.H.; Park, J.K.; Kim, K.M.; Lee, H.J.; Kim, S. In vitro and in vivo antithrombotic and cytotoxicity effects of ferulic acid. J. Biochem. Mol. Toxicol. 2018, 32, e22004. [Google Scholar] [CrossRef] [PubMed]
  203. Peng, C.C.; Hsieh, C.L.; Wang, H.E.; Chung, J.Y.; Chen, K.C.; Peng, R.Y. Ferulic acid is nephrodamaging while gallic acid is renal protective in long term treatment of chronic kidney disease. Clin. Nutr. 2012, 31, 405–414. [Google Scholar] [CrossRef]
  204. Agrahari, P.; Singh, D.K. Seasonal variation in abiotic factors and ferulic acid toxicity in snail-attractant pellets against the intermediate host snail Lymnaea acuminata. Zoonoses Public Health 2013, 60, 478–486. [Google Scholar] [CrossRef] [PubMed]
  205. Veras, K.S.; Fachel, F.N.S.; de Araujo, B.V.; Teixeira, H.F.; Koester, L.S. Oral Pharmacokinetics of Hydroxycinnamic Acids: An Updated Review. Pharmaceutics 2022, 14, 2663. [Google Scholar] [CrossRef]
  206. Zhang, Y.; Li, Z.; Zhang, K.; Yang, G.; Wang, Z.; Zhao, J.; Hu, R.; Feng, N. Ethyl oleate-containing nanostructured lipid carriers improve oral bioavailability of trans-ferulic acid as compared with conventional solid lipid nanoparticles. Int. J. Pharm. 2016, 511, 57–64. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of FA.
Figure 1. Chemical structure of FA.
Ijms 24 08011 g001
Figure 2. A schematic drawing presenting signaling pathways affected by ferulic acid. (A) Under normal conditions, Nrf2 is sequestered in cytoplasm by keap1. Ferulic acid (FA) induces the translocation of Nrf2 from the cytoplasm to the nucleus and promotes the expression of Nrf2, which activates antioxidant response element (ARE) and increases transcription of Nrf2-regulated genes, such as HO-1. FA activates the Nrf2/HO-1 signaling, exerting an anti-oxidative effect. (B) The process of activating p38 MAPK and NF-κB signal cascades through TLR4 signaling, leading to the expression of inflammatory cytokines. NF-κB signaling requires IKK subunits, which regulate pathway activation through IκB phosphorylation. The JAK/STAT signaling pathway is activated by cytokines and STATs are dephosphorylated in the nucleus, leading to the activation of downstream inflammatory cytokines. FA acts as an anti-inflammatory agent by inhibiting the p38 MAPK, NF-κB, and JAK/STAT pathways. (C) FA can block the activation of TGF-β1/Smads signaling and reverse the nuclear translocation of Smads to resist fibrosis. Furthermore, FA has the ability to alleviate ECM by regulating MMPs. (D) FA promotes tumor regression and cell apoptosis by increasing the expression of p53 and Bax, while inhibiting proliferation by decreasing the expression of ERK and AKT. AKT: protein kinase B; Bax: Bcl2-associated X protein; ECM: extracellular matrix; ERK: extracellular signal-regulated kinase; HO-1: heme oxygenase-1; IκB: inhibitor of NF-κB; IKK: IkappaB kinase; JAK: Janus kinase; Keap1: Kelch-like-ECH-associated protein 1; MAPK: mitogen-activated protein kinase; MMPs: matrix metalloproteinases; NF-κB: nuclear factor-kappaB; Nrf2: nuclear factor erythroid-2-related factor-2; Smads: small mothers against decapentaplegics; STAT: signal transducer and activator of transcription; TGF-β1: transforming growth factor-β1; TLR4: toll-like receptor 4.
