Next Article in Journal
Uropathogenic E. coli and Hybrid Pathotypes in Mexican Women with Urinary Tract Infections: A Comprehensive Molecular and Phenotypic Overview
Previous Article in Journal
The Root Extract of Rosa multiflora Ameliorates Nonalcoholic Steatohepatitis Development via Blockade of De Novo Lipogenesis and Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Licorice Chalcones as Molecular Genes and Signaling Pathways Modulator—A Review of Experimental Implications for Nicotine-Induced Non-Small Cell Lung Cancer Treatment

by
Naser A. Alsharairi
Heart, Mind and Body Research Group, Griffith University, Gold Coast, QLD 4222, Australia
Curr. Issues Mol. Biol. 2024, 46(6), 5894-5908; https://doi.org/10.3390/cimb46060352
Submission received: 27 May 2024 / Revised: 11 June 2024 / Accepted: 11 June 2024 / Published: 13 June 2024

Abstract

:
Lung cancer (LC) represents the leading cause of global cancer deaths, with cigarette smoking being considered a major risk factor. Nicotine is a major hazardous compound in cigarette smoke (CS), which stimulates LC progression and non-small cell lung cancer (NSCLC) specifically through activation of the nicotinic acetylcholine receptor (α7nAChR)-mediated cell-signaling pathways and molecular genes involved in proliferation, angiogenesis, and metastasis. Chalcones (CHs) and their derivatives are intermediate plant metabolites involved in flavonol biosynthesis. Isoliquiritigenin (ILTG), licochalcone A–E (LicoA–E), and echinatin (ECH) are the most common natural CHs isolated from the root of Glycyrrhiza (also known as licorice). In vitro and/or vivo experiments have shown that licorice CHs treatment exhibits a range of pharmacological effects, including antioxidant, anti-inflammatory, and anticancer effects. Despite advances in NSCLC treatment, the mechanisms of licorice CHs in nicotine-induced NSCLC treatment remain unknown. Therefore, the aim of this paper is to review experimental studies through the PubMed/Medline database that reveal the effects of licorice CHs and their potential mechanisms in nicotine-induced NSCLC treatment.

Graphical Abstract

1. Introduction

Globally, lung cancer (LC) is regarded as the most common malignant tumor causing death, with tobacco smoking being considered the greatest risk factor [1,2,3]. Non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) are the two major types associated with tobacco smoking, with NSCLC comprising 85% of all LC cases while SCLC represents the remaining 15% [3]. NSCLC is further categorized into large cell carcinoma, adenocarcinoma, and squamous subtypes [3].
Tobacco smoke consists of multiple chemicals identified as carcinogens, such as “4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone” (NNK) and “N-Nitrosonornicotine” (NNN), which contribute to NSCLC risk [4]. While nicotine is not a carcinogen, it could be the cause of the increasing NSCLC risk in smokers [5]. Both nitrosamines (NNK, NNN) and nicotine demonstrated significant induction of the nicotinic acetylcholine receptor (α7nAChRs) and beta-adrenergic receptor (β-AdrR) expressed in several brain areas (e.g., hypothalamus), which could, in turn, promote tumor growth, proliferation, angiogenesis, and metastasis in NSCLC cells [4,5,6]. The mechanisms through which nicotine causes NSCLC have been widely described in the literature [6,7]. Figure 1 shows the mechanisms of nicotine in NSCLC progression.
Chalcones (CHs), also termed 1,3-diphenyl-2-propen-1-ones, are plant metabolites that serve as precursors for isoflavonoids and flavonoids biosynthesis in edible and medicinal plants [8]. CHs also act as positive allosteric modulators (PAMs) of human α7nAChR channels polyhydroxy-substituted chalcone and diphenyl propanones [9]. CHs have a ketoethylenic moiety in their structure and consist of benzene rings (A and B) that are connected through a three-carbon α, β-unsaturated carbonyl system. Moreover, CHs possess a two-fold conjugate bond and exhibit different π-electron delocalization on both rings [8,10,11].
Natural CHs have been widely studied and often exhibit anticancer, antidiabetic, anti-inflammatory, antioxidant, and neuroprotective activities [12,13,14,15]. Natural CHs are categorized based on the type of subtraction into methoxy, amino, alkyl, aryl, and hydroxy CHs [16]. Isoliquiritigenin (ILTG), licochalcones A–E (LicoA–E), and echinatin (ECH) are natural hydroxy-methoxy CHs, mainly isolated from the Glycyrrhiza genus (also known as licorice), which belongs to the leguminosae/Fabaceae family, which consists of several species, where G. glabra, G. uralensis, and G. inflate are the most common ones [17]. These CHs exert anti-inflammatory, anti-angiogenesis, apoptosis, and cell cycle arrest activities in cancer cells through a wide range of molecular mechanisms such as cyclin-dependent kinase 1 (CDK1), proliferating cell nuclear antigen (PCNA), and nuclear transcription factor kappaB (NFκB) [17,18,19,20,21,22]. The chemical structures of natural CHs derived from Glycyrrhiza (ILTG, LicoA–E, and ECH) are shown in Figure 2.
Licorice CHs have been regarded as safe, but the main issue lies with the range of safe doses in treatment [18]. There is still a research gap on whether licorice ILTG for use in treating cancer is safe and nontoxic [25]. However, in vitro and in vivo experimental evidence has suggested that licorice LicoA and ILTG counteract the toxicity of chemotherapeutic drugs, along with enhancing anticancer activity [19]. A few in vivo experiments in rats showed that the bioavailability of ILTG is frequently low, which is attributed to the disappearance and/or rapid formation of ILTG metabolites in the liver and small intestine [26,27]. The administration of ILTG to rats at a dose of 20 mg/kg led to low absorption efficiencies and a rapid disappearance of ILTG in different tissues (liver, heart, lung, and kidney) [28]. An experiment has revealed a very small amount of the ILTG metabolite isoliquiritin apioside absorbed by passive diffusion in the intestinal Caco-2 cell monolayer [29]. It was reported that the absorbed LicoA and ECH in rat plasma were 3.3% and 6.81%, respectively, after oral (15 mg/kg) and intravenous (5 mg/kg) administeration of LicoA and ECH [30,31].
Research evidence suggests that licorice-derived flavonoids exert autophagic effects against NSCLC cells in vivo through downregulating the CDK4-cyclin D1 signaling pathway [32]. Natural flavonoids, including flavones, anthocyanins, proanthocyanidins, and quercetin derived from medicinal plants (e.g., Scutellaria baicalensis, Vaccinium macrocarpon, Myrica rubra, Rhododendron formosanum, Ginkgo biloba, Polygonum aviculare), are thought to be effective in nicotine-induced NSCLC treatment due to their anti-proliferative, anti-angiogenesis, anti-inflammatory, and apoptotic/autophagic properties [33,34,35]. However, the effects and mechanisms of licorice CHs in nicotine-induced NSCLC treatment remain largely unknown. Thus, this is the first review to cover experimental studies summarizing the therapeutic effects of licorice CHs in nicotine-induced NSCLC and exploring the mechanisms of action that underlie their effects.

2. Methods

A literature search was performed to identify articles in the PubMed/Medline database until 15 May 2024. The search was conducted using the “AND” Boolean function to combine the following keywords: “NSCLC”, “Glycyrrhiza”, “licorice”, “ILTG”, “Lico”, and “ECH”. Experimental studies published in English on the effects and/or mechanisms of licorice CHs in nicotine-induced NSCLC treatment were included. Studies were excluded if they focused on emphysema, as it has different cellular and/or molecular mechanisms underlying nicotine-induced lung injury compared to NSCLC [36]. The search identified 24 experimental studies that met the search criteria, which included 19 in vitro, 1 in vivo, and 4 mixed (in vitro and in vivo).

3. Licorice Isoliquiritigenin in Nicotine-Induced NSCLC Treatment

Among CHs, ILTG (4,2′,4′-trihydroxychalcone) is considered the most active one, which acts as a PAM for α7nAChR due to its ability to facilitate agonist-induced activation of α7 choline receptors without interacting with other subtypes of selective nAChR agonists [9]. The PAM activity of ILTG results from the substitution of the hydroxyl (OH) groups from position 2 to 4 of ring B, combined with the presence of a hydroxy substitution at the 2′ or 4′ position of ring A [9]. Treatment with ILTG showed inhibition of nicotine-induced inflammasome activation in the brain of female Sprague-Dawley rats [37]. It has been reported that amyloid-beta (Aβ)1–42 aggregation could be inhibited by ILTG in vitro, which may be due to the presence of a 4-substituted side chain in the A ring of ILTG, thereby producing π-π and hydrophobic bonds with Aβ1–42 [38]. ILTG was also found to attenuate Aβ42-induced oxidative stress in microglia by reducing the production of inflammatory cytokines and nitric oxide (NO) through the downregulation of NFκB and the upregulation of nuclear factor erythroid-2 related factor 2 (Nrf2) signaling pathways [39]. Activation of the α7nAChR-mediated signaling pathways and molecular genes involved in NSCLC cell proliferation and anti-apoptosis were found to aggregate Aβ40 and Aβ42 levels, which inhibit protein 53 (p53)-induced apoptosis, leading to the upregulation of the phosphatidylinositol-3 kinase/threonine kinase/NFκB (PI3K/Akt/NFκB) signaling pathway and metalloproteinase 2/9 (MMP2/9) expression [40]. Therefore, ILTG might have anti-cancer effects by regulating cellular processes in NSCLC cells through downregulation of the α7nAChR-mediated signaling pathways and gene expression involved in Aβ aggregation.
ILTG also has the potential to inhibit monoamine oxidase (MAO) in vitro by interacting with active site residues at dopamine and vasopressin receptors, thereby reducing neuronal disorders [41]. MAO promotes metastatic potential in NCI-A549 NSCLC cells in such a way that it increases migration and reactive oxygen species (ROS) production through the activation of multiple signaling pathways, including signal transducer and activator of transcription (STAT1/3/6), cAMP-responsive element-binding protein (CREP), early growth response 1 (EGR1), and peroxisome proliferator-activated receptor γ (PPARγ) [42]. MAO is indeed highly expressed in NSCLC cells, and MAO knockout obviously inhibits human papillomavirus (HPV)-16 E7 oncoprotein-induced tumor growth, hypoxia-inducible factor-1 (HIF-1α) protein accumulation, ROS production, and the expression of epithelial-mesenchymal transition (EMT)-associated markers in NCI-H460 NSCLC cells [43]. MAO inhibitors G10 and G11 have been reported to exhibit high inhibitory effects on paclitaxel-resistant NCI-A549/H460 NSCLC cell proliferation, growth, and metastasis [44,45], through the downregulation of the Akt/HIF-1α signaling pathway and MMP2/p21 expression [45]. Thus, ILTG may have significant inhibitory effects on MAO-induced migration, proliferation, growth, and metastasis of NSCLC cells through the downregulation of a7nAChR-mediated signaling pathways and molecular genes.
ILTG has shown in a few experiments its therapeutic potential in nicotine-induced NSCLC by inhibiting proliferation, invasion, migration, and tumor growth, along with the induction of apoptosis and autophagy via modulating the α7nAChR-mediated signaling pathways and/or molecular genes. Treatment with ILTG resulted in the inhibition of proliferation, invasion, and migration by inducing apoptosis in NSCLC cells in vitro at a concentration of 20 μM. This was demonstrated by activating the expression of pro-apoptosis-associated genes, and inhibiting the expression of anti-apoptosis, proliferation, invasion, and migration-associated genes, as well as the PI3K/Akt signaling pathway [46]. In vitro experiments showed anti-proliferative and apoptotic effects on NSCLC cells following treatment with 20 and 40 μmol/L ILTG by blocking the cell cycle at the G1 phase via upregulating expression of apoptosis and cell-cycle-associated genes [47]. ILTG at a concentration of 40 μM induces proliferation inhibition and G2/M cell cycle arrest in NSCLC cells through a mechanism that involves upregulating cell cycle-related genes [48]. The combinational treatment of NSCLC cells with ILTG and flavonoid glycosides (liquiritin, isoliquiritin) extracted from G. uralensis resulted in the induction of apoptosis, cytotoxicity, and cell cycle arrest at the G2/M phase in vitro [49]. ILTG was found to be effective in inhibiting tumor growth of NSCLC cells in vitro when treated at a concentration of 50 μg/mL by suppressing the localization of β-catenin to the nucleus [50]. It has been shown that ILTG inhibits the proliferation, invasion, and migration of NSCLC cells, with significant cytotoxicity observed at different concentrations in vitro. This inhibition was mediated by downregulating oncofetal IGF2 mRNA-binding protein 3 (IGF2BP3) expression, which promoted the mRNA stability of twist family bHLH transcription factor 1 (TWIST1) [51]. An in vitro experiment on NSCLC cells showed that apoptosis and cytotoxicity were significantly enhanced after treatment with ILTG nanosuspension in comparison to pure ILTG at different concentrations. However, the mechanisms behind these effects have not been elucidated [52].
In vitro and in vivo experiments reported inhibition of NSCLC cell migration, invasion, and metastasis when treated with ILTG at a concentration of 10 μM in a mechanism possibly involving blocked cytoskeleton reorganization and focal adhesion assembly through reducing the abundance of tyrosine (Tyr)-phosphorylated non-receptor tyrosine kinase (Src) and focal adhesion kinase (FAK) [53]. ILTG was also found to significantly induce apoptosis in tyrosine kinase inhibitor (TKI)-resistant and sensitive NSCLC cells both in vitro and in vivo at a concentration of 40 μM by targeting mutant/wild type epidermal growth factor receptor (EGFR) through downregulating Akt and extracellular signal-regulated kinase (ERK)1/2 signaling pathways, B-cell lymphoma-2 (Bcl-2) expression, and upregulating poly ADP ribose polymerase (PARP) cleavage, B-cell chronic lymphocytic leukemia-lymphoma like 11 gene (Bim), and caspase 3 expression [54].
Table 1 and Figure 3 highlight the therapeutic effects and mechanisms of licorice ILTG in nicotine-induced NSCLC.

4. Licorice Licochalcone in Nicotine-Induced NSCLC Treatment

Lico is a natural CH extracted from licorice and categorized into five major classes (licoA, licoB, licoC, licoD, and licoE), which have been shown to exhibit antidiabetic, antiallergic, antioxidant, anti-inflammatory, antimicrobial, antiviral, and anticancer activities [21]. All licos possess an OH group on the A and B rings. LicoA and LicoE have a 1,1-dimethyl-2-propenyl” substituent at position C-5. In LicoC and LicoD, there is a prenyl substituent (3-methylbut-2-en-1-yl) at the C-3 and C-3′ positions, respectively (Figure 2). However, the potential mechanisms of these licos in nicotine-induced NSCLC treatment remain unclear. Licos play a significant role in inhibiting the migration, invasion, and proliferation of cancer cells, along with promoting apoptosis and cell cycle arrest through suppressing the activity of α7nAChR-mediated signaling pathways, including PI3K/Akt/mTOR, ERK, and Wnt/β-catenin [55]. LicoE treatment resulted in inhibited cell migration, invasion, metastasis, angiogenesis, and induced apoptosis in mouse lung tissues via upregulating the expression of HIF-1α, CDK, MMP-9, cyclooxygenase-2 (COX-2), vascular endothelial growth factor (VEGF), lymphocyte common antigen 45 (CD45), lymphatic vessel endothelial receptor-1 (LVER-1), and downregulating the expression of Bcl-2-associated X protein (Bax) and cleaved caspase-3 [56]. LicoA, LicoB, and LicoD have been shown to exert anti-inflammatory effects on murine macrophage cell line RAW264.7 by inhibiting α7nAChR-mediated signaling cascades, including lipopolysaccharide (LPS)-induced protein kinase A (PKA), NO production, NF-κB p65 phosphorylation at serine 276, tumor necrosis factor (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) expression [57,58]. Treatment with licoB showed a reduction of oxidative damage in RAW264.7 cells in vivo. LicoB decreases the production of ROS and inflammatory cytokines (e.g., TNFα) and increases the levels of antioxidant substances through the activation of Nrf2 and the downregulation of NF-κB signaling pathways [59]. Therefore, licos may exert anticancer effects in nicotine-induced NSCLC via modulation of the α7nAChR-mediated signaling pathways and/or molecular genes.
Activation of the α7nAChR-mediated signaling pathways, known to be involved in a range of cellular processes, including cell proliferation and anti-apoptosis, was found to increase Aβ levels in NSCLC cells [40]. LicoA and LicoB demonstrated anti-Aβ aggregation activity by collapsing the hydrogen bonds inside the Aβ1–42 protofibril to varying degrees [60]. LicoE inhibits Aβ1–42 aggregation through suppression of the choline transporter-like protein 1 function in microglia and TNF-α mRNA expression [61]. Thus, licos may inhibit excessive accumulation of Aβ in NSCLC cells, leading to the suppression of α7nAChR and its downstream signaling pathways.
Several in vitro experiments investigating the therapeutic potential of licoA in nicotine-induced NSCLC suggest it exerts several activities, including anti-tumor growth, anti-proliferative, anti-migration, anti-invasion, anti-viability, apoptosis, and autophagy, via modulation of the α7nAChR-mediated signaling pathways and/or molecular genes. Treatment with licoA at a 13 μg/mL concentration showed significant inhibitory effects on the nuclear localization of β-catenin in NSCLC cells by disrupting the activity of Wnt/β-catenin signaling [50]. LicoA treatment demonstrated anti-proliferative and apoptotic effects in NSCLC cells at a concentration of 10 μM through upregualting microRNA (miRNAs) expression, leading to promote endoplasmic reticulum (ER) stress by inhibiting Nrf2 expression. However, treatment with licoA at higher concentrations (in particular 40 μM) enhances autophagy and unfolded protein response (UPR), but without affecting the downstream apoptotic genes of Bim and Bcl-2 in NSCLC cells [62]. LicoA promotes the migration and invasion-inhibiting effects in NSCLC when treated at a concentration of 2–20 μM through the suppression of Sp1 (specificity protein 1) expression and MMP signaling pathways [63]. LicoA suppresses proliferation and promotes apoptosis, autophagy, and cell cycle arrest at the S and G2/M phases in NSCLC cells at 40 and 80 μM concentrations through the modulation of apoptotic and anti-apoptotic gene expression [64]. LicoA induces apoptotic and autophagic cytotoxicity in NCI-A549 and NCI-H460 cells at different concentrations by altering the expression of apoptosis and autophagy-related genes through the downregulation of ERK and c-Jun N-terminal kinase (JNK) signaling pathways [65]. LicoA has been reported to significantly repress NSCLC cell viability by inhibiting hypoxia-mediated HIF-1α activation and its related genes at a concentration of 20 μM [66]. It has been demonstrated that licoA inhibits proliferation and promotes apoptosis and cell cycle arrest at the G2/M phase at different concentrations by altering cell cycle and apoptosis-related gene expression via the upregulation of the ER stress pathway [67]. LicoA inhibits the growth of gefitinib-resistant NSCLC cells at 50–70 μM concentrations via suppressing heat shock protein 90 (Hsp90) expression, resulting in a significant depletion of oncogenic genes [68]. Treatment of NSCLC cells with licoA at 5–20 μM concentrations led to a significant inhibition of viability and an inducer of apoptosis, autophagy, and cell cycle arrest at the sub-G1 phase. The mechanisms behind these effects are associated with increased lactate dehydrogenase (LDH) release and upregulated apoptotic and autophagic-related gene expression [69]. NSCLC cells treated with licoA at a concentration of 10 μM resulted in inhibited proliferation and induced apoptosis by downregulating interferon-gamma (IFN-γ)-induced programmed death-ligand 1 (PD-L1) protein expression through the generation of ROS, the activation of protein kinase RNA-like endoplasmic reticulum kinase-eukaryotic initiation factor 2α (PERK-eIF2α) axis, and the inhibition of eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) phosphorylation [70].
Treatment with licoA represses cell viability and induces apoptosis in vivo by upregulating apoptosis-related genes and inhibiting tyrosine-protein kinase Met (c-Met) phosphorylation and its downstream kinases-mediated gefitinib resistance in NSCLC cells through promoting c-Casitas B-lineage lymphoma (c-Cbl), which is responsible for licoA-induced c-Met degradation [71]. LicoA treatment at 20 and 40 μM concentrations resulted in a significant inhibition of proliferation in vitro and xenograft tumor growth in vivo, along with the induction of apoptosis and cell cycle arrest at the G1 phase. The mechanisms behind these effects in NSCLC cells are related to the upregulation and/or downregulation of molecular genes and signaling pathways involved in cell proliferation and apoptosis [72]. Treatment with licoA inhibits in vitro and in vivo cell growth of osimertinib-sensitive and osimertinib-resistant NSCLC cells at different concentrations, with no significant cytotoxicity against these cells observed. LicoA treatment also induces apoptosis by suppressing survivin protein via the downregulation of EGFR and its downstream kinases [73].
LicoB treatment has shown anticancer effects on gefitinib-sensitive and gefitinib-resistant NSCLC cells in vitro at 5–20 μM concentrations by inducing proliferation inhibition, apoptosis, and cell cycle arrest at the G2/M phase. LicoB treatment significantly inhibited EGFR, mesenchymal epithelial transition factor receptor (MET) activity, apoptotic gene expression, and enhanced ROS, ER stress, and pro-apoptotic gene expression [74]. Treatment with licoD at the same concentrations also resulted in suppressed phosphorylation and the kinase activity of EGFR and MET, induced apoptosis, and blocked cell cycle progression at the G2/M phase in gefitinib-sensitive and gefitinib-resistant NSCLC cells in vitro [75].
Table 2 summarizes the effects of licorice licos in nicotine-induced NSCLC treatment. Taken together, licoA, licoB, and licoD may be effective in nicotine-induced NSCLC treatment via modulating the α7nAChR-mediated molecular genes and/or cellular signaling pathways (Figure 4).

5. Licorice Echinatin in Nicotine-Induced NSCLC Treatment

ECH, a CH isolated from licorice, is structurally similar to licoA, having one OH group on the A and B rings (positions 4′ and 4) (Figure 2). ECH exerts strong antioxidant effects by generating two radical adduct formation products (ECH-ECH dimer and ECH-2-1,1-diphenyl-2-picrylhydrazyl radical adducts) through hydrogen atom transfer antioxidant mechanisms [76]. However, whether ECH has a therapeutic role in nicotine-induced NSCLC remains unknown. Only one in vitro experiment has examined the therapeutic effect of ECH against NSCLC cells. The experiment demonstrated anti-proliferative, apoptotic, and autophagic effects in gefitinib-sensitive and resistant NSCLC cells (NCI-HCC827, NCI-HCC827GR) when treated at a concentration of 5–15 μM by inhibiting the expression of EGFR, MET, Akt, ERK, and increasing ROS production, all known as α7nAChR-mediated molecular genes involved in NSCLC development [77].

6. Limitations

Most experiments investigating the anticancer effects of licorice CHs in nicotine-induced NSCLC have been performed in vitro, with only a few in vivo mouse models being used. Licorice CHs have never been assessed for their clinical significance, and there are no tools for monitoring the effectiveness and safety of these CHs in NSCLC patients. The extraction and identification of CHs have not been clearly determined. A few in vitro experiments showed high cytotoxicity of ILTG and licoA treatment, which could be useful in controlling the growth of NSCLC cells by enhancing apoptosis and autophagy in these cells. All experiments showed dose-dependent anticancer effects, but the optimal dose in treatment remains a key challenge. Licorice CHs have not been assessed for their bioavailability in all experiments.

