Next Article in Journal
Metabolomic Evidence for Peroxisomal Dysfunction in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
Previous Article in Journal
Novel Polyurethane Scaffolds Containing Sucrose Crosslinker for Dental Application
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insights into the Mechanisms of Action of Proanthocyanidins and Anthocyanins in the Treatment of Nicotine-Induced Non-Small Cell Lung Cancer

by
Naser A. Alsharairi
Heart, Mind & Body Research Group, Griffith University, Gold Coast, QLD 4122, Australia
Int. J. Mol. Sci. 2022, 23(14), 7905; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23147905
Submission received: 1 July 2022 / Revised: 15 July 2022 / Accepted: 16 July 2022 / Published: 18 July 2022
(This article belongs to the Section Molecular Oncology)

Abstract

:
In traditional medicine, different parts of plants, including fruits, have been used for their anti-inflammatory and anti-oxidative properties. Plant-based foods, such as fruits, seeds and vegetables, are used for therapeutic purposes due to the presence of flavonoid compounds. Proanthocyanidins (PCs) and anthocyanins (ACNs) are the major distributed flavonoid pigments in plants, which have therapeutic potential against certain chronic diseases. PCs and ACNs derived from plant-based foods and/or medicinal plants at different nontoxic concentrations have shown anti-non-small cell lung cancer (NSCLC) activity in vitro/in vivo models through inhibiting proliferation, invasion/migration, metastasis and angiogenesis and by activating apoptosis/autophagy-related mechanisms. However, the potential mechanisms by which these compounds exert efficacy against nicotine-induced NSCLC are not fully understood. Thus, this review aims to gain insights into the mechanisms of action and therapeutic potential of PCs and ACNs in nicotine-induced NSCLC.

1. Introduction

Lung cancer (LC) is considered the main diagnosed cancer causing death worldwide [1]. LC is broadly categorized into two major histologic classes: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC is further subclassified into squamous, adenocarcinoma and large cell carcinoma, which constitute 85% of smoking-attributable LC cases [2,3]. Tobacco products contain several carcinogenic compounds, such as tobacco-specific nitrosamines (i.e., NNN and NNK), which enhance production of DNA adducts in the lungs of smokers, thereby causing mutations of several NSCLC suppressor genes including protein p53 [4,5,6]. Although nicotine is considered non-carcinogenic, it may contribute to NSCLC development [7,8,9,10,11]. Nicotine enhances proliferation, angiogenesis and metastasis and inhibits apoptosis/autophagy in NSCLC cells by activating nicotinic acetylcholine receptors (nAChRs), especially the α7 subunit, and its downstream signaling pathways including the proto-oncogene serine/threonine kinase (Rb-RAF1), the phosphatidylinositol-3 kinase/serine/threonine kinase (PI3K/Akt), the mammalian target of rapamycin (mTOR), the nonreceptor tyrosine/kinase focal adhesion/protein kinase (Src/FAK/PKC) and the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) [12]. Nicotine also induces epithelial-to-mesenchymal transition (EMT), which is the key step in enhancing tumor progression in NSCLC cells, resulting in upregulation of several transcription and growth factors via activation of α7nAChR-mediated signaling pathways [12]. The mechanisms by which nicotine enhances tumor progression in NSCLC cells have been previously described in greater details [13]. In brief, nicotine binds to α7nAChR, activating the cellular signaling pathways involved in proliferation, metastasis, angiogenesis and anti-apoptosis/autophagy, which increases expression of EMT-associated molecules in NSCLC cells such as hypoxia inducible factor-1 (HIF-1α), vascular endothelial growth factor (VEGF), transforming growth factor β (TGF-β), deca-pentaplegic homolog (Smad), B-cell lymphoma-2 (Bcl-2), metalloproteinases (MMPs), cyclinD1, Snail, twist, vimentin, fibronectin and N-cadherin [13]. Figure 1 summarizes the mechanisms for nicotine in the development of NSCLC.
There is no clear recommendations on safe dietary supplements for use in treating NSCLC, particularly in smokers [14]. However, combination therapy of monoclonal antibody-based immunotherapy and chemotherapeutic agents has been shown to be a promising treatment strategy for NSCLC [15]. In addition, natural flavonoids present in fruits (e.g., citrus) in combination with chemotherapeutic agents, such as cisplatin, have shown anti-NSCLC effects, demonstrated by inhibition of the α7nAChR-mediated signaling pathways involved in cellular processes including proliferation, inflammation and anti-apoptosis [13,16].The health benefits of fruits are due to the high levels of bioactive flavonoids they contain, such as PCs and ACNs [17,18]. A large number of studies have documented the health benefits of natural flavonoids derived from fruits and plant foods, in relation to their biological attributes such as anti-diabetes/anti-cancer activity, reducing cardiovascular diseases and improving the blood lipid profile [17,18,19]. Thus, there is a real need to investigate natural flavonoid compounds, such as PCs and ACNs, as therapeutic agents for nicotine-induced NSCLC.
Flavanols include a group of natural compounds categorized according to their chemical structures into catechins and PCs, which are found in various plant-based foods. PCs, also known as condensed tannins, are polymeric and/or oligomeric pigments found in common plant-based foods (e.g., fruits, vegetables, cereal grains, and legumes), Cinnamomi Cortex (barks of Cinnamomum cassia used as a traditional Chinese medicine) and Vaccinium berries, for which a range of therapeutic effects have been reported including anti-cancer, antimicrobial, anti-diabetic, anti-obesity, cardioprotective and antioxidant properties [20]. Procyanidins are the most homo-oligomeric PC derivatives comprised of epicatechin/catechin monomeric, connected via the C4→C6/C4→C8 bond (B-type linkage) and the C2→O7 bond (A-type linkage), and dominated by dimers (e.g., procyanidin A1-A2/B1-B8), trimers (e.g., selligueain A/B and procyanidin C1/C2), and tetramers (degree of polymerization ranged from 5 to 11) [20,21,22].
ACNs, featuring six common glycosylated forms of anthocyanidins (i.e., malvidin, pelargonidin, cyanidin, petunidin, delphinidin and peonidin) with hydroxyl (OH) moieties in their structure at the 3 position on the C-ring are water-soluble pigments belonging to flavonoids responsible for producing various colors in fruits, berries and vegetables that exert a protective effect against diabetes, cardiovascular and neurodegenerative diseases and cancers (including LC) [23,24,25].
In various plant species, PCs and ACNs share the first and last steps of flavonoid biosynthesis and its regulatory genes including dihydroflavonol reductase, flavanone 3β-hydroxylase and chalcone isomerase/synthase [26,27]. PCs and ACNs haveshown to be the key compounds contributing to the intense color of cereals, fruits, vegetables and blueberries [28], and they have been reported to reduce the production of reactive oxygen species (ROS) in some berry fruits, which may make them potential anti-antioxidant agents [29]. Development and ripening of plant species involve complex processes that result in substantial structural and metabolic changes such as changes in texture and flavor formation and pigment accumulation [30]. These processes are coordinated by abscisic acid (ABA) and other ripening-related genes, which regulate PCs and ACNs biosynthesis [30]. Ripening is considered an important stage where major flavonoids are accumulated [31]. A diverse range of plant phytohormones including cytokinin, gibberellins, auxin, jasmonic acid and brassinosteroids are involved in regulating different aspects of fruit development and ripening through multilevel regulatory mechanisms [31]. PCs are the main compounds detected at the early stage of plant development, whereas ACNs accumulate early in ripening [31]. ACNs act as the major colored pigments attracting animals and insects for seed dispersal in berry fruits [30]. In climacteric fruit ripening, ethylene plays a key hormonal role in stimulating the differential expression of many gene encoding transcription factors that regulate pigments accumulation, cell wall softening, texture change and aroma, flavor and skin color development [32,33].
A recent reviews showed that particular flavonoid compounds (e.g., quercetin, kaempferol, epigallocatechin gallate, and delphinidin) derived from plant-based foods act against NSCLCin in vitro/in vivo models [16,34]. However, the molecular mechanism of PCs and ACNs with regard to their preventive effects against nicotine-induced NSCLC have not been fully elucidated. A recent review showed that flavone-rich extracts from Scutellaria baicalensis, and baicalein, baicalin, wogonin, wogonoside and oroxylin A, in particular, serve as potential adjuvant agents for the treatment of nicotine-induced NSCLC. These compounds exert anti-inflammatory, anti-angiogenesis, anti-proliferative and apoptotic/autophagic effects through inhibiting several cellular signaling pathways as potential molecular mechanisms [13].
The therapeutic effects of PCs and ACNs against nicotine-induced NSCLC may depend on their bioavailability, that is, absorption, distribution and excretion. ACNs are distinguishable from other flavonoids because of its apparent low bioavailability [35,36]. However, studies have demonstrated that ACNs have higher bioavailability and bioactivity in vitro than in vivo [37]. ACNs are absorbed in the blood, distributed to target tissues and excreted into bile as the glycosylated structure [35]. The absorption efficiencies of the hydrophobic ACNs are greater than the hydrophilic ones in vitro [38]. In vitro studies showed that PAs oligomers are degraded into monomers and dimers in acidic gastric juice by human fecal microflora, which are then absorbed as intact molecules in the colon and excreted into urine. In vivo, epicatechin (EC) and methylated EC, as the major metabolites of PAs dimers, are absorbed in the intestinal mucosa in very low contents, catabolized by fecal microflora fermentation into low molecular phenolic acids and then excreted into urine [39]. Given that PCs and ACNs are used as therapeutic agents against many chronic diseases, targeting these compounds might help our understanding of the mechanisms involved in nicotine-induced NSCLC treatment, particularly in smokers. To date, no reviews have discussed the molecular mechanisms of PCs and ACNs in nicotine-induced NSCLC treatment. Thus, this paper highlights the mechanisms of action and therapeutic potential of PCs and ACNs in nicotine-induced NSCLC.

2. Methods

A literature review was undertaken to identify the published studies exploring the potential molecular mechanisms of PCs and ACNs in nicotine-induced NSCLC treatment from 2000 up to June 2022 via searches of PubMed/MEDLINE. The following search terms were used:“proanthocyanidins” OR “procyanidin” AND “anthocyanins” OR “malvidin” OR “pelargonidin” OR “cyanidin” OR “petunidin” OR “delphinidin” OR “peonidin” AND “NSCLC”OR “lung cancer” AND “nicotine” OR “cigarette smoke”. The search included all types of study design published in English. All studies were identified and reviewed by the author. From a total of 202 articles searched using the databases, 30 studies were selected based on the search terms.

3. Proanthocyanidins in Nicotine-Induced NSCLC Treatment

Several studies demonstrated the therapeutic effects of PC-rich extracts from plant-based foods and/or medicinal plants at different nontoxic concentration against nicotine-induced NSCLC. Cranberry-derived PCs suppress tumor cell growth in NSCLC cells [40,41], but the mechanisms for this action have not been well investigated [40]. Treatment with cranberry PCs resulted in a significant induction of apoptosis and cell cycle arrest in NSCLC cells via upregulating the expression of pro-apoptotic-related markers (e.g., cytochrome c and caspase 3) [42].
PCs from grape seed extract have shown promising results in nicotine-induced NSCLC treatment. For example, using the in vivo proteolysis/antitumor assay and the in vitro proteolysis/angiogenesis assay, PCs inhibit angiogenesis-mediated tumor growth in NSCLC cells, in part by suppressing vascular extracellular matrix (ECM) proteolysis byMMP-2 [43]. Treatment of NSCLC cells with PCs using the in vivo tumor xenograft assay and the in vitro 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for cell proliferation/survival resulted in suppression of cell proliferation in vitro/vivo, and inhibition of angiogenesis and induction the apoptotic cell death of tumor cells in vivo. Such effects are mediated by upregulation of insulin-like growth factor binding protein-3 (IGFBP-3) levels and inhibition of proliferating cell nuclear antigen (PCNA) in the tumor microenvironment [44]. A study used the in vivo tumor xenograft assay and the in vitro cell deathenzyme-linked immunosorbent assay (ELISA) and MTT assay for assessing proliferation of NSCLC cells showed that PCs cause proliferation inhibition and apoptosis induction via the inhibition of cyclooxygenase-2 (COX-2) expression and prostaglandin-2 (PGI-2) receptors in NSCLC cells [45]. The mechanism underlying the anti-migration effect of PCs on NSCLC cells involve inhibiting nitric oxide (NO) synthase, N(G)-nitro-L-arginine methyl ester (L-NAME), and the ERK1/2 and MAPK signaling pathways [46]. The anti-proliferative/apoptotic effects of PCs onNSCLC cells are mediated via the activation of caspase 3 expression, prostacyclin synthase (PTGIS)/PGI2 (as measured by 6-keto PGF1α), and 15-lipoxigenase-2/15(S)-hydroxyeicosatetraenoic acid (15-LOX-2)/15-HETE production [47]. PCs showed the inhibitory effects on the cigarette smoke condensate (CSC)-induced migration of NSCLC cells through inhibition of NADPH oxidase (NOX)-induced oxidative stress and EMT transition [48]. Treatment with PCs using the colorimetric caspase-3 activity assay in vivo and in vitro showed apoptotic effects through increased expression of pro-apoptotic markers (e.g., poly ADP ribose polymerase (PARP); Bcl-2-associated X protein (Bax)), and decreased expression of apoptotic markers (e.g., Bcl-2 and cyclins) [49]. A study used the in vivo tumor xenograft assay and the in vitro MTT and miR-106b ISH assays showed that PCs promote anti-proliferative/invasive effects on NSCLC cells via downregulating miR-106b expression and upregulating cyclin-dependent kinase inhibitor 1A (CDKN1A) mRNA and p21 expression [50].
A few studies on NSCLC cells after treatment with the Cinnamomi Cortex extract PCs showed a significant reduction in nuclear factor-E2-related factor 2(Nrf2) expression, and insulin-like growth factor-1 receptors (IGF-1R) were responsible for induced proliferation [51,52]. Cinnamomi Cortex extract procyanidin C1 exert anti-metastatic activity by suppressing TGF-β-induced EMT in NSCLC cells [53].
Treatment with PCs inhibits hydrogen peroxide (H2O2)-induced NSCLC cell viability, as shown by reduced ROS and malondialdehyde (MDA) production, hydrogen peroxide(H2O2)-induced oxidative stress, and promoted the expression of Nrf2 target genes [54]. PCs inhibit proliferation, viability, along with induction of apoptosis and G2/M cell cycle arrest in NSCLC cells. This is triggered by inhibiting the EMT-related molecules (e.g., N-cadherin and vimentin), expression of apoptotic markers (e.g., Bcl-2), and increasing expression of pro-apoptotic markers (e.g., Bax) via downregulating the Janus kinase/signal transducer and activator of transcription3 (JAK2/STAT3) signaling pathway [55].
Treatment with prodelphinidin B-2 3′-O-gallate, a proanthocyanidin gallate, resulted in the upregulation of key transcription factors such as the soluble Fas ligand (sFasL) and membrane-bound Fas ligand (mFasL), which are responsible for the anti-proliferative and apoptotic activities in NSCLC cells [56,57]. Cinnamtannin D1, an A-type procyanidin trimer, from Rhododendron formosanum extracts has been found to exhibit autophagic effects on NSCLC cells via inhibition of cellular signaling pathways (e.g., mTOR) [58].
These results suggest that plant-derived natural PCs may play a significant role as anti-NSCLC agents by suppressing proliferation, migration, invasion, viability, metastasis, angiogenesis, and promoting apoptosis/autophagy via inhibition/activation of transcription factors and/or multiple cellular signaling pathways induced by α7nAChR in NSCLC cells. Table 1 highlights the molecular mechanisms of PCs in nicotine-induced NSCLC treatment.

4. Anthocyanins in Nicotine-Induced NSCLC Treatment

ACNs-rich extracts from plant-based foods/medicinal plants at various nontoxic concentrations have exerted therapeutic effects against nicotine-induced NSCLC, as demonstrated by several studies. Delphinidin and cyanidin have been shown to inhibit platelet-derived growth factor (PDGF)(AB)-induced VEGF expression in vascular smooth muscle cells through attenuation of the p38 MAPK and c-JUN NH2-terminal kinase (JNK) signaling pathways [59], known as α7nAChR-mediated cascades involved in NSCLC.ACNs extracted from Vitis coignetiae Pulliat inhibit the expression of several transcription and growth factors (e.g., MMP2/9 and VEGF) involved in proliferation, angiogenesis, invasion, and migration of NSCLC cells [60]. The extracts from Morus alba L. (cyanidin 3-rutinoside and cyanidin 3-glucoside) inhibit the migratory and invasive activities of NSCLC cells [61]. Cyanidin-3-glucoside not only inhibits the proliferation, invasion, and migration, but also induces apoptosis. The mechanism underlying such an effect is associated with inhibiting p53-induced gene 3 (TP53I3) expression in NSCLC cells and the downregulation of cellular signaling pathways (PI3K/Akt/mTOR) involved in NSCLC progression [62].
Delphinidin inhibits tumor growth, angiogenesis, proliferation, and induces apoptosis in NSCLC cells using the in vivo Matrigel plug assay and the in vitro MTT/ELISA assay by upregulating pro-apoptotic expression (e.g., Bax and caspase 3/9), along with downregulating epidermal growth factor receptor (EGFR), cobalt chloride (CoCl2)-induced HIF-1α, Bcl-2, PCNA, cyclin D1, and VEGF mRNA expression via inhibiting of several signaling pathways [63,64].
A combination of five ACN extracts (i.e., delphinidin, peonidin, petunidin, cyanidin, and malvidin) from bilberry and blueberry resulted in inhibited growth and metastasis, and induced apoptosis and cell cycle arrest of NSCLC cells in vitro. The mechanism of action of ACNs involves inhibiting activation of multiple signaling pathways, including TNFα-induced NF-kB, Notch, Wnt/β-catenin, and their key transcription factors (i.e., cyclin D1/B1, VEGF, p-ERK, bcl-2, and MMP9). Furthermore, when evaluated with an in vivo model using the in vivo xenograft assay, delphinidin alone, and the ACN mixture, resulted in significantly inhibited growth of H1299 cells [65]. ACNs derived from Syzygium cumini L. (known as Indian blackberry) showed significant anti-proliferative effects on NSCLC cells, but the mechanisms for this action have not been observed [66].
These results suggest that ACNs may have a significant role in anti-proliferative, anti-invasive, anti-angiogenic, anti-metastatic, and apoptotic/autophagic effects in NSCLC cells by suppressing the activation of key transcription/growth factors and α7nAChR-mediatedcellular signaling pathways. The molecular mechanisms of ACNs in nicotine-induced NSCLC treatment are summarized in Table 2.

5. Combination of Proanthocyanidins and Anthocyanins with Chemotherapeutics/Radiotherapy in Nicotine-Induced NSCLC Treatment

Several studies suggest that PCs and ACNs used in combination with radiotherapy and/or chemotherapy have the ability to modulate the activity of cellular signaling pathways involved in proliferation, inflammation as well as in anti-apoptosis and/or cell cycle arrest in NSCLC cells. Treatment with PCs extracted from Cinnamomi Cortex inhibits Nrf2-mediated proliferation through promoting sensitivity of NSCLC cells to the cytotoxic action of chemotherapeutic drug etoposide and doxorubicin [67]. A recent study using the CCK8 and ELISA assays reported anti-inflammatory and apoptosis-inducing effects of PCs on NSCLC cells when used in combination with ionizing radiation. The results indicated that such combinations inhibit tumor growth in vivo and reduce interferons/interleukins production and increase pro-apoptotic p53 target genes in vitro via upregulating the MAPK signaling pathway [68]. Delphinidin treatment with γ-ionizing radiation (IR) induced autophagy and apoptosis in NSCLC cells by increasing pro-apoptotic expression (e.g., caspase-3 and LC3), and reduced p-ERK1/2 expression via activation of the MAPK signaling pathway [69]. Cyanidin-3-glucoside, but not pelargonidin-3-glucoside, has been recently reported to inhibit the mRNA level of claudin-2 (CLDN2)-induced chemotherapy resistance to doxorubicin through the downregulation of p-Akt and the upregulation of p38 MAPK, leading to apoptosis and necrotic cell death in NSCLC cells [70]. Table 3 summarizes the molecular mechanisms of PCs and ACNs in combination with radiotherapy/chemotherapy in nicotine-induced NSCLC treatment.

6. PCs and ACNs as Positive Modulators of Human α7nAChR

Findings of this review suggest that PCs and ACNs exert anti-tumor functions against NSCLC, due to their potential as positive modulators of α7nAChR. Treatment of NSCLC cells with varying concentrations suppresses the growth of these cells by 50% relative to the untreated control (GI50). The concentrations used both in vivo and in vitro studies are not associated with gross/visible toxicity. ACNs overdose possesses no significant cytotoxic effects on NSCLC cells in vivo. The cytotoxicities of anti-NSCLC drugs and/or ionizing radiation are significantly improved in the presence of 100–300 µg/mL Cinnamomi Cortex PCs in vitro, 20 µg/mL grape seed PCs in vivo/vitro, and ≤50 µM ACNs in vitro.
PCs vary in the number, configuration, arrangement and substitution of OH groups on the B ring. In general, all PCs have OH groups on C-3, C-5, and C-7 positions, respectively, and on their B rings [22]. The anti-NSCLC activity of PCs may rely on the number of OH groups on their B-ring, and the OH group at C-3 plays a significant role in their significant anti-tumor ability [71]. Procyanidins have five OH groups on positions C3,5,7,4′,5′, with one OH group on the C ring and two OH groups on the A and B rings [22]. Procyanidins may interact with α7nAChR binding site and thus could be considered as a potential target for the treatment of nicotine-induced NSCLC.
The number and position of OH and methoxyl groups on the flavylium B-ring of ACNs may play a significant role in the suppression of α7nAChR and its downstream signaling pathways. The presence of one OH group or methoxy substituents on the B-ring may result in a considerable loss of inhibitory effects in NSCLC cells. Delphinidin, which has three OH moieties at C-3′,4′,5′ of the B-ring, exerts greater inhibitory activity than cyanidin and pelargonidin, which have two and one OH group, respectively. The presence of two OH groups, one methoxyl group in petunidin, and one OH group with methoxy substituents in malvidin and peonidin make them less inhibitory effects on NSCLC cells [25]. Thus, ACNs would be expected to have high levels of effective anti-NSCLC agents in binding to the α7nAChR-mediated signaling pathways, which may result in suppression of the receptor in a concentration-dependent manner.
Understanding conditions associated with nicotine-α7-nAChR signaling might provide an opportunity to explore the therapeutic potential of PCs and ACNs in nicotine-induced NSCLC. Among several types of α subunits, the homomeric α7nAChR has been shown to be the key receptor activating NNK and/or nicotine-mediated cell proliferation, angiogenesis and anti-apoptosis in NSCLC cells in vivo and in vitro [10,72]. High levels of the α7 mRNA levels in squamous cell carcinoma tumors are found in smokers than nonsmokers [73]. NNK has been shown to be a high affinity agonist for α7nAChR in LC cells [74]. Nicotine is converted to NNK via activation of the α7nAChR and β-adrenergic receptor (β-AR) [8]. NNK binds to α7nAChR and β-AR causes activation of NSCLC-promoting signaling pathways [10,72]. NNK induces production of ROS in vitro, which causes oxidative DNA damage in NSCLC cells by activating the Wingless-type protein (Wnt) signaling pathway [75]. NNK or nicotine induces NSCLC cell proliferation, invasion and angiogenesis in vivo and in vitro through α7nAChR with activation of COX-2 expression, leading to an upregulation of PGE2, which is implicated in NSCLC development [76,77]. NNK enhances proliferation of NSCLC cells in vitro by activating PCNA expression via PI3K/AKT/Stat3/ERK1/2 signaling pathway activation [78]. In vitro, treatment with nicotine causes an induction effect on α7nAChR, along with significant stimulation of p-ERK1/2 cascade, accompanied by promotes the nAChR-mediated production of neurotransmitter noradrenaline in the brain, which enhances β-AR signaling-mediated DNA synthesis of NSCLC cells, resulting in promoted cell proliferation [79]. Nicotine influences NSCLC biology involve inhibition of apoptosis induced by chemotherapy and radiotherapy, and induction of proliferation and angiogenesis of NSCLC cells. Such effect is mediated by α7nAChR, which stimulates signaling pathways such as PI3K/Akt/mTOR, suppresses of the mitochondrial death pathway or induces of pro-angiogenic factors [80]. The binding of nicotine to the α7nAChRin vivo/vitro upregulates multiple downstream pathways (e.g., MAPK/ERK, Src, JAK, and STAT) and induces the secretion of growth/angiogenic factors (e.g., EGF and VEGF) in NSCLC cells, and such effect could be mediated by inactivation of p53 expression [9]. Binding of nicotine to α7nAChR enhanced voltage-gated Ca2+ channels, which result in induction of NSCLC cell angiogenesis by activating of Ca2+ influx [10,72]. Nicotine induces NSCLC angiogenesis, migration and invasion in vitro by inhibiting HIF-1α and its downstream target gene VEGF. Such effect is mediated via α7nAChR-mediated Src, Ca+2, PI3K, MAPK/ERK1/2, and mTOR signaling pathways activation [81]. Nicotine promotes NSCLC cell proliferation in vitro via protein arrestin, β1 (ARRB1)-mediated activation of E2F transcription factors 1 (E2F1), and Src and Rb-Raf-1 signaling downstream of α7nAChR [82,83]. Nicotine or NNK induces NSCLC cell proliferation in vivo and in vitro by activating Akt phosphorylation and its related downstream substrates, including GSK-3β, mTOR and NFκB [84]. In vitro, proliferation, invasion and angiogenesis of NSCLC cells are found to be promoted by nicotine via α7nAChR with activation of Signal transducer and activator of transcription1 (STAT1), E2F1, as well as induction of Src, PI3K, and Rb-Raf signaling pathways [85]. The apoptotic family members, including Bcl-XL, c-Myc, and Bcl2, are induced in response to nicotine/NNK treatment in NSCLC cells in vitro via α7nAChR-mediated ERK1/2, Raf1 and MAPKs signaling pathways activation, resulting in suppression of apoptosis [86]. In vivo, nicotine or NNK-induced activation of the PI3K/Akt/mTOR pathway via stimulation of α7nAChR inhibits Bax expression [87]. NNK treatment inhibits miRNAs expression in NSCLC cells in vivo. This is observed through upregulation of cytochrome P450 (CYP) 2A3 expression as a mechanism of such effect [88]. NNK induces NSCLC progression in vitro by altering the expression of mismatch DNA repair (MMR), leading to miRNAs deregulation, particularly miR-422a, miR-155, and miR-21 [89]. NNK induces proliferation and inhibits apoptosis in NSCLC cells in vivo by downregulating miR-124 expression, leading to the activation of Akt, thereby enhancing NSCLC progression [90].
PCs may have the ability to inhibit β-amyloid (Aβ) aggregation [91], which is the key hallmark of NSCLC [92,93]. Aβ is one of the key mechanisms responsible for LC, as shown in increased Aβ isoforms in A549 cells, particularly Aβ40 and Aβ42. Inactivation of p53 was found to increase Aβ40 and Aβ42 levels in A549 cells via upregulation of the PI3K/Akt/NFκB signaling pathway and MMP2/9 expression, known to increase viability and reduce apoptosis in NSCLC cells, suggesting that activation of p53 may be helpful in reducing the levels of Aβ40 and Aβ42 responsible for viability and anti-apoptosis in NSCLC cells [93]. Aβ40 and Aβ42 contain amino acids serine (Ser) and tyrosine (Tyr) (H6DSGY10 and G25SNKG29), which are known to be the major toxic Aβ forms. Ser26 and Tyr10 of Aβ peptides have been shown to interact with acetylcholinesterase (AChE), a cholinergic enzyme-mediated neurotransmitter acetylcholine (Ach) associated with amyloid fibril formation in A549 cells, leading to increased cytotoxic response in these cells [94]. Ach produced by AchE from acetyl-coenzyme A (acetyl-CoA) and choline plays a keyrole as paracrine/autocrine growth factor in the lung homeostasis, which binds to nAChRs on the NSCLC cells [95,96]. The expression of AchE has been reported to be reduced in NSCLC cells. The decrease in AchE activity leads to elevate Ach levels in NSCLC cells, which could promote cell proliferation, migration, invasion, angiogenesis, and reduce apoptosis/autophagy through activating Ca2+ channels, TGF-β-induced EMT and α7nAChR-mediated signaling pathways such as PI3K/Akt, ERK/MAPK, GSK-3β, and STAT3 [97]. This suggests that Ach may have therapeutic potential in nicotine-induced NSCLC cells by upregulating expression of p53, which promotes apoptosis/autophagy and reduce proliferation and angiogenesis in NSCLC cells. Given that PCs reduce cellular growth and proliferation in NSCLC cells via upregulating p53 expression, targeting such natural compound in treatment regimes might help in downregulating cellular signaling pathways, as the mechanisms involved in Aβ-induced proliferation and anti-apoptosis in NSCLC cells.
Activation of Ach was also found to promote inflammation and pro-inflammatory cytokines in pulmonary tissue [98]. Increased concentrations of pro-inflammatory cytokines (i.e., IL-6, IL-8, IL-1β, and TNF-α) have been reported in NSCLC cells, particularly H460 and A549 cells [99]. The potential mechanism underlying such increases involves activating the α7nAChR-mediated JAK1-dependent STAT3 signaling by receptor tyrosine kinase (RTK) such as VEGFR and mutant-EGFR [100,101]. Therefore, ACNs may have significant anti-inflammatory functions via reducing of Aβ aggregation in NSCLC cells, leading to the modulation of α7nAChR and its downstream signaling pathways.
Previous studies showed that microRNAs (miRs), including miR-205-3p, a class of noncoding RNA molecules, act as potential markers for the progression of NSCLC [102,103]. A recent in vitro experiment has suggested that the Aβ precursor protein-binding family B member 2 (APBB2)-mediated activation of dysregulated miR-205-3p expression in different NSCLC cells, including H460, H1299, 95-D, and A549, resulted in activation of proliferation and inhibition of apoptosis in NSCLC cells [104]. PCs and ACNs may directly bind to Aβ to reduce its aggregation, suggesting that these compounds could be useful in reducing miR-205-3p expression involved in the pathogenesis of NSCLC. PCs and ACNs may thus be useful for their anti-Aβ aggregation activity via reducing miR-205-3p expression, resulting in an inhibition of proliferation and induction of apoptosis in NSCLC cells.
ACNs may bind to the active site of α-L-rhamnosidases (α-R) enzyme, having an inhibitory effect on NSCLC cells by inhibiting the cellular signaling pathways involved in proliferation and anti-apoptosis/autophagy. α-R is involved in ACNs metabolism, which hydrolysis of glycosidic bonds, and thus, may improve the bioavailability as well as the anti-tumor activity of ACNs in nicotine-induced NSCLC cells. For example, α-R enzyme has been demonstrated to catalyze the hydrolysis of cyanidin-3-rutinoside to cyanidin-3-glucoside in black raspberry from Aspergillus spp. [105] and in red mulberry (Morus alba L.) fruit [106], which are known for their anti-NSCLC activity. Thus, α-R may provide highly biologically active ACNs, which suppress proliferation and induction of apoptosis/autophagy in NSCLC cells.
Taken together, nicotine-mediated activation of cell proliferation, migration, invasion, angiogenesis, and inhibition of apoptosis in NSCLC cells can be modulated when PCs and ACNs interact with the active site of α7AChR, displaying complex modulation of several transcription/growth factors via signaling pathways involved in α7nAChR activation, suggesting that such compounds may have a significant role in the treatment of NSCLC (Figure 2).

7. Limitations

Although the available in vivo and in vitro studies suggest a therapeutic potential of PCs and ACNs in nicotine-induced NSCLC, these studies are used different dosage regimens in the treatment, which questions the reliability of results from these studies. Different doses of PCs and ACNs may affect diverse transcription factors and multiple cellular signaling pathways, and thereby affecting targeted nicotine-induced NSCLC therapy. The optimal effective dose of PCs and ACNs are difficult to determine, as it varied between studies. PCs and ACNs could be effective in nicotine-induced NSCLC treatment if used at concentration of up to 100 μM/µg/mL, but further studies are needed to apply this in vitro anti-NSCLC effects.

8. Conclusions

PCs and ACNs are flavonoid pigments with anti-NSCLC effects that exist in various plant-based foods and/or medicinal plants. The potential molecular mechanisms by which PCs and ACNs from plants-based foods and medicinal plants exert anti-tumor activity with therapeutic potential in nicotine-induced NSCLC have not been elucidated. Treatment with PCs and ACNs at various nontoxic concentrations have demonstrated several activities against nicotine-induced NSCLC, including anti-proliferative, anti-migration, anti-metastatic, anti-invasive, anti-angiogenic, and apoptosis/autophagy induction. PC-rich extracts from grape seed and/or Cinnamomi Cortex used in combination with irradiation/chemotherapy may have useful anti-proliferative, anti-inflammatory and apoptotic efficacy against nicotine-induced NSCLC. Delphinidin and cyanidin may promote apoptotic/autophagic activity by enhancing the chemosensitivity and/or radiosensitivity of NSCLC cells.
Nicotine and/or NNK may contribute to NSCLC development by promoting cell proliferation, migration, invasion, angiogenesis, and inhibiting apoptosis. Such effects may occur through α7nAChR-mediated signaling pathways activation. PCs and ACNs may act as selective agonists of α7nAChR, where they can inhibit the activation of α7nAChR in NSCLC cells. PCs and ACNs may directly bind to α7nAChR, suggesting that these compounds could be useful in modulating multiple transcription/growth factors and signaling pathways. PCs may act as anti-NSCLC agents through inhibiting excessive accumulation of Aβ involved in nicotine-induced NSCLC. In addition, the α-R enzyme involved in deglycosylation of ACNs in plant-based foods may play a key therapeutic role in nicotine-induced NSCLC, as they are involved in the anti-proliferative and apoptotic/autophagic activities against NSCLC cells. Therefore, plant-based foods and/or medicinal plants-derived PCs and ACNs may have therapeutic efficacy as anti-NSCLC medicinal agents in smokers due to their anti-proliferative, anti-angiogenic, anti-migration, anti-invasive, anti-inflammation, anti-metastatic, and apoptosis/autophagy activities, but further studies are needed to better understand the underlying mechanisms.

Funding

This review received no financial support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Bray, F.; Jacques, F.; 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] [PubMed] [Green Version]
  2. Molina, J.R.; Yang, P.; Cassivi, S.D.; Schild, S.E.; Adjei, A.A. Non-small cell lung cancer: Epidemiology, risk factors, treatment, and survivorship. Mayo Clin. Proc. 2008, 83, 584–594. [Google Scholar] [CrossRef]
  3. Alsharairi, N.A. The effects of dietary supplements on asthma and lung cancer risk in smokers and non-smokers: A review of the literature. Nutrients 2019, 11, 725. [Google Scholar] [CrossRef] [Green Version]
  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] [PubMed] [Green Version]
  5. Hecht, S.S. Lung Carcinogenesis by tobacco smoke. Int. J. Cancer 2012, 131, 2724–2732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hecht, S.S.; Stepanov, I.; Carmella, S.G. Exposure and metabolic activation biomarkers of carcinogenic tobacco-spesficnitrosommines. ACS Chem. Res. 2016, 49, 106–114. [Google Scholar] [CrossRef] [Green Version]
  7. Tournier, J.-M.; Birembaut, P. Nicotinic acetylcholine receptors and predisposition to lung cancer. Curr. Opin. Oncol. 2011, 23, 83–87. [Google Scholar] [CrossRef]
  8. Warren, G.W.; Singh, A.K. Nicotine and lung cancer. J. Carcinog. 2013, 12, 1. [Google Scholar] [CrossRef]
  9. Schaal, S.; Chellappan, S.P. Nicotine-mediated cell proliferation and tumor progression in smoking-related cancers. Mol. Cancer Res. 2014, 12, 14–23. [Google Scholar] [CrossRef] [Green Version]
  10. Wang, S.; Hu, Y. α7 nicotinic acetylcholine receptors in lung cancer (Review). Oncol. Lett. 2018, 16, 1375–1382. [Google Scholar] [CrossRef] [Green Version]
  11. Zhao, H.; Wang, Y.; Ren, X. Nicotine promotes the development of non-small cell lung cancer through activating LINC00460 and PI3K/Akt signaling. Biosci. Rep. 2019, 39, BSR20182443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. 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]
  13. 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]
  14. Alsharairi, N.A. Supplements for smoking-related lung diseases. Encyclopedia 2021, 1, 76–86. [Google Scholar] [CrossRef]
  15. Dafni, U.; Tsourti, Z.; Vervita, K.; Peters, S. Immune checkpoint inhibitors, alone or in combination with chemotherapy, as first-line treatment for advanced non-small cell lung cancer. A systematic review and network meta-analysis. Lung Cancer 2019, 134, 127–140. [Google Scholar] [CrossRef] [PubMed]
  16. Zanoaga, O.; Braicu, C.; Jurj, A.; Rusu, A.; Buiga, R.; Berindan-Neagoe, I. Progress in research on the role of flavonoids in lung cancer. Int. J. Mol. Sci. 2019, 20, 4291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Skrovankova, S.; Sumczynski, D.; Mlcek, J.; Jurikova, T.; Sochor, J. Bioactive compounds and antioxidant activity in different types of berries. Int. J. Mol. Sci. 2015, 16, 24673–24706. [Google Scholar] [CrossRef] [Green Version]
  18. Sayago-Ayerdi, S.; García-Martínez, D.L.; Ramírez-Castillo, A.C.; Ramírez-Concepción, H.R.; Viuda-Martos, M. Tropical fruits and their co-products as bioactive compounds and their health effects: A review. Foods 2021, 10, 1952. [Google Scholar] [CrossRef]
  19. Samtiya, M.; Aluko, R.E.; Dhewa, T.; Moreno-Rojas, J.M. Potential health benefits of plant food-derived bioactive components: An overview. Foods 2021, 10, 839. [Google Scholar] [CrossRef]
  20. Smeriglio, A.; Barreca, A.; Bellocco, E.; Trombetta, D. Proanthocyanidins and hydrolysable tannins: Occurrence, dietary intake and pharmacological effects. Br. J. Pharmacol. 2017, 174, 1244–1262. [Google Scholar] [CrossRef] [Green Version]
  21. Rue, E.A.; Rush, M.D.; van Breemen, R.B. Procyanidins: A comprehensive review encompassing structure elucidation via mass spectrometry. Phytochem. Rev. 2017, 17, 1–16. [Google Scholar] [CrossRef]
  22. Mannino, G.; Chinigò, G.; Serio, G.; Genova, T.; Gentile, C.; Munaron, L.; Bertea, C.M. Proanthocyanidins and where to find them: A meta-analytic approach to investigate their chemistry, biosynthesis, distribution, and effect on human health. Antioxidants 2021, 10, 1229. [Google Scholar] [CrossRef] [PubMed]
  23. Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lin, B.-W.; Gong, C.-C.; Song, H.-F.; Cui, Y.-Y. Effects of anthocyanins on the prevention and treatment of cancer. Br. J. Pharmacol. 2017, 174, 1226–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mattioli, R.; Francioso, A.; Mosca, L.; Silva, P. Anthocyanins: A comprehensive review of their chemical properties and health effects on cardiovascular and neurodegenerative diseases. Molecules 2020, 25, 3809. [Google Scholar] [CrossRef]
  26. Winkel-Shirley, B. Flavonoid biosynthesis. A colorful model for genetics, biochemistry, cell biology, and biotechnology. Plant Physiol. 2001, 126, 485–493. [Google Scholar] [CrossRef] [Green Version]
  27. Marles, M.A.S.; Ray, H.; Gruber, M.Y. New perspectives on proanthocyanidin biochemistry and molecular regulation. Phytochemistry 2003, 64, 367–383. [Google Scholar] [CrossRef]
  28. Limtrakul, P.; Semmarath, W.; Mapoung, S. Anthocyanins and proanthocyanidins in natural pigmented rice and their bioactivities. In Phytochemicals in Human Health; Intechopen: London, UK, 2019. [Google Scholar] [CrossRef] [Green Version]
  29. Wu, X.; Gu, L.; Prior, R.L.; McKay, S. Characterization of anthocyanins and proanthocyanidins in some cultivars of Ribes, Aronia, and Sambucus and their antioxidant capacity. J. Agric. Food Chem. 2004, 52, 7846–7856. [Google Scholar] [CrossRef]
  30. Karppinen, K.; Zoratti, L.; Nguyenquynh, N.; Häggman, H.; Jaakola, L. On the developmental and environmental regulation of secondary metabolism in Vaccinium spp. berries. Front Plant Sci. 2016, 7, 655. [Google Scholar] [CrossRef] [Green Version]
  31. Kumar, R.; Khurana, A.; Sharma, A.K. Role of plant hormones and their interplay in development and ripening of fleshy fruits. J. Exp. Bot. 2014, 65, 4561–4575. [Google Scholar] [CrossRef] [Green Version]
  32. Osorio, S.; Scossa, F.; Fernie, A.R. Molecular regulation of fruit ripening. Front. Plant Sci. 2013, 4, 198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bapat, V.A.; Trivedi, P.K.; Ghosh, A.; Sane, V.A.; Ganapathi, T.R.; Nath, P. Ripening of fleshy fruit: Molecular insight and the role of ethylene. Biotechnol. Adv. 2010, 28, 94–107. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, Q.; Pan, H.; Li, J. Molecular insights into potential contributions of natural polyphenols to lung cancer treatment. Cancers 2019, 11, 1565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. McGhie, T.K.; Walton, M.C. The bioavailability and absorption of anthocyanins: Towards a better understanding. Mol. Nutr. Food Res. 2007, 51, 702–713. [Google Scholar] [CrossRef] [PubMed]
  36. Kuntz, S.; Rudloff, S.; Asseburg, H.; Borsch, C.; Fröhling, B.; Unger, F.; Dold, S.; Spengler, B.; Römpp, A.; Kunz, C. Uptake and bioavailability of anthocyanins and phenolic acids from grape/blueberry juice and smoothie in vitro and in vivo. Br. J. Nutr. 2015, 113, 1044–1055. [Google Scholar] [CrossRef] [Green Version]
  37. Tena, N.; Martín, J.; Asuero, A.G. State of the art of anthocyanins: Antioxidant activity, sources, bioavailability, and therapeutic effect in human health. Antioxidants 2020, 9, 451. [Google Scholar] [CrossRef]
  38. Liu, Y.; Zhang, D.; Wu, Y.; Wang, D.; Wei, Y.; Wu, J.; Ji, B. Stability and absorption of anthocyanins from blueberries subjected to a simulated digestion process. Int. J. Food Sci. Nutr. 2014, 65, 440–448. [Google Scholar] [CrossRef]
  39. Zhang, L.; Wang, Y.; Li, D.; Ho, C.-T.; Li, J.; Wan, X. The absorption, distribution, metabolism and excretion of procyanidins. Food Funct. 2016, 7, 1273–1281. [Google Scholar] [CrossRef]
  40. Ferguson, P.J.; Kurowska, E.; Freeman, D.J.; Chambers, A.F.; Koropatnick, D.J. A flavonoid fraction from cranberry extract inhibits proliferation of human tumor cell lines. J. Nutr. 2004, 134, 1529–1535. [Google Scholar] [CrossRef] [Green Version]
  41. Neto, C.C.; Krueger, C.G.; Lamoureaux, T.L.; Kondo, M.; Vaisberg, A.J.; Hurta, R.A.R.; Curtis, S.; Matchett, M.D.; Yeung, H.; Sweeney, M.I.; et al. Maldi-TOF MS characterization of proanthocyanidins from cranberry fruit (Vaccinium macrocarpon) that inhibit tumor cell growth and matrix metalloproteinase expression in vitro. J. Sci. Food. Agric. 2006, 86, 18–25. [Google Scholar] [CrossRef]
  42. Kresty, L.A.; Howell, A.B.; Baird, M. Cranberry proanthocyanidins mediate growth arrest of lung cancer cells through modulation of gene expression and rapid induction of apoptosis. Molecules 2011, 16, 2375–2390. [Google Scholar] [CrossRef]
  43. Zhai, W.-Y.; Jia, C.-P.; Zhao, H.; Xu, Y.-S. Procyanidins inhibit tumor angiogenesis by crosslinking extracellular matrix. Chin. J Cancer Res. 2011, 23, 99–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Akhtar, S.; Meeran, S.M.; Katiyar, N.; Katiyar, S.K. Grape seed proanthocyanidins inhibit the growth of human non-small cell lung cancer xenografts by targeting insulin-like growth factor binding protein-3, tumor cell proliferation, and angiogenic factors. Clin. Cancer Res. 2009, 15, 821–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sharma, S.D.; Meeran, S.M.; Katiyar, S.K. Proanthocyanidins inhibit in vitro and in vivo growth of human non-small cell lung cancer cells by inhibiting the prostaglandin E(2) and prostaglandin E(2) receptors. Mol. Cancer Ther. 2010, 9, 569–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Punathil, T.; Katiyar, S.K. Inhibition of non-small cell lung cancer cell migration by grape seed proanthocyanidins is mediated through the inhibition of nitric oxide, guanylate cyclase, and ERK1/2. Mol. Carcinog. 2009, 48, 232–242. [Google Scholar] [CrossRef] [PubMed]
  47. Mao, J.T.; Smoake, J.; Park, H.K.; Lu, Q.-Y.; Xue, B. Grape seed procyanidin extract mediates antineoplastic effects against lung cancer via modulations of prostacyclin and 15-HETE eicosanoid pathways. Cancer Prev. Res. 2016, 9, 925–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Vaid, M.; Katiyar, S.K. Grape seed proanthocyanidins inhibit cigarette smoke condensate-induced lung cancer cell migration through inhibition of NADPH oxidase and reduction in the binding of p22(phox) and p47(phox) proteins. Mol. Carcinog. 2015, 54, E61–E71. [Google Scholar] [CrossRef] [PubMed]
  49. Singh, T.; Sharma, S.D.; Katiyar, S.K. Grape proanthocyanidins induce apoptosis by loss of mitochondrial membrane potential of human non-small cell lung cancer cells in vitro and in vivo. PLoS ONE 2011, 6, e27444. [Google Scholar] [CrossRef]
  50. Xue, B.; Lu, Q.-Y.; Massie, L.; Qualls, C.; Mao, J.T. Grape seed procyanidin extract against lung cancer: The role of microrna-106b, bioavailability, and bioactivity. Oncotarget 2018, 9, 15579–15590. [Google Scholar] [CrossRef] [Green Version]
  51. Ohnuma, T.; Anzai, E.; Suzuki, Y.; Shimoda, M.; Saito, S.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Selective antagonization of activated Nrf2 and inhibition of cancer cell proliferation by procyanidins from Cinnamomi Cortex extract. Arch. Biochem. Biophys. 2015, 585, 17–24. [Google Scholar] [CrossRef]
  52. Ohnuma, T.; Sakamoto, K.; Shinoda, A.; Takagi, C.; Ohno, S.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Procyanidins from Cinnamomi Cortex promote proteasome-independent degradation of nuclear Nrf2 through phosphorylation of insulin-like growth factor-1 receptor in A549 cells. Arch. Biochem. Biophys. 2017, 635, 66–73. [Google Scholar] [CrossRef] [PubMed]
  53. Kin, R.; Kato, S.; Kaneto, N.; Sakurai, H.; Hayakawa, Y.; Li, F.; Tanaka, K.; Saiki, I.; Yokoyama, S. Procyanidin C1 from Cinnamomi Cortex inhibits TGF-β-induced epithelial-to-mesenchymal transition in the A549 lung cancer cell line. Int. J. Oncol. 2013, 43, 1901–1906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Sun, C.; Jin, W.; Shi, H. Oligomeric proanthocyanidins protects A549 cells against H2O2-induced oxidative stress via the Nrf2-ARE pathway. Int. J. Mol. Med. 2017, 39, 1548–1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wu, Y.; Liu, C.; Niu, Y.; Xia, J.; Fan, L.; Wu, Y.; Gao, W. Procyanidins mediates antineoplastic effects against non-small cell lung cancer via the JAK2/STAT3 pathway. Transl. Cancer Res. 2021, 10, 2023–2035. [Google Scholar] [CrossRef]
  56. Kuo, P.L.; Hsu, Y.L.; Lin, T.C.; Lin, C.C. The antiproliferative activity of prodelphinidin B-2 3’-O-gallate from green tea leaf is through cell cycle arrest and Fas-mediated apoptotic pathway in A549 cells. Food Chem. Toxicol. 2005, 43, 315–323. [Google Scholar] [CrossRef]
  57. Kuo, P.L.; Hsu, Y.L.; Lin, T.A.; Lin, C.C. Prodelphinidin B-2 3,3’-di-O-gallate from Myrica rubra inhibits proliferation of A549 carcinoma cells via blocking cell cycle progression and inducing apoptosis. Eur. J. Pharmacol. 2004, 501, 41–48. [Google Scholar] [CrossRef]
  58. Way, T.-D.; Tsai, S.-J.; Wang, C.-M.; Jhan, Y.-L.; Ho, C.-T.; Chou, C.-H. Cinnamtannin D1 from Rhododendron formosanum induces autophagy via the inhibition of Akt/mTOR and activation of ERK1/2 in non-small-cell lung carcinoma cells. J. Agric. Food Chem. 2015, 63, 10407–10417. [Google Scholar] [CrossRef]
  59. Oak, M.-H.; Bedoui, J.E.; Madeira, S.V.F.; Chalupsky, K.; Schini-Kerth, V.B. Delphinidin and cyanidin inhibit PDGF(AB)-induced VEGF release in vascular smooth muscle cells by preventing activation of p38 MAPK and JNK. Br. J. Pharmacol. 2006, 149, 283–290. [Google Scholar] [CrossRef] [Green Version]
  60. Lu, J.N.; Panchanathan, R.; Lee, W.S.; Kim, H.J.; Kim, D.H.; Choi, Y.H.; Kim, G.; Shin, S.C.; Hong, S.C. Anthocyanins from the fruit of Vitis coignetiae Pulliatinhibit TNF-augmented cancer proliferation, migration, and invasion in A549 cells. Asian Pac. J. Cancer Prev. 2017, 18, 2919–2923. [Google Scholar]
  61. Chen, P.-N.; Chu, S.-C.; Chiou, H.-L.; Kuo, W.-H.; Chiang, C.-L.; Hsieh, Y.-S. Mulberry anthocyanins, cyanidin 3-rutinoside and cyanidin 3-glucoside, exhibited an inhibitory effect on the migration and invasion of a human lung cancer cell line. Cancer Lett. 2006, 235, 248–259. [Google Scholar] [CrossRef]
  62. Chen, X.; Zhang, W.; Xu, X. Cyanidin-3-glucoside suppresses the progression of lung adenocarcinoma by downregulating TP53I3 and inhibiting PI3K/AKT/mTOR pathway. World J. Surg. Oncol. 2021, 19, 232. [Google Scholar] [CrossRef]
  63. Pal, H.C.; Sharma, S.; Strickland, L.R.; Agarwal, J.; Athar, M.; Elmets, C.A.; Afaq, F. Delphinidin reduces cell proliferation and induces apoptosis of non-small-cell lung cancer cells by targeting EGFR/VEGFR2 signaling pathways. PLoS ONE. 2013, 8, e77270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kim, M.-H.; Jeong, Y.-J.; Cho, H.-J.; Hoe, H.-S.; Park, K.-K.; Park, Y.-Y.; Choi, Y.-H.; Kim, C.-H.; Chang, H.-W.; Park, Y.-J.; et al. Delphinidin inhibits angiogenesis through the suppression of HIF-1α and VEGF expression in A549 lung cancer cells. Oncol. Rep. 2017, 37, 777–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kausar, H.; Jeyabalan, J.; Aqil, F.; Chabba, D.; Sidana, J.; Singh, I.P.; Gupta, R.C. Berry anthocyanidins synergistically suppress growth and invasive potential of human non-small-cell lung cancer cells. Cancer Lett. 2012, 325, 54–62. [Google Scholar] [CrossRef] [PubMed]
  66. Aqil, F.; Gupta, A.; Munagala, R.; Jeyabalan, J.; Kausar, H.; Sharma, R.J.; Singh, I.P.; Gupta, R.C. Antioxidant and antiproliferative activities of anthocyanin/ellagitannin-enriched extracts from Syzygium cumini L. (Jamun, the Indian Blackberry). Nutr. Cancer 2012, 64, 428–438. [Google Scholar] [CrossRef] [Green Version]
  67. Ohnuma, T.; Matsumoto, T.; Itoi, A.; Kawana, A.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Enhanced sensitivity of A549 cells to the cytotoxic action of anticancer drugs via suppression of Nrf2 by procyanidins from Cinnamomi Cortex extract. Biochem. Biophys. Res. Commun. 2011, 413, 623–629. [Google Scholar] [CrossRef]
  68. Xu, Y.; Huang, Y.; Chen, Y.; Cao, K.; Liu, Z.; Wan, Z.; Liao, Z.; Li, B.; Cui, J.; Yang, Y.; et al. Grape seed proanthocyanidins play the roles of radioprotection on normal lung and radiosensitization on lung cancer via differential regulation of the MAPK signaling pathway. J. Cancer 2021, 12, 2844–2854. [Google Scholar] [CrossRef] [PubMed]
  69. Kang, S.H.; Bak, D.-H.; Chung, B.Y.; Bai, H.-W.; Kang, B.S. Delphinidin enhances radio-therapeutic effects via autophagy induction and JNK/MAPK pathway activation in non-small cell lung cancer. Korean J. Physiol. Pharmacol. 2020, 24, 413–422. [Google Scholar] [CrossRef]
  70. Eguchi, H.; Matsunaga, H.; Onuma, S.; Yoshino, Y.; Matsunaga, T.; Ikari, A. Down-regulation of claudin-2 expression by cyanidin-3-glucoside enhances sensitivity to anticancer drugs in the spheroid of human lung adenocarcinoma A549 cells. Int. J. Mol. Sci. 2021, 22, 499. [Google Scholar] [CrossRef]
  71. Liao, W.; Chen, L.; Ma, X.; Jiao, R.; Li, X.; Wang, Y. Protective effects of kaempferol against reactive oxygen species-induced hemolysis and its antiproliferative activity on human cancer cells. Eur. J. Med. Chem. 2016, 114, 24–32. [Google Scholar] [CrossRef]
  72. Cheng, W.; Chen, K.; Lee, K.; Feng, P.; Wu, S. Nicotinic-nAChR signaling mediates drug resistance in lung cancer. J. Cancer 2020, 11, 1125–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Bordas, A.; Cedillo, J.L.; Arnalich, F.; Esteban-Rodriguez, I.; Guerra-Pastrián, L.; de Castro, J.; Martín-Sánchez, C.; Atienza, G.; Fernández-Capitan, C.; Rios, J.J.; et al. Expression patterns for nicotinic acetylcholine receptor subunit genes in smoking-related lung cancers. Oncotarget 2017, 8, 67878–67890. [Google Scholar] [CrossRef] [Green Version]
  74. Schuller, H.M. Nitrosamines as nicotinic receptor ligands. Life Sci. 2007, 80, 2274–2280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hirata, N.; Yamada, S.; Sekino, Y.; Kanda, Y. Tobacco nitrosamine NNK increases ALDH-positive cells via ROS-Wnt signaling pathway in A549 human lung cancer cells. J. Toxicol. Sci. 2017, 42, 193–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wen, J.; Fu, J.-H.; Zhang, W.; Guo, M. Lung carcinoma signaling pathways activated by smoking. Chin. J. Cancer. 2011, 30, 551–558. [Google Scholar] [CrossRef] [Green Version]
  77. Martinez-Marti, A.; Navarro, A.; Felip, E. COX-2 inhibitors in NSCLC: Never-ending story or misplaced? Transl. Lung Cancer Res. 2018, 7 (Suppl. S3), S191–S194. [Google Scholar] [CrossRef]
  78. Huang, R.-Y.; Li, M.-Y.; Hsin, M.K.Y.; Underwood, M.J.; Ma, L.T.; Mok, T.S.K.; Warner, T.D.; Chen, G.G. 4-Methylnitrosamino-1-3-pyridyl-1-butanone (NNK) promotes lung cancer cell survival by stimulating thromboxane A2 and its receptor. Oncogene 2011, 30, 106–116. [Google Scholar] [CrossRef] [Green Version]
  79. Al-Wadei, H.A.N.; Al-Wadei, M.H.; Schuller, H.M. Cooperative regulation of non-small cell lung carcinoma by nicotinic and beta-adrenergic receptors: A novel target for intervention. PLoS ONE 2012, 7, e29915. [Google Scholar] [CrossRef] [Green Version]
  80. Czyżykowski, R.; Połowinczak-Przybyłek, J.; Potemski, P. Nicotine-induced resistance of non-small cell lung cancer to treatment-possible mechanisms. Adv. Hyg. Exp. Med./Postepy Hig. I Med. Dosw. 2016, 70, 186–193. [Google Scholar] [CrossRef]
  81. Zhang, Q.; Tang, X.; Zhang, Z.-F.; Velikina, R.; Shi, S.; Le, A.D. Nicotine induces hypoxia-inducible factor-1alpha expression in human lung cancer cells via nicotinic acetylcholine receptor-mediated signaling pathways. Clin. Cancer Res. 2007, 13, 4686–4694. [Google Scholar] [CrossRef] [Green Version]
  82. Dasgupta, P.; Rastogi, S.; Pillai, S.; Ordonez-Ercan, D.; Morris, M.; Haura, E.; Chellappan, S. Nicotine induces cell proliferation by β-arrestin–mediated activation of Src and Rb–Raf-1 pathways. J. Clin. Investig. 2006, 116, 2208–2217. [Google Scholar] [CrossRef] [PubMed]
  83. Dasgupta, P.; Rizwani, W.; Pillai, S.; Davis, R.; Banerjee, S.; Hug, K.; Lloyd, M.; Coppola, D.; Haura, E.; Chellappan, S.P. ARRB1-mediated regulation of E2F target genes in nicotine-induced growth of lung tumors. J. Natl. Cancer Inst. 2011, 103, 317–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tsurutani, J.; Castillo, S.S.; Brognard, J.; Granville, G.A.; Zhang, C.; Gills, J.J.; Sayyah, J.; Dennis, P.A. Tobacco components stimulate Akt-dependent proliferation and NFkappaB-dependent survival in lung cancer cells. Carcinogenesis 2005, 26, 1182–1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Schaal, C.; Chellappan, S. Nicotine-mediated regulation of nicotinic acetylcholine receptors in non-small cell lung adenocarcinoma by E2F1 and STAT1 transcription factors. PLoS ONE 2016, 11, e0156451. [Google Scholar] [CrossRef] [PubMed]
  86. Deng, X. Bcl2 family functions as signaling target in nicotine-/NNK-induced survival of human lung cancer cells. Scientifica 2014, 2014, 215426. [Google Scholar] [CrossRef] [Green Version]
  87. Memmott, R.M.; Dennis, P.A. The role of the Akt/mTOR pathway in tobacco-carcinogen induced lung tumorigenesis. Clin. Cancer Res. 2010, 16, 4–10. [Google Scholar] [CrossRef] [Green Version]
  88. Kalscheuer, S.; Zhang, X.; Zeng, Y.; Upadhyaya, P. Differential expression of microRNAs in early-stage neoplastic transformation in the lungs of F344 rats chronically treated with the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Carcinogenesis 2008, 29, 2394–2399. [Google Scholar] [CrossRef]
  89. Doukas, S.G.; Vageli, D.P.; Lazopoulos, G.; Spandidos, D.A.; Sasaki, C.T.; Tsatsakis, A. The effect of NNK, a tobacco smoke carcinogen, on the miRNA and mismatch DNA repair expression profiles in lung and head and neck squamous cancer cells. Cells 2020, 9, 1031. [Google Scholar] [CrossRef] [Green Version]
  90. Jin, H.; Li, Q.; Cao, F.; Wang, S.-N.; Wang, R.-T.; Wang, Y.; Tan, Q.-Y.; Li, C.-R.; Zou, H.; Wang, D.; et al. miR-124 inhibits lung tumorigenesis induced by K-ras mutation and NNK. Mol. Ther. Nucleic Acids. 2017, 9, 145–154. [Google Scholar] [CrossRef] [Green Version]
  91. Xie, H.; Wang, J.-R.; Yau, L.-F.; Liu, Y.; Liu, L.; Han, Q.-B.; Zhao, Z.; Jiang, Z.-H. Catechins and procyanidins of Ginkgo biloba show potent activities towards the inhibition of β-amyloid peptide aggregation and destabilization of preformed fibrils. Molecules 2014, 19, 5119–5134. [Google Scholar] [CrossRef] [Green Version]
  92. Jin, W.; Bu, X.; Liu, Y.; Shen, L.; Zhuang, Z.; Jiao, S.; Zhu, C.; Wang, Q.; Zhou, H.; Zhang, T.; et al. Plasma amyloid-beta levels in patients with different types of cancer. Neurotox. Res. 2017, 31, 283–288. [Google Scholar] [CrossRef] [PubMed]
  93. 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] [PubMed]
  94. Atali, S.; Dorandish, S.; Devos, J.; Williams, A.; Price, D.; Taylor, J.; Guthrie, J.; Heyl, D.; Evans, H.G. Interaction of amyloid beta with humanin and acetylcholinesterase is modulated by ATP. FEBS Open Biol. 2020, 10, 2805–2823. [Google Scholar] [CrossRef] [PubMed]
  95. Niu, X.-M.; Lu, S. Acetylcholine receptor pathway in lung cancer: New twists to an old story. World J. Clin. Oncol. 2014, 5, 667–676. [Google Scholar] [CrossRef] [PubMed]
  96. Friedman, J.R.; Richbart, S.D.; Merritt, J.C.; Brown, K.C.; Nolan, N.A.; Akers, A.T.; Lau, J.K.; Robateau, Z.R.; Miles, S.L.; Dasgupta, P. Acetylcholine signaling system in progression of lung cancers. Pharmacol. Ther. 2019, 194, 222–254. [Google Scholar] [CrossRef]
  97. Xi, H.-J.; Wu, R.-P.; Liu, J.-J.; Zhang, L.-J.; Li, Z.-S. Role of acetylcholinesterase in lung cancer. Thorac. Cancer. 2015, 6, 390–398. [Google Scholar] [CrossRef]
  98. Pinheiro, N.M.; Miranda, C.J.C.P.; Perini, A.; Câmara, N.O.S.; Costa, S.K.P.; Alonso-Vale, M.I.C.; Caperuto, L.C.; Tibério, I.L.F.C.; Prado, M.A.M.; Martinsm, M.A.; et al. Pulmonary inflammation is regulated by the levels of the vesicular acetylcholine transporter. PLoS ONE. 2015, 10, e0120441. [Google Scholar] [CrossRef] [Green Version]
  99. Huang, Q.; Du, J.; Fan, J.; Lv, Z.; Qian, X.; Zhang, X.; Han, J.; Chen, C.; Wu, F.; Jin, Y. The effect of proinflammatory cytokines on IL-17RA expression in NSCLC. Med. Oncol. 2014, 31, 144. [Google Scholar] [CrossRef]
  100. Mohrherr, J.; Uras, I.Z.; Moll, H.P.; Casanova, E. STAT3: Versatile functions in non-small cell lung cancer. Cancers 2020, 12, 1107. [Google Scholar] [CrossRef]
  101. Parakh, S.; Ernst, M.; Poh, A.R. Multicellular effects of STAT3 in non-small cell lung cancer: Mechanistic insights and therapeutic opportunities. Cancers 2021, 13, 6228. [Google Scholar] [CrossRef]
  102. Jiang, M.; Zhang, P.; Hu, G.; Xiao, Z.; Xu, F.; Zhong, T.; Huang, F.; Kuang, H.; Zhang, W. Relative expressions of miR-205-5p, miR-205-3p, and miR-21 in tissues and serum of non-small cell lung cancer patients. Mol. Cell Biochem. 2013, 383, 67–75. [Google Scholar] [CrossRef]
  103. Sheng, W.; Guo, W.; Lu, F.; Liu, H.; Xia, R.; Dong, F. Upregulation of Linc00284 promotes lung cancer progression by regulating the miR-205-3p/c-met axis. Front. Genet. 2021, 12, 694571. [Google Scholar] [CrossRef] [PubMed]
  104. Xu, L.; Xiong, J.; Zhang, Y.; Dai, Y.; Ren, X.; Ren, Y.; Han, D.; Wei, S.; Qi, M. miR-205-3p promotes lung cancer progression by targeting APBB2. Mol. Med. Rep. 2021, 24, 588. [Google Scholar] [CrossRef] [PubMed]
  105. Lim, T.; Jung, H.; Hwang, K.T. Bioconversion of cyanidin-3-rutinoside to cyanidin-3-glucoside in black raspberry by crude α-L-rhamnosidase from aspergillus species. J. Microbiol. Biotechnol. 2015, 25, 1842–1848. [Google Scholar] [CrossRef] [PubMed]
  106. Zhou, X.-J.; Zhu, C.-T.; Zhang, L.-Y.; You, S.; Wu, F.-A.; Wang, J. Enrichment and purification of red pigments from defective mulberry fruits using biotransformation in a liquid-liquid-solid three-phase system. Environ. Sci. Pollut. Res. Int. 2021, 28, 24432–24440. [Google Scholar] [CrossRef]
Figure 1. Mechanisms for nicotine in the development of NSCLC.
Figure 1. Mechanisms for nicotine in the development of NSCLC.
Ijms 23 07905 g001
Figure 2. The therapeutic potential of PCs and ACNs in nicotine-induced NSCLC. (↓) decrease, (↑) increase.
Figure 2. The therapeutic potential of PCs and ACNs in nicotine-induced NSCLC. (↓) decrease, (↑) increase.
Ijms 23 07905 g002
Table 1. The molecular mechanisms of PCs in nicotine-induced NSCLC treatment.
Table 1. The molecular mechanisms of PCs in nicotine-induced NSCLC treatment.
Study TypeNSCLC Cell TypeExtract/CompoundConcentrationsActivityMechanisms of ActionReference
In vitroDMS114Cranberry presscake200–300 µmol/LAnti-proliferative, apoptosisNA[40]
In vitroH460Cranberry (Vaccinium macrocarpon)20–80 µg/mLAnti-proliferativeMMP2, MMP9↓[41]
In vitroH460Cranberry (Vaccinium macrocarpon)50 µg/mLApoptosis, cell cycle arrestP21, P73, PARP, cytochrome c, caspase3/4/8↑
Bcl-2↓
[42]
In vitro/vivoA549Grape seed100 µg/mL (in vitro)
30 mg PC/kg bodyweight (in vivo)
Inhibition of tumor angiogenesisMMP2↓[43]
In vitro/vivoA549, H1299Grape seed60, 80 µg/mL (in vitro)
Administration of PCs (0.1%, 0.2%, and 0.5%, bodyweight) (in vivo)
Anti-proliferative, anti-angiogenic, apoptosisPCNA↓
IGFBP-3↑
[44]
In vitro/vivoH157, H226, H460, H1299, A549Grape seed20, 40, and 60 µg/mL (in vitro)
Administration of PCs (0.5%, bodyweight) (in vivo)
Anti-proliferative, apoptosisCOX-2, PGI-2↓[45]
In vitroA549, H1299Grape seed10, 20, 40, and 60 µg/mLAnti-migrationNO, L-NAME, MAPK, ERK1/2↓[46]
In vitro A549Grape seed6 µg/mLAnti-proliferative, apoptosiscaspase 3, PTGIS/PGI2↑[47]
In vitroA549, H1299, H460Grape seed20 and 40 µg/mLAnti-migrationE-cadherin, NOX, p22/p47(phox)↓
N-cadherin, fibronectin, vimentin↑
[48]
In vitro/vivoA549, H1299Grape seed20, 40, and 60 µg/mL (in vitro)
50, 100, and
200 mg PC/kg bodyweight (in vivo)
ApoptosisG1arrest, Bax, caspases-3/9, Cdki, PARP↑
Bcl-2,Bcl-xl, Cdk2/4/6, cyclins↓
[49]
In vitro/vivoA549Grape seed45 µg/mL (in vitro)
112 mg PC/kg bodyweight (in vivo)
Anti-proliferative, anti-invasiveCDKN1A, p21↑,
miR-106b↓
[50]
In vitroA549Cinnamomi Cortex2.5 µg/mLInhibition of cell viability and proliferationNrf2↓[51]
In vitroA549Cinnamomi Cortex10 µg/mLInhibition of cell proliferationNrf2, IGF-1R↓[52]
In vitroA549Cinnamomi Cortex12.5, 25, 50, and 100 µg/mLAnti-metastaticTGF-β, snail, E-cadherin, smad2↓[53]
In vitroA549PCs≥100 mg/LInhibition of cell viabilityROS, MDA, Nrf2↓
HO-1, NQO1, TXNRD1, glutathione, catalase, superoxide dismutase↑
[54]
In vitroA549PCs12.5, 25, 50, 100, and 200 µMInhibition of cell viability and proliferation, apoptosis, cell cycle arrestN-cadherin, vimentin, Bcl-2, MMP2/9, JAK2/STAT3↓,
Bax↑
[55]
In vitroA549Green tea leaf1, 5, 10, 20 µMAnti-proliferative, apoptosis, cell cycle arrestP21, P53, Fas/sFasL, Fas/APO-1↑[56]
In vitroA549Myrica rubra0.5, 2.5, 5, and 10 µMAnti-proliferative, apoptosis, cell cycle arrestFas/APO-1, P21/WAF1, P53, Fas/sFasL↑[57]
In vitroA549, H460Rhododendron formosanum125, 150, and 175 µMAutophagyAkt/mTOR↓[58]
(↑) increase; (↓) decrease; NA: not mentioned.
Table 2. The molecular mechanisms of ACNs in nicotine-induced NSCLC treatment.
Table 2. The molecular mechanisms of ACNs in nicotine-induced NSCLC treatment.
Study TypeNSCLC Cell TypeExtract/CompoundConcentrationsActivityMechanisms of ActionReference
In vitroA549Vitis coignetiae Pulliat200 µg/mLAnti-proliferative, anti-invasive, anti-angiogenic, anti-migrationMMP2/9, cyclin D1, C-myc, COX-2, VEGF↓[60]
In vitroA549Morus alba L.25, 50, and 100 µMAnti-migration, anti-invasiveMMP2, c-Jun, C-fos, NF-kB↓[61]
In vitroA549, H1299Cyanidin-3-glucoside5, 10, 20, 40, and 80 µMAnti-proliferative, anti-migration, anti-invasive, apoptosisTP53I3 andPI3K/Akt/mTOR↓[62]
In vitro/vivoA549, H441, SK-MES-1Delphinidin5–100 µM (in vitro)
1, 2 mg PC/kg bodyweight (in vivo)
Inhibition of tumor growth, anti-proliferative, anti-angiogenic, apoptosisEGFR, Bcl-2, PCNA, cyclin D1, VEGFA, Akt/PI3K/MAPK↓
Bax, caspase-3/9↑
[63]
In vitro/vivoA549Delphinidin10, 20, and 40 µM (in vitro)
80 µM (in vivo)
Anti-angiogenicEGF, CoCl2, HIF-1α, ERK, VEGF mRNA, Akt/mTOR/PI3K/p70S6K↓[64]
In vitro/vivoA549, H1299Bilberry and blueberry3.125–12.5 µM (in vitro)
1.5 mg PC/kg bodyweight (in vivo)
Anti-metastatic, anti-invasive, apoptosis, cell cyclearrestTNFα-induced NF-kB, Notch, Wnt/β-catenin, cyclinD1/B1, VEGF, p-ERK, bcl-2, MMP9 ↓[65]
In vitroA549Syzygium cumini L.2.5, 5, 10, 20, and 25 µMAnti-proliferativeNA[66]
(↑) increase, (↓) decrease, NA; not mentioned.
Table 3. The molecular mechanisms of PCs and ACNs in combination with radiotherapy/chemotherapy in nicotine-induced NSCLC treatment.
Table 3. The molecular mechanisms of PCs and ACNs in combination with radiotherapy/chemotherapy in nicotine-induced NSCLC treatment.
Study TypeNSCLC Cell TypeExtract/CompoundConcentrationsActivityMechanisms of ActionReference
In vitroA549Cinnamomi Cortex100–300 µg/mLAnti-proliferativeNrf2↓[67]
In vitro/In vivoA549Grape seed20 µg/mL (in vitro)
2 mg PC/kg bodyweight (in vivo)
Anti-inflammation, apoptosis ROS, IL-6, IFN-γ↓
P53, p-JNK, MAPK↑
[68]
In vitroA549Delphinidin10, 20, and 50 µMApoptosis, autophagy p-ERK1/2↓,
caspase-3, LC3, p-JNK, MAPK ↑
[69]
In vitroA549Cyanidin-3-glucoside1, 10, 20, and 50 µMApoptosis, autophagy CLDN2, p-Akt↓,
P38 MAPK↑
[70]
(↑) increase; (↓) decrease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23147905

AMA Style

Alsharairi NA. Insights into the Mechanisms of Action of Proanthocyanidins and Anthocyanins in the Treatment of Nicotine-Induced Non-Small Cell Lung Cancer. International Journal of Molecular Sciences. 2022; 23(14):7905. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23147905

Chicago/Turabian Style

Alsharairi, Naser A. 2022. "Insights into the Mechanisms of Action of Proanthocyanidins and Anthocyanins in the Treatment of Nicotine-Induced Non-Small Cell Lung Cancer" International Journal of Molecular Sciences 23, no. 14: 7905. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23147905

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