Figure 2. A schematic drawing presenting signaling pathways affected by ferulic acid. (A) Under normal conditions, Nrf2 is sequestered in cytoplasm by keap1. Ferulic acid (FA) induces the translocation of Nrf2 from the cytoplasm to the nucleus and promotes the expression of Nrf2, which activates antioxidant response element (ARE) and increases transcription of Nrf2-regulated genes, such as HO-1. FA activates the Nrf2/HO-1 signaling, exerting an anti-oxidative effect. (B) The process of activating p38 MAPK and NF-κB signal cascades through TLR4 signaling, leading to the expression of inflammatory cytokines. NF-κB signaling requires IKK subunits, which regulate pathway activation through IκB phosphorylation. The JAK/STAT signaling pathway is activated by cytokines and STATs are dephosphorylated in the nucleus, leading to the activation of downstream inflammatory cytokines. FA acts as an anti-inflammatory agent by inhibiting the p38 MAPK, NF-κB, and JAK/STAT pathways. (C) FA can block the activation of TGF-β1/Smads signaling and reverse the nuclear translocation of Smads to resist fibrosis. Furthermore, FA has the ability to alleviate ECM by regulating MMPs. (D) FA promotes tumor regression and cell apoptosis by increasing the expression of p53 and Bax, while inhibiting proliferation by decreasing the expression of ERK and AKT. AKT: protein kinase B; Bax: Bcl2-associated X protein; ECM: extracellular matrix; ERK: extracellular signal-regulated kinase; HO-1: heme oxygenase-1; IκB: inhibitor of NF-κB; IKK: IkappaB kinase; JAK: Janus kinase; Keap1: Kelch-like-ECH-associated protein 1; MAPK: mitogen-activated protein kinase; MMPs: matrix metalloproteinases; NF-κB: nuclear factor-kappaB; Nrf2: nuclear factor erythroid-2-related factor-2; Smads: small mothers against decapentaplegics; STAT: signal transducer and activator of transcription; TGF-β1: transforming growth factor-β1; TLR4: toll-like receptor 4.
Ijms 24 08011 g002
Figure 3. Pharmacological effects associated with ferulic acid in different pulmonary diseases. ALI/ARDS: acute lung injury/acute respiratory distress syndrome; Bax: Bcl2-associated X protein; ECM: extracellular matrix; HO-1: heme oxygenase-1; IL-8: interleukin-8; IPF: idiopathic pulmonary fibrosis; MAPK: mitogen-activated protein kinase; MMPs: matrix metalloproteinases; MyD88: myeloid differentiation primary response 88; NA: neuraminidase; NF-κB: nuclear factor-kappaB; Nrf2: nuclear factor erythroid-2-related factor-2; ROS: reactive oxygen species; Smads: small mothers against decapentaplegics; TGF-β1: transforming growth factor-β1; TLR7/9: toll-like receptor 7/9.
Figure 3. Pharmacological effects associated with ferulic acid in different pulmonary diseases. ALI/ARDS: acute lung injury/acute respiratory distress syndrome; Bax: Bcl2-associated X protein; ECM: extracellular matrix; HO-1: heme oxygenase-1; IL-8: interleukin-8; IPF: idiopathic pulmonary fibrosis; MAPK: mitogen-activated protein kinase; MMPs: matrix metalloproteinases; MyD88: myeloid differentiation primary response 88; NA: neuraminidase; NF-κB: nuclear factor-kappaB; Nrf2: nuclear factor erythroid-2-related factor-2; ROS: reactive oxygen species; Smads: small mothers against decapentaplegics; TGF-β1: transforming growth factor-β1; TLR7/9: toll-like receptor 7/9.
Ijms 24 08011 g003
Figure 4. Proposed signaling pathway for the effect of ferulic acid against pulmonary fibrosis. In pulmonary fibrosis, transforming growth factor-β (TGF-β) triggers small mothers against decapentaplegic (Smad) 2/3, of which the phosphorylated form compounds with Smad4, and then enters into the nucleus to regulate gene transcription by binding to cofactors or DNA sequences. In addition, TGF-β inhibits the deposition of ECM by suppressing MMPs. Ferulic acid inhibits the TGF-β signaling pathway to alleviate pulmonary fibrosis. ECM: excessive extracellular matrix; MMPs: matrix metalloproteinases.