7. Conclusions

This review highlights the effects of licorice CHs in nicotine-induced NSCLC treatment via various mechanisms of action. ILTG, licoA–E, and ECH are natural CHs isolated from licorice with anticancer effects in vitro and/or in vivo. However, the mechanisms of licorice CHs in nicotine-induced NSCLC treatment remain largely unknown. ILTG may produce anticancer effects in nicotine-induced NSCLC by PAM of α7nAChR. ILTG may also act as a potential inhibitory compound for MAO-induced cellular processes in NSCLC cells through the suppression of α7nAChR-mediated signaling pathways. ILTG and licos may have a significant role in repressing Aβ aggregation in NSCLC cells, leading to inhibited α7nAChR and its downstream signaling pathways implicated in NSCLC development.
In vitro and/or in vivo experiments have shown that licorice CHs exert multiple therapeutic effects in nicotine-induced NSCLC, including anti-proliferative, anti-tumor growth, anti-invasion, anti-migration, anti-viability, apoptosis, cell cycle arrest, and autophagy through modulating α7nAChR-mediated signaling pathways and molecular genes. Treatment with ILTG, licoA, licoB, and licoD has demonstrated anticancer effects on TKI, osimertinib, and gefitinib-sensitive and resistant NSCLC cells by inducing apoptosis and/or cell cycle arrest via upregulating apoptosis-related genes.
Although all experiments demonstrated that they were effective in NSCLC treatment, the exact mechanisms involved in the therapeutic effects of licorice CHs against NSCLC cells induced by nicotine as a stimulator of α7nAChR need further investigation. More in vitro and in vivo experiments are warranted to evaluate the toxicity, safety, and optimal effective dose of licorice CHs in nicotine-induced NSCLC treatment. Further clinical studies are needed to investigate the therapeutic role of licorice CHs in nicotine-induced NSCLC, particularly in smokers.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

4EBP1Eukaryotic translation initiation factor 4E-binding protein 1
AktThreonine kinase
Amyloid-beta
BaxBcl-2-associated X protein
Bcl-2B-cell lymphoma-2
Bcl-xlB-cell lymphoma-extra large
BimB-cell chronic lymphocytic leukemia-lymphoma like 11 gene
c-Cblc-Casitas B-lineage lymphoma
CD45Lymphocyte common antigen 45
CDC2Cell division control protein 2 homologue
CDC25Cell division control protein 25 homologue
CDC42Cell division control protein 42 homologue
CDKCyclin-dependent kinase
c-FLIPLCellular-FLICE inhibitory protein
CHOPC/EBP homologous protein
CHsChalcones
c-MetTyrosine-protein kinase Met
COX-2Cyclooxygenase-2
CREPcAMP-responsive element-binding protein
CSCigarette smoke
DMEMDulbecco’s Modified Eagle Medium
ECHEchinatin
EGFREpidermal growth factor receptor
EGR1Early growth response 1
eIF2αEukaryotic initiation factor 2α
EMTEpithelial-mesenchymal transition
EREndoplasmic reticulum
ERKExtracellular signal-regulated kinase
FAKFocal adhesion kinase
FBXO5F-box protein 5
GADD45Growth Arrest and DNA Damage-inducible 45
GLUT1Glucose transporter 1
GRP7878-kDa glucose-regulated protein
GSKGlycogen synthase kinase
GTPaseGuanosine triphosphatase
Her2Human epidermal growth factor receptor 2
HGFHepatocyte growth factor
HIF-1αHypoxia inducible factor-1
HPVHuman papillomavirus
Hsp90Heat shock protein 90
IAPsInhibitor of apoptosis proteins
IFN-γInterferon-gamma
IGF2BP3Oncofetal IGF2 mRNA-binding protein 3
ILTGIsoliquiritigenin
JNKc-Jun N-terminal kinase
LCLung cancer
LC3LC3-phospholipid conjugate
LDHLactate dehydrogenase
LEVR-1Lymphatic vessel endothelial receptor-1
LicosLicochalcones
LPSLipopolysaccharide
MAOMonoamine oxidase
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemoattractant protein-1
MDM2Murine double minute 2
MetMesenchymal epithelial transition factor receptor
mFasLMembrane-bound Fas ligand
miRNAsMicroRNAs
MMPMetalloproteinase
mTORMammalian target of rapamycin
NF-kBNuclear transcription factor-kappaB
NNK4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone
NNNN-Nitrosonornicotine
NONitric oxide
Nrf2Nuclear factor erythroid-2 related factor 2
NSCLCNon-small cell lung cancer
OHHydroxyl group
p21Protein 21
p53Protein 53
p62Protein 62
p65Protein 65
p70Protein 70
PAMsPositive allosteric modulators
PARPPoly ADP ribose polymerase
PCNAProliferating cell nuclear antigen
PDK1Pyruvate dehydrogenase kinase 1
PD-L1Programmed death-ligand 1
PERKProtein kinase RNA-like endoplasmic reticulum kinase
PI3KPhosphatidylinositol-3 kinase
PKAProtein kinase A
PPARγPeroxisome proliferator-activated receptor γ
PTENPhosphatase and tensin homolog
RafRetinoblastoma tumor suppressor protein-proto-oncogene
RIP1Receptor-interacting protein-1
ROSReactive oxygen species
SCLCSmall cell lung cancer
sFasLSoluble Fas ligand
Sp1Specificity protein 1
SrcNon-receptor tyrosine kinase
STATSignal transducer and activator of transcription
TKITyrosine kinase inhibitor
TNFαTumor necrosis factor
TWIST1Twist family bHLH transcription factor 1
TyrTyrosine
UPRUnfolded protein response
VEGFVascular endothelial growth factor
WAF1/CIP1Cyclin-dependent kinase inhibitor p21
α7nAChRNicotinic acetylcholine receptor
β-AdrRbeta-adrenergic receptor

References

  1. Ji, X.; Chen, J.; Ye, J.; Xu, S.; Lin, B.; Hou, K. Epidemiological analysis of global and regional lung cancer mortality: Based on 30-year data analysis of global burden disease database. Healthcare 2023, 11, 2920. [Google Scholar] [CrossRef] [PubMed]
  2. Thandra, K.C.; Barsouk, A.; Saginala, K.; Aluru, J.S.; Barsouk, A. Epidemiology of lung cancer. Contemp. Oncol. 2021, 25, 45–52. [Google Scholar]
  3. Schabath, M.B.; Cote, M.L. Cancer progress and priorities: Lung cancer. Cancer Epidemiol. Biomark. Prev. 2019, 28, 1563–1579. [Google Scholar] [CrossRef] [PubMed]
  4. Xue, J.; Yang, S.; Seng, S. Mechanisms of cancer induction by tobacco-specific NNK and NNN. Cancers 2014, 6, 1138–1156. [Google Scholar] [CrossRef]
  5. Warren, G.W.; Singh, A.K. Nicotine and lung cancer. J. Carcinog. 2013, 12, 1. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, S.; Hu, Y. α7 nicotinic acetylcholine receptors in lung cancer. Oncol. Lett. 2018, 16, 1375–1382. [Google Scholar] [CrossRef] [PubMed]
  7. Hajiasgharzadeh, K.; Sadigh-Eteghad, S.; Mansoori, B.; Mokhtarzadeh, A.; Shanehbandi, D.; Doustvandi, M.A.; Asadzadeh, Z.; Baradaran, B. Alpha7 nicotinic acetylcholine receptors in lung inflammation and carcinogenesis: Friends or foes? J. Cell. Physiol. 2019, 234, 14666–14679. [Google Scholar] [CrossRef]
  8. Rudrapal, M.; Khan, J.; Bin Dukhyil, A.Z.; Alarousy, R.M.I.I.; Attah, E.I.; Sharma, T.; Khairnar, S.J.; Bendale, A.R. Chalcone scaffolds, bioprecursors of flavonoids: Chemistry, bioactivities, and pharmacokinetics. Molecules 2021, 26, 7177. [Google Scholar] [CrossRef] [PubMed]
  9. Balsera, B.; Mulet, J.; Fernández-Carvajal, A.; de la Torre-Martínez, R.; Ferrer-Montiel, A.; Hernández-Jiménez, J.G.; Estévez-Herrera, J.; Borges, R.; Freitas, A.E.; López, M.G.; et al. Chalcones as positive allosteric modulators of α7 nicotinic acetylcholine receptors: A new target for a privileged structure. Eur. J. Med. Chem. 2014, 86, 724–739. [Google Scholar] [CrossRef] [PubMed]
  10. Jasim, H.A.; Nahar, L.; Jasim, M.A.; Moore, S.A.; Ritchie, K.J.; Sarker, S.D. Chalcones: Synthetic chemistry follows where nature leads. Biomolecules 2021, 11, 1203. [Google Scholar] [CrossRef]
  11. Elkanzi, N.A.A.; Hrichi, H.; Alolayan, R.A.; Derafa, W.; Zahou, F.M.; Bakr, R.B. Synthesis of chalcones derivatives and their biological activities: A Review. ACS Omega 2022, 7, 27769–27786. [Google Scholar] [CrossRef] [PubMed]
  12. Irfan, R.; Mousavi, S.; Alazmi, M.; Saleem, R.S.Z. A Comprehensive review of aminochalcones. Molecules 2020, 25, 5381. [Google Scholar] [CrossRef] [PubMed]
  13. Constantinescu, T.; Lungu, C.N. Anticancer activity of natural and synthetic chalcones. Int. J. Mol. Sci. 2021, 22, 11306. [Google Scholar] [CrossRef] [PubMed]
  14. Salehi, B.; Quispe, C.; Chamkhi, I.; El Omari, N.; Balahbib, A.; Sharifi-Rad, J.; Bouyahya, A.; Akram, M.; Iqbal, M.; Docea, A.O.; et al. Pharmacological properties of chalcones: A review of preclinical including molecular mechanisms and clinical evidence. Front. Pharmacol. 2021, 11, 592654. [Google Scholar] [CrossRef] [PubMed]
  15. Birsa, M.L.; Sarbu, L.G. Hydroxy chalcones and analogs with chemopreventive properties. Int. J. Mol. Sci. 2023, 24, 10667. [Google Scholar] [CrossRef] [PubMed]
  16. Maisto, M.; Marzocchi, A.; Keivani, N.; Piccolo, V.; Summa, V.; Tenore, G.C. Natural chalcones for the management of obesity disease. Int. J. Mol. Sci. 2023, 24, 15929. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, D.; Liang, J.; Zhang, J.; Wang, Y.; Chai, X. Natural chalcones in Chinese materia medica: Licorice. Evid. Based Complement. Altern. Med. 2020, 2020, 3821248. [Google Scholar] [CrossRef] [PubMed]
  18. Wahab, S.; Annadurai, S.; Abullais, S.S.; Das, G.; Ahmad, W.; Ahmad, M.F.; Kandasamy, G.; Vasudevan, R.; Ali, M.S.; Amir, M. Glycyrrhiza glabra (Licorice): A comprehensive review on its phytochemistry, biological activities, clinical evidence and toxicology. Plants 2021, 10, 2751. [Google Scholar] [CrossRef] [PubMed]
  19. Tuli, H.S.; Garg, V.K.; Mehta, J.K.; Kaur, G.; Mohapatra, R.K.; Dhama, K.; Sak, K.; Kumar, A.; Varol, M.; Aggarwal, D.; et al. Licorice (Glycyrrhiza glabra L.)-derived phytochemicals target multiple signaling pathways to confer oncopreventive and oncotherapeutic effects. Onco Targets Ther. 2022, 15, 1419–1448. [Google Scholar] [CrossRef]
  20. Wu, Y.; Wang, Z.; Du, Q.; Zhu, Z.; Chen, T.; Xue, Y.; Wang, Y.; Zeng, Q.; Shen, C.; Jiang, C.; et al. Pharmacological effects and underlying mechanisms of licorice-derived flavonoids. Evid. Based Complement. Altern. Med. 2022, 2022, 9523071. [Google Scholar] [CrossRef]
  21. Pia, G.D.M.; Sara, F.; Mario, F.; Lorenza, S. Biological effects of licochalcones. Mini Rev. Med. Chem. 2019, 19, 647–656. [Google Scholar]
  22. Chen, Z.; Ding, W.; Yang, X.; Lu, T.; Liu, Y. Isoliquiritigenin, a potential therapeutic agent for treatment of inflammation-associated diseases. J. Ethnopharmacol. 2024, 318 Pt B, 117059. [Google Scholar] [CrossRef]
  23. Cerulli, A.; Masullo, M.; Montoro, P.; Piacente, S. Licorice (Glycyrrhiza glabra, G. uralensis, and G. inflata) and their constituents as active cosmeceutical ingredients. Cosmetics 2022, 9, 7. [Google Scholar] [CrossRef]
  24. Fu, Y.; Chen, J.; Li, Y.-J.; Zheng, Y.-F.; Li, P. Antioxidant and anti-inflammatory activities of six flavonoids separated from licorice. Food Chem. 2013, 141, 1063–1071. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, K.-L.; Yu, Y.-C.; Hsia, S.-M. Perspectives on the role of isoliquiritigenin in cancer. Cancers 2021, 13, 115. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, Y.K.; Chin, Y.-W.; Bae, J.-K.; Seo, J.S.; Choi, Y.H. Pharmacokinetics of isoliquiritigenin and its metabolites in rats: Low bioavailability is primarily due to the hepatic and intestinal metabolism. Planta Med. 2013, 79, 1656–1665. [Google Scholar] [CrossRef]
  27. Choi, Y.H.; Kim, Y.-J.; Chae, H.-S.; Chin, Y.-W. In vivo gastroprotective effect along with pharmacokinetics, tissue distribution and metabolism of isoliquiritigenin in mice. Planta Med. 2015, 81, 586–593. [Google Scholar] [CrossRef]
  28. Qiao, H.; Zhang, X.; Wang, T.; Liang, L.; Chang, W.; Xia, H. Pharmacokinetics, biodistribution and bioavailability of isoliquiritigenin after intravenous and oral administration. Pharm. Biol. 2014, 52, 228–236. [Google Scholar] [CrossRef]
  29. Wang, X.-X.; Liu, G.-Y.; Yang, Y.-F.; Wu, X.-W.; Xu, W.; Yang, X.-W. Intestinal absorption of triterpenoids and flavonoids from Glycyrrhizae radix et rhizoma in the human Caco-2 monolayer cell model. Molecules 2017, 22, 1627. [Google Scholar] [CrossRef]
  30. Weng, Q.; Chen, L.; Ye, L.; Lu, X.; Yu, Z.; Wen, C.; Chen, Y.; Huang, G. Determination of licochalcone A in rat plasma by UPLC–MS/MS and its pharmacokinetics. Acta Chromatogr. 2019, 31, 262–265. [Google Scholar] [CrossRef]
  31. Li, T.; Ye, W.; Huang, B.; Lu, X.; Chen, X.; Lin, Y.; Wen, C.; Wang, X. Determination and pharmacokinetic study of echinatin by UPLC-MS/MS in rat plasma. J. Pharm. Biomed. Anal. 2019, 168, 133–137. [Google Scholar] [CrossRef] [PubMed]
  32. Zhu, J.; Huang, R.; Yang, R.; Xiao, Y.; Yan, J.; Zheng, C.; Xiao, W.; Huang, C.; Wang, Y. Licorice extract inhibits growth of non-small cell lung cancer by down-regulating CDK4-cyclin D1 complex and increasing CD8+ T cell infiltration. Cancer Cell Int. 2021, 21, 529. [Google Scholar] [CrossRef] [PubMed]
  33. Alsharairi, N.A. Scutellaria baicalensis and their natural flavone compounds as potential medicinal drugs for the treatment of nicotine-induced non-small-cell lung cancer and asthma. Int. J. Environ. Res. Public Health 2021, 18, 5243. [Google Scholar] [CrossRef] [PubMed]
  34. Alsharairi, N.A. Insights into the mechanisms of action of proanthocyanidins and anthocyanins in the treatment of nicotine-induced non-small cell lung cancer. Int. J. Mol. Sci. 2022, 23, 7905. [Google Scholar] [CrossRef]
  35. Alsharairi, N.A. Quercetin derivatives as potential therapeutic agents: An updated perspective on the treatment of nicotine-induced non-small cell lung cancer. Int. J. Mol. Sci. 2023, 24, 15208. [Google Scholar] [CrossRef] [PubMed]
  36. Blaskovic, S.; Donati, Y.; Ruchonnet-Metrailler, S.; Avila, Y.; Schittny, D.; Schlepütz, C.M.; Schittny, J.C.; Barazzone-Argiroffo, C. Early life exposure to nicotine modifies lung gene response after elastase-induced emphysema. Respir. Res. 2022, 23, 44. [Google Scholar] [CrossRef] [PubMed]
  37. d’Adesky, N.D.; de Rivero Vaccari, J.P.; Bhattacharya, P.; Schatz, M.; Perez-Pinzon, M.A.; Bramlett, H.M.; Raval, A.P. Nicotine alters estrogen receptor-beta-regulated inflammasome activity and exacerbates ischemic brain damage in female rats. Int. J. Mol. Sci. 2018, 19, 1330. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, Y.; Zhang, Z.-Y.; Li, Y.-P.; Li, D.; Huang, S.-L.; Gu, L.-Q.; Xu, J.; Huang, Z.-S. Syntheses and evaluation of novel isoliquiritigenin derivatives as potential dual inhibitors for amyloid-beta aggregation and 5-lipoxygenase. Eur. J. Med. Chem. 2013, 66, 22–31. [Google Scholar] [CrossRef] [PubMed]
  39. Fu, Y.; Jia, J. Isoliquiritigenin confers neuroprotection and alleviates amyloid-β42-induced neuroinflammation in microglia by regulating the Nrf2/NF-κB signaling. Front. Neurosci. 2021, 15, 638772. [Google Scholar] [CrossRef]
  40. Dorandish, S.; Williams, A.; Atali, S.; Sendo, S.; Price, D.; Thompson, C.; Guthrie, J.; Heyl, D.; Evans, H.G. Regulation of amyloid-β levels by matrix metalloproteinase-2/9 (MMP2/9) in the media of lung cancer cells. Sci. Rep. 2021, 11, 9708. [Google Scholar] [CrossRef]
  41. Prajapati, R.; Seong, S.H.; Park, S.E.; Paudel, P.; Jung, H.A.; Choi, J.S. Isoliquiritigenin, a potent human monoamine oxidase inhibitor, modulates dopamine D1, D3, and vasopressin V1A receptors. Sci. Rep. 2021, 11, 23528. [Google Scholar] [CrossRef]
  42. Dhabal, S.; Das, P.; Biswas, P.; Kumari, P.; Yakubenko, V.P.; Kundu, S.; Cathcart, M.K.; Kundu, M.; Biswas, K.; Bhattacharjee, A. Regulation of monoamine oxidase A (MAO-A) expression, activity, and function in IL-13-stimulated monocytes and A549 lung carcinoma cells. J. Biol. Chem. 2018, 293, 14040–14064. [Google Scholar] [CrossRef]
  43. Huang, B.; Zhou, Z.; Liu, J.; Wu, X.; Li, X.; He, Q.; Zhang, P.; Tang, X. The role of monoamine oxidase A in HPV-16 E7-induced epithelial-mesenchymal transition and HIF-1α protein accumulation in non-small cell lung cancer cells. Int. J. Biol. Sci. 2020, 16, 2692–2703. [Google Scholar] [CrossRef]
  44. Yang, X.; Zhao, D.; Li, Y.; Li, Y.; Cui, W.; Li, Y.; Li, H.; Li, X.; Wang, D. Potential monoamine oxidase A inhibitor suppressing paclitaxel-resistant non-small cell lung cancer metastasis and growth. Thorac. Cancer 2020, 11, 2858–2866. [Google Scholar] [CrossRef]
  45. Yang, X.-G.; Li, Y.-Y.; Zhao, D.-X.; Cui, W.; Li, H.; Li, X.-Y.; Li, Y.-X.; Wang, D. Repurposing of a monoamine oxidase A inhibitor-heptamethine carbocyanine dye conjugate for paclitaxel-resistant non-small cell lung cancer. Oncol. Rep. 2021, 45, 306–1314. [Google Scholar] [CrossRef]
  46. Tian, T.; Sun, J.; Wang, J.; Liu, Y.; Liu, H. Isoliquiritigenin inhibits cell proliferation and migration through the PI3K/AKT signaling pathway in A549 lung cancer cells. Oncol. Lett. 2018, 16, 6133–6139. [Google Scholar] [CrossRef]
  47. Hsu, Y.; Kuo, P.; Chiang, L.; Lin, C. Isoliquiritigenin inhibits the proliferation and induces the apoptosis of human non-small cell lung cancer A549 cells. Clin. Exp. Pharmacol. Physiol. 2004, 31, 414–418. [Google Scholar] [CrossRef]
  48. Li, T.; Satomi, Y.; Katoh, D.; Shimada, J.; Baba, M.; Okuyama, T.; Nishino, H.; Kitamura, N. Induction of cell cycle arrest and p21(CIP1/WAF1) expression in human lung cancer cells by isoliquiritigenin. Cancer Lett. 2004, 207, 27–35. [Google Scholar]
  49. Zhou, Y.; Ho, W.S. Combination of liquiritin, isoliquiritin and isoliquirigenin induce apoptotic cell death through upregulating p53 and p21 in the A549 non-small cell lung cancer cells. Oncol. Rep. 2014, 31, 298–304. [Google Scholar] [CrossRef]
  50. Heng, W.S.; Cheah, S. Identification of phytochemical-based β-catenin nuclear localization inhibitor in NSCLC: Differential targeting population from member of isothiocyanates. Molecules 2021, 26, 399. [Google Scholar] [CrossRef]
  51. Cui, Y.; Wu, Y.; Wang, C.; Wang, Z.; Li, Y.; Jiang, Z.; Zhao, W.; Pan, Z. Isoliquiritigenin inhibits non-small cell lung cancer progression via m6A/IGF2BP3-dependent TWIST1 mRNA stabilization. Phytomedicine 2022, 104, 154299. [Google Scholar] [CrossRef]
  52. Qiao, F.; Zhao, Y.; Mai, Y.; Guo, J.; Dong, L.; Zhang, W.; Yang, J. Isoliquiritigenin Nanosuspension enhances cytostatic effects in A549 lung cancer cells. Planta Med. 2020, 86, 538–547. [Google Scholar] [CrossRef]
  53. Chen, C.; Shenoy, A.K.; Padia, R.; Fang, D.; Jing, Q.; Yang, P.; Su, S.; Huang, S. Suppression of lung cancer progression by isoliquiritigenin through its metabolite 2, 4, 2’, 4’-Tetrahydroxychalcone. J. Exp. Clin. Cancer Res. 2018, 37, 243. [Google Scholar] [CrossRef]
  54. Jung, S.K.; Lee, M.; Lim, D.Y.; Kim, J.E.; Singh, P.; Lee, S.; Jeong, C.; Lim, T.; Chen, H.; Chi, Y.; et al. Isoliquiritigenin induces apopto-sis and inhibits xenograft tumor growth of human lung cancer cells by targeting both wild type and L858R/T790M mutant EGFR. J. Biol. Chem. 2014, 289, 35839–35848. [Google Scholar] [CrossRef]
  55. Deng, N.; Qiao, M.; Li, Y.; Liang, F.; Li, J.; Liu, Y. Anticancer effects of licochalcones: A review of the mechanisms. Front. Pharmacol. 2023, 14, 1074506. [Google Scholar] [CrossRef]
  56. Kwon, S.J.; Park, S.Y.; Kwon, G.T.; Lee, K.W.; Kang, W.-H.; Choi, M.-S.; Yun, J.W.; Jeon, J.-H.; Jun, J.G.; Park, J.H.Y. Licochalcone E present in licorice suppresses lung metastasis in the 4T1 mammary orthotopic cancer model. Cancer Prev. Res. 2013, 6, 603–613. [Google Scholar] [CrossRef]
  57. Furusawa, J.-I.; Funakoshi-Tago, M.; Tago, K.; Mashino, T.; Inoue, H.; Sonoda, Y.; Kasahara, T. Licochalcone A significantly suppresses LPS signaling pathway through the inhibition of NF-kappaB p65 phosphorylation at serine 276. Cell Signal 2009, 21, 778–785. [Google Scholar] [CrossRef]
  58. Furusawa, J.-I.; Funakoshi-Tago, M.; Mashino, T.; Tago, K.; Inoue, H.; Sonoda, Y.; Kasahara, T. Glycyrrhiza inflata-derived chalcones, Licochalcone A, Licochalcone B and Licochalcone D, inhibit phosphorylation of NF-kappaB p65 in LPS signaling pathway. Int. Immunopharmacol. 2009, 9, 499–507. [Google Scholar] [CrossRef]
  59. Dong, M.; Yang, Z.; Gao, Q.; Deng, Q.; Li, L.; Chen, H. Protective effects of isoliquiritigenin and licochalcone B on the immunotoxicity of BDE-47: Antioxidant effects based on the activation of the Nrf2 pathway and inhibition of the NF-κB pathway. Antioxidants 2024, 13, 445. [Google Scholar] [CrossRef]
  60. Fang, M.; Su, K.; Wang, X.; Guan, P.; Hu, X. Study on molecular mechanisms of destabilizing Aβ(1–42) protofibrils by licochalcone A and licochalcone B using molecular dynamics simulations. J. Mol. Graph. Model. 2023, 122, 108500. [Google Scholar] [CrossRef]
  61. Muto, E.; Okada, T.; Yamanaka, T.; Uchino, H.; Inazu, M. Licochalcone E, a β-Amyloid aggregation inhibitor, regulates microglial M1/M2 polarization via inhibition of CTL1-mediated choline uptake. Biomolecules 2023, 13, 191. [Google Scholar] [CrossRef]
  62. Chen, G.; Ma, Y.; Jiang, Z.; Feng, Y.; Han, Y.; Tang, Y.; Zhang, J.; Ni, H.; Li, X.; Li, N. Lico A causes ER stress and apoptosis via up-regulating miR-144-3p in human lung cancer cell line H292. Front. Pharmacol. 2018, 9, 837. [Google Scholar] [CrossRef]
  63. Huang, H.-C.; Tsai, L.-L.; Tsai, J.-P.; Hsieh, S.-C.; Yang, S.-F.; Hsueh, J.-T.; Hsieh, Y.-H. Licochalcone A inhibits the migration and invasion of human lung cancer cells via inactivation of the Akt signaling pathway with downregulation of MMP-1/-3 expression. Tumour Biol. 2014, 35, 12139–12149. [Google Scholar] [CrossRef]
  64. Lin, Y.-J.; Liang, W.-M.; Chen, C.-J.; Tsang, H.; Chiou, J.-S.; Liu, X.; Cheng, C.-F.; Lin, T.-H.; Liao, C.-C.; Huang, S.-M.; et al. Network analysis and mechanisms of action of Chinese herb-related natural compounds in lung cancer cells. Phytomedicine 2019, 58, 152893. [Google Scholar] [CrossRef]
  65. Luo, W.; Sun, R.; Chen, X.; Li, J.; Jiang, J.; He, Y.; Shi, S.; Wen, H. ERK activation-mediated autophagy induction resists licochalcone A-induced anticancer activities in lung cancer cells in vitro. Onco Targets Ther. 2021, 13, 13437–13450. [Google Scholar] [CrossRef]
  66. Park, M.K.; Ji, J.; Haam, K.; Han, T.-H.; Lim, S.; Kang, M.-J.; Lim, S.S.; Ban, H.S. Licochalcone A inhibits hypoxia-inducible factor-1α accumulation by suppressing mitochondrial respiration in hypoxic cancer cells. Biomed. Pharmacother. 2021, 133, 111082. [Google Scholar] [CrossRef]
  67. Qiu, C.; Zhang, T.; Zhang, W.; Zhou, L.; Yu, B.; Wang, W.; Yang, Z.; Liu, Z.; Zou, P.; Liang, G. Licochalcone A inhibits the proliferation of human lung cancer cell lines A549 and H460 by inducing G2/M cell cycle arrest and ER stress. Int. J. Mol. Sci. 2017, 18, 1761. [Google Scholar] [CrossRef]
  68. Seo, Y.H. Discovery of licochalcone A as a natural product inhibitor of Hsp90 and its effect on gefitinib resistance in non-small cell lung cancer (NSCLC). Bull. Korean Chem. Soc. 2013, 34, 1917. [Google Scholar] [CrossRef]
  69. Tang, Z.H.; Chen, X.; Wang, Z.-Y.; Chai, K.; Wang, Y.-F.; Xu, X.-H.; Wang, X.-W.; Lu, J.-H.; Wang, Y.-T.; Chen, X.-P.; et al. Induction of C/EBP homologous protein-mediated apoptosis and autophagy by licochalcone A in non-small cell lung cancer cells. Sci. Rep. 2016, 6, 26241. [Google Scholar] [CrossRef]
  70. Yuan, L.-W.; Jiang, X.-M.; Xu, Y.-L.; Huang, M.-Y.; Chen, Y.-C.; Yu, W.-P.; Su, M.-X.; Ye, Z.-H.; Chen, X.; Wang, Y.; et al. Licochalcone A inhibits interferon-gamma-induced programmed death-ligand 1 in lung cancer cells. Phytomedicine 2021, 80, 153394. [Google Scholar] [CrossRef]
  71. Han, S.; Li, X.; Gan, Y.; Li, W. Licochalcone A promotes the ubiquitination of c-Met to abrogate gefitinib resistance. Biomed. Res. Int. 2022, 2022, 5687832. [Google Scholar] [CrossRef]
  72. Fan, X.; Guan, G.; Wang, J.; Jin, M.; Wang, L.; Duan, X. Licochalcone A induces cell cycle arrest and apoptosis via suppressing MAPK signaling pathway and the expression of FBXO5 in lung squamous cell cancer. Oncol. Rep. 2023, 50, 214. [Google Scholar] [CrossRef]
  73. Gao, F.; Li, M.; Yu, X.; Liu, W.; Zhou, L.; Li, W. Licochalcone A inhibits EGFR signalling and translationally suppresses survivin expression in human cancer cells. J. Cell Mol. Med. 2021, 25, 813–826. [Google Scholar] [CrossRef]
  74. Oh, H.-N.; Lee, M.-H.; Kim, E.; Yoon, G.; Chae, J.-I.; Shim, J.-H. Licochalcone B inhibits growth and induces apoptosis of human non-small-cell lung cancer cells by dual targeting of EGFR and MET. Phytomedicine 2019, 63, 153014. [Google Scholar] [CrossRef]
  75. Oh, H.-N.; Lee, M.-H.; Kim, E.; Kwak, A.-W.; Yoon, G.; Cho, S.-S.; Liu, K.; Chae, J.-I.; Shim, J.-H. Licochalcone D induces ROS-dependent apoptosis in gefitinib-sensitive or resistant lung cancer cells by targeting EGFR and MET. Biomolecules 2020, 10, 297. [Google Scholar] [CrossRef]
  76. Liang, M.; Li, X.; Ouyang, X.; Xie, H.; Chen, D. Antioxidant mechanisms of echinatin and licochalcone A. Molecules 2018, 24, 3. [Google Scholar] [CrossRef]
  77. Oh, H.-N.; Lee, M.-H.; Kim, E.; Kwak, A.-W.; Seo, J.-H.; Yoon, G.; Cho, S.-S.; Choi, J.-S.; Lee, S.-M.; Seo, K.-S.; et al. Dual inhibition of EGFR and MET by Echinatin retards cell growth and induces apoptosis of lung cancer cells sensitive or resistant to gefitinib. Phytother. Res. 2020, 34, 388–400. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of nicotine in NSCLC carcinogenesis.
Figure 1. Mechanisms of nicotine in NSCLC carcinogenesis.
Cimb 46 00352 g001
Figure 2. Chemical structures of licorice CHs [23,24].
Figure 2. Chemical structures of licorice CHs [23,24].
Cimb 46 00352 g002
Figure 3. Mechanisms of ILTG in nicotine-induced NSCLC treatment. (↓) decrease, (↑) increase.
Figure 3. Mechanisms of ILTG in nicotine-induced NSCLC treatment. (↓) decrease, (↑) increase.
Cimb 46 00352 g003
Figure 4. Mechanisms of licos in nicotine-induced NSCLC treatment. (↓) decrease, (↑) increase.
Figure 4. Mechanisms of licos in nicotine-induced NSCLC treatment. (↓) decrease, (↑) increase.
Cimb 46 00352 g004
Table 1. Therapeutic effects of licorice ILTG in nicotine-induced NSCLC.
Table 1. Therapeutic effects of licorice ILTG in nicotine-induced NSCLC.
Experimental ModelsNSCLC Cell LineDose/AdministrationTherapeutic EffectsRef.
In vitroA549A549 cells were treated with ILTG at 20 μM concentration for 24–72 h incubation at 37 °CAnti-proliferative, anti-invasion, anti-migration, apoptosis[46]
In vitroA549A549 cells were treated with ILTG at 0, 2, 10, 20, and 40 μmol/L concentrations for 12–72 h of incubation at 37 °CAnti-proliferative, apoptosis, G1 phase cell cycle arrest[47]
In vitroA549A549 cells were treated with ILTG at 10, 20, 30, 40, and 40 μM concentrations for 24–72 h of incubation at 42 and 70 °CAnti-proliferative, G2/M phase cell cycle arrest[48]
In vitroA549A549 cells were treated with ILTG and flavonoid glycosides (liquiritin, isoliquiritin) at 1.95–500 μg/mL concentrations for 24–72 h of incubationApoptosis, G2/M phase cell cycle arrest[49]
In vitroSK-LU-1SK-LU-1cells were treated with ILTG at 1.56, 3.13, 6.25, 12.5, 25, and 50 μg/mL concentrations for 24–72 h of incubationInhibition of cell growth[50]
In vitroA549, H1299NSCLC cells were treated with ILTG at 6.25, 12.5, and 25 μM concentrations for 24–72 h of incubation at 37 °CAnti-proliferative, anti-invasion, anti-migration[51]
In vitroA549A549 cells were treated with pure ILTG and ILTG nanosuspensions at 0.03, 0.06, 0.09, 0.12, 0.15, and 0.15 μM concentrations for 24–72 h incubation at 37 °CApoptosis[52]
In vitro/vivoA549, H1299, H1975NSCLC cells were treated with ILTG at 0, 3, and 10 μM concentrations for 24 h of incubation at 37 °C
Athymic nude mice were injected with a mixture of DMEM and Matrigel in a 1:1 ratio and then classified into 4 groups, in which each group received a vehicle: ILTG (25 mg/kg), tetrahydroxychalcone (25 mg/kg), or AZD0530 (20 mg/kg)
Anti-migration, inhibition of cell growth, and tumorigenesis[53]
In vitro/vivoA549, HCC827GR, H1975, H1650, and HCC827NSCLC cells were treated with ILTG at 0, 10, 20, and 40 μM concentrations for 24 h of incubation at 37 °C
Athymic nude mice were divided into 4 groups: vehicle, ILTG (1 mg/kg), ILTG (5 mg/kg), and gefitinib (5 mg/kg)
Apoptosis[54]
Table 2. The therapeutic effects of licorice licos in nicotine-induced NSCLC.
Table 2. The therapeutic effects of licorice licos in nicotine-induced NSCLC.
Experimental ModelsLico ClassificationNSCLC Cell LineDose/AdministrationTherapeutic EffectsRef.
In vitroLicoASK-LU-1SK-LU-1cells were treated with licoA at 1.56, 3.13, 6.25, 12.5, 25, and 50 μg/mL concentrations for 24–72 h of incubationInhibition of tumor growth[50]
In vitroLicoAH292H292 cells were treated with licoA at 0, 1, 10, 20, 40, and 80 μM concentrations for 24–96 h of incubation at 37 °CAnti-proliferative, apoptosis, and autophagy[62]
In vitroLicoAA549, H460NSCLC cells were treated with licoA at 0, 2, 5, 10, and 20 μM concentrations for 24 and 48 h of incubation at 37 °CAnti-migration, anti-invasion[63]
In vitroLicoAA549A549 cells were treated with licoA at 20, 40, and 80 μM concentrations for 24 h of incubation at 37 °CApoptosis, S and G2/M phase cell cycle arrest, autophagy[64]
In vitroLicoAA549, H23, H460, H1299, SPC-A1NSCLC cells were treated with licoA at 0, 5, 10, 20, 30, and 40 μM concentrations for 48 h of incubation at 37 °C for 7 daysApoptosis, autophagy[65]
In vitroLicoAHCT116, H1299, H322NSCLC cells were treated with licoA at 5, 10, 15, and 20 μM concentrations for 6 h of incubation at 37 °CAnti-viability[66]
In vitroLicoAA549, H460NSCLC cells were treated with licoA at 20, 40, and 60 μM concentrations for 4 and 12 h of incubation at 37 °CAnti-proliferative, apoptosis, G2/M phase cell cycle arrest[67]
In vitroLicoAH1975H1975 cells were treated with licoA at 0, 10, 30, 50, and 70 μM concentrations for 24–72 h of incubation at 37 °CAnti-proliferative[68]
In vitroLicoAA549, H1299NSCLC cells were treated with licoA at 0, 5, 10, 15, and 20 μM concentrations for 24 h of incubation at 37 °CApoptosis, sub-G1 phase cell cycle arrest, autophagy[69]
In vitroLicoAA549, H1299, H1650NSCLC cells were treated with licoA at 5, 10, 15, and 20 μM concentrations for 24 and 48 h of incubation at 37 °CAnti-proliferative, apoptosis[70]
In vivoLicoAHCC827, PC-9Female athymic nude mice were injected with 20 mg/kg licoA for a period of six weeksAnti-viability, apoptosis[71]
In vitro/vivoLicoAH226, H1703NSCLC cells were treated with licoA at 0, 10, 20, and 40 μM concentrations for 24–72 h incubation at 37 °C
BALB/c-nu mice (male) were divided into 4 groups: control (saline containing 20% SBE-β-CD), licoA (7.5 mg/kg), licoA (15 mg/kg), and cisplatin (2 mg/kg)
Anti-proliferative, inhibition of tumor growth, apoptosis, G1 phase cell cycle arrest[72]
In vitro/vivoLicoAA549, HCC827, H1975, and H3255NSCLC cells were treated with licoA at 0, 5, 10, 20, 40, 80, and 200 μM concentrations for 24–96 h of incubation at 37 °C
Athymic nude mice (female) were injected with 10 mg/kg licoA
Inhibition tumor growth, apoptosis[73]
In vitroLicoBHCC827, HCC827GRNSCLC cells were treated with licoB at 5, 10, 15, and 20 μM concentrations for 48 h of incubation at 37 °CAnti-proliferative, apoptosis, G2/M phase cell cycle arrest[74]
In vitroLicoDHCC827, HCC827GRNSCLC cells were treated with licoD at 5, 10, 15, and 20 μM concentrations for 48 h of incubation at 37 °CAnti-proliferative, apoptosis, G2/M phase cell cycle arrest[75]
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

Alsharairi, N.A. The Role of Licorice Chalcones as Molecular Genes and Signaling Pathways Modulator—A Review of Experimental Implications for Nicotine-Induced Non-Small Cell Lung Cancer Treatment. Curr. Issues Mol. Biol. 2024, 46, 5894-5908. https://doi.org/10.3390/cimb46060352

AMA Style

Alsharairi NA. The Role of Licorice Chalcones as Molecular Genes and Signaling Pathways Modulator—A Review of Experimental Implications for Nicotine-Induced Non-Small Cell Lung Cancer Treatment. Current Issues in Molecular Biology. 2024; 46(6):5894-5908. https://doi.org/10.3390/cimb46060352

Chicago/Turabian Style

Alsharairi, Naser A. 2024. "The Role of Licorice Chalcones as Molecular Genes and Signaling Pathways Modulator—A Review of Experimental Implications for Nicotine-Induced Non-Small Cell Lung Cancer Treatment" Current Issues in Molecular Biology 46, no. 6: 5894-5908. https://doi.org/10.3390/cimb46060352

Article Metrics

Back to TopTop