Figure 4. Proposed signaling pathway for the effect of ferulic acid against pulmonary fibrosis. In pulmonary fibrosis, transforming growth factor-β (TGF-β) triggers small mothers against decapentaplegic (Smad) 2/3, of which the phosphorylated form compounds with Smad4, and then enters into the nucleus to regulate gene transcription by binding to cofactors or DNA sequences. In addition, TGF-β inhibits the deposition of ECM by suppressing MMPs. Ferulic acid inhibits the TGF-β signaling pathway to alleviate pulmonary fibrosis. ECM: excessive extracellular matrix; MMPs: matrix metalloproteinases.
Ijms 24 08011 g004
Figure 5. Insights into the multi-target actions of ferulic acid in acute lung injury/acute respiratory distress syndrome. Ferulic acid (FA) can directly target TLR4 and inhibit the TLR4/NF-κB pathway, decreasing the expression of phospho-NF-κB and downstream inflammatory mediators. Moreover, FA inhibits the expression of MAPK signaling pathway-related proteins. Additionally, ethyl ferulate improves acute lung injury/acute respiratory distress syndrome (ALI/ARDS) in an AMPK/Nrf2-dependent manner. AMPK: adenosine monophosphate-activated protein kinase; HO-1: heme oxygenase-1; MAPK: mitogen-activated protein kinase; Nrf2: nuclear factor erythroid-2-related factor-2; TLR4: toll-like receptor4.
Figure 5. Insights into the multi-target actions of ferulic acid in acute lung injury/acute respiratory distress syndrome. Ferulic acid (FA) can directly target TLR4 and inhibit the TLR4/NF-κB pathway, decreasing the expression of phospho-NF-κB and downstream inflammatory mediators. Moreover, FA inhibits the expression of MAPK signaling pathway-related proteins. Additionally, ethyl ferulate improves acute lung injury/acute respiratory distress syndrome (ALI/ARDS) in an AMPK/Nrf2-dependent manner. AMPK: adenosine monophosphate-activated protein kinase; HO-1: heme oxygenase-1; MAPK: mitogen-activated protein kinase; Nrf2: nuclear factor erythroid-2-related factor-2; TLR4: toll-like receptor4.
Ijms 24 08011 g005
Table 1. Physico-chemical properties of FA.
Table 1. Physico-chemical properties of FA.
PropertyIndex
Molecular weight194.18
Melting point170.0 ± 2.0 °C
Boiling point372.3 ± 27.0 °C
Density1.316 ± 0.06 g/cm3
Cis isomerYellow oily substance
Trans isomerMonoclinic crystal
SolubilitySoluble in hot water, ethanol and ethyl acetate, slightly soluble in diethyl ether, and poorly soluble in benzene and petroleum ether
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhai, Y.; Wang, T.; Fu, Y.; Yu, T.; Ding, Y.; Nie, H. Ferulic Acid: A Review of Pharmacology, Toxicology, and Therapeutic Effects on Pulmonary Diseases. Int. J. Mol. Sci. 2023, 24, 8011. https://doi.org/10.3390/ijms24098011

AMA Style

Zhai Y, Wang T, Fu Y, Yu T, Ding Y, Nie H. Ferulic Acid: A Review of Pharmacology, Toxicology, and Therapeutic Effects on Pulmonary Diseases. International Journal of Molecular Sciences. 2023; 24(9):8011. https://doi.org/10.3390/ijms24098011

Chicago/Turabian Style

Zhai, Yiman, Tingyu Wang, Yunmei Fu, Tong Yu, Yan Ding, and Hongguang Nie. 2023. "Ferulic Acid: A Review of Pharmacology, Toxicology, and Therapeutic Effects on Pulmonary Diseases" International Journal of Molecular Sciences 24, no. 9: 8011. https://doi.org/10.3390/ijms24098011

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop