Abstract
Notch-1 signaling plays a crucial role in stem cell maintenance and in repair mechanisms in various mucosal surfaces, including airway mucosa. Persistent injury can induce an aberrant activation of Notch-1 signaling in stem cells leading to an increased risk of cancer initiation and progression. Chronic inflammatory respiratory disorders, including chronic obstructive pulmonary disease (COPD) is associated with both overactivation of Notch-1 signaling and increased lung cancer risk. Increased oxidative stress, also due to cigarette smoke, can further contribute to promote cancer initiation and progression by amplifying inflammatory responses, by activating the Notch-1 signaling, and by blocking regulatory mechanisms that inhibit the growth capacity of stem cells. This review offers a comprehensive overview of the effects of aberrant Notch-1 signaling activation in stem cells and of increased oxidative stress in lung cancer. The putative role of natural compounds with antioxidant properties is also described.
Keywords: stem cells, Notch signaling, oxidative stress, cigarette smoke, lung cancer, nutraceutics
Notch-1 signaling activation preserves stem cells and cancer stem cells. Oxidative stress is the result of an imbalance between pro and antioxidant mechanisms. Cigarette smoke increases oxidative stress and affects Notch-1 signaling activation.
Graphical Abstract
Graphical Abstract.
1. Introduction
Tissue stem/progenitor cells are crucial in the homeostasis and tissue repair of various mucosal surfaces following injury, including the airway mucosa [1]. There are several subgroups of stem cells (embryonic stem cells and somatic or adult non-embryonic stem cells) identified by different cellular markers that correlate with various cellular events. Continued activation of repair processes to restore damage due to a persistent inflammatory insult can lead to the transformation of stem cells into cancer stem cells (CSCs) [2]. Responses to stimuli from the surrounding microenvironment, activation of various signaling pathways, epigenetic modifications, and self-renewal processes activate dedifferentiation processes can further contribute to generate undifferentiated cancer cell progenitors.
The Notch pathway plays an important role in many processes that preserve stem cells because it regulates mechanisms involved in angiogenesis, self-renewal, adhesion, and differentiation [1]. These physiological functions of Notch signaling explain why it is one of the most activated pathways in cancer cells participating in all cancer processes from initiation to cancer metastasis [2].
Increased oxidative stress can also contribute to cancer initiation and progression [3]. In this regard, it has been demonstrated that oxidative stress activates several signaling pathways that control processes of self-renewal ability of stem or CSCs. During chronic inflammatory processes, increased reactive oxygen species (ROS) and reactive nitrogen species (RNS), such as peroxynitrites and nitrogen oxides, can generate mutagenic DNA adducts and can fuel processes favoring cancer progression including drug resistance and angiogenesis [3].
The present review illustrates the role of interaction among stem cells, Notch-1, and oxidative stress in the onset and progression of cancer lesions of the airways focusing attention on the associated chronic inflammatory diseases and on the role played by cigarette smoke exposure. The role of natural compounds in preventing cancer development and progression by modulating stem cells, Notch-1, and oxidative stress interactions is also discussed.
2. Stem cells, stem cell niches, and CSCs
Stem cells are classified into mesenchymal stem cells, hematopoietic stem cells, pluripotent or multipotent stem cells, and tissue-specific stem cells. Stem cells maintain tissue homeostasis thanks to their proliferative capacity and their ability to become specialized cell populations. Stem cells and progenitor cells differ in their self-renewal capacity. In this regard, it has been demonstrated that stem cell undergoes asymmetric cell division that leads to the generation of a stem cell with perpetual self-renewal capacity and a progenitor cell that is committed to differentiation and proliferation without self-renewal capacity [4]. Two main kinds of stem cells are recognized in humans: embryonic stem cells and adult stem cells (ASCs). In tissues or organs, ASCs are undifferentiated cells that thanks to their ability of self-renewal and of multipotency can differentiate into all of the major specialized cell types of a tissue or organ. These cells, frequently in a resting state, enter the cell cycle sensing stimuli from their niche when necessary, as during wound healing upon tissue injury. In this regard, cyclin-dependent kinase inhibitors, p53 and p21 axis, epigenetic alterations including DNA methylation and histone alterations, and the Notch signaling pathway can all be able to modify the quiescent state of ASCs promoting the access to a proliferative and metabolic active state [5].
The loss of stem cell number and activity over time can explain organ dysfunction with age. Reduced proliferation rate, morphological, and biochemical changes of tissue stem cells depict a senescent state of these cells. The senescence processes can also be triggered by chronic insult exposure [6]. It is now more evident that alterations in stem cell niches in chronic degenerative diseases are responsible for stem cell aging due to the presence of suppressive stimuli that overcome activation stimuli. Stem cell niches include fibroblasts, endothelial, and immune cells that, releasing multiple factors, affect the stem cell niche microenvironment. Increased expression of signals such as Nox4 in stem cell niches impairs alveolar type 2 (AT2) to alveolar type 1 (AT1) differentiation through elevated ROS levels [7]. Accordingly, antioxidants can be useful in improving regeneration processes of alveoli restoring AT2 to AT1 differentiation processes in chronic diseases with uncontrolled oxidative stress [8]. Furthermore, aberrant activation of repair processes in response to chronic insults can promote mutagenic DNA adducts in stem cells thus favoring tumor initiation and then the consequent development of CSCs [8]. CSCs can be alternately originated by the induction of dedifferentiation processes. In this regard, it is known that all differentiated cells can undergo dedifferentiation processes and, by expressing the appropriate transcription factors, can acquire a pluripotency state [9]. CSCs are a small group of cells with potential multidirectional differentiation capacity since they are capable of self-renewal and can undergo differentiation to generate the phenotypic heterogeneity observed in cancer. Many tumors have highly proliferating cells and slowly proliferating cells inside them [10]. CSCs other than promoting cancer initiation may be responsible for tumor progression, metastasis, resistance to therapy, and subsequent tumor recurrence [10]. Cancer relapse after chemotherapy or radiotherapy is initiated by CSCs since this subpopulation is highly resistant to these treatments [11, 12]. Highly drug-resistant CSC population cells use redox systems to counteract the cytotoxic activities of anticancer agents. The CSCs, in a similar fashion to non-CSCs, are encompassed by a specialized cell microenvironment that sends activating signals to these cells [13, 14]. Collectively all these data support the notion that the main difference between normal and CSCs resides in no control over the cell number by CSCs [3].
3. Notch-1 signaling activation and CSCs
The Notch pathway plays a key role in the development and maintenance of the hematopoietic cell population related to the development, proliferation, and differentiation of stem cells [1]. It is known that the Notch pathway plays a role in the different cellular pathways in the various stages of growth of hematopoietic cells, also involving hematopoietic CSCs in the mechanism of self-regeneration or differentiation of common lymphoid precursors in T or B cells [2].
3.1 Description of the Notch pathway
Four Notch receptors (Notch-1 to Notch-4) and five ligands are expressed in humans: Delta-like 1, 3, 4 (Dll1, Dll3, Dll4), and Jagged 1, 2. It is known that Notch receptors are made up of three domains: extracellular Notch domain (NECD), transmembrane Notch domain (NTM), and intracellular Notch domain (NICD) [15, 16]. Notch signaling activation starts upon interaction with a ligand that induces two proteolytic cleavages. The first trigger of cleavage is catalyzed by ADAM family metalloproteases and leads to the removal of the extracellular domain of Notch. The residual part of the Notch protein undergoes a second cleavage through Notch gamma-secretase generating the activated form of the Notch receptor, NICD. The NICD molecule migrates into the nucleus and combines with other proteins, consequently modulating its main target gene expression, Hes and Hey family [15]. Inside the nucleus, NICD forms a complex with the DNA-bound transcription factor CSL and a Mastermind family coactivator (MAML) [16].
All four Notch receptors can be activated during physiological or pathological conditions. The consequence of Notch-1 activation on cell proliferation and the cellular apoptotic mechanisms can be regulated by modulating the expression of cell cycle regulatory proteins [17]. All four Notch receptors can be activated during physiological or pathological conditions. To our knowledge, the main difference relies on the overactivation of the Notch pathway during pathological conditions. Increased oxidative stress could be considered as one of the main triggers that promotes this overactivation.
3.2 Notch, CSCs, and the induction of epithelial–mesenchymal transition processes
The activation of the Notch pathway and thus the expression of its target gene, HES1, plays a fundamental role in the maintenance of CSCs, in a framework similar to what occurs during their embryonic development [18] (Fig. 1). The Notch signaling is also involved in the induction of epithelial–mesenchymal transition (EMT) processes [19]. EMT is a highly coordinated process that, when activated, through key transcription factors such as Snail, Twist, and Zeb, leads to the loss of epithelial cell polarity together with the loss of cell–cell adhesion and the acquisition of mesenchymal cell properties [19]. Although the EMT physiologically concurs with the events finalized to tissue repair, during lung cancer processes, this process can be used by cancer cells to promote metastasis. Stemness is a property associated with EMT processes. CSCs are characterized by increased EMT transcription factors, along with reduced intercellular adhesion protein expression. Notch signaling can promote EMT processes through different mechanisms [20]. Notch signaling activation increases NF-κB thus inducing metalloproteases, TGF-beta, or Twist thus promoting EMT. In addition, Notch can induce the expression of transcription factors including Snail that increase mesenchymal markers, such as vimentin, other than reducing epithelial markers, including E-cadherin [20]. It is also well established that EMT in cancer has been depicted as a dedifferentiation event with a relevant role in the acquisition of stem cell characteristics [21].
Figure 1.
(A) Upon Notch ligand and Notch-1 receptor interaction, ADAM-family metalloproteases catalyze a first cleavage that leads to the removal of the extracellular domain of the Notch-1 receptor. (B) Gamma-secretase mediates a second Notch cleavage that generates the activated form of the Notch receptor, NICD. (C) The NICD migrates into the nucleus, where interacting with other proteins (CSL and MAML), modulates primary target genes, such as the Hes and Hey family. (D) Fully activated Notch signaling and target molecules maintain and preserve stem cell niches and promote EMT and angiogenesis processes. Pharmacological approaches to block the Notch pathway are shown: mAbs against receptor and ligand, gamma-secretase inhibitor (GSI), and Notch transcription complex inhibitor (CB-103).
3.3 Pharmacological targeting
Notch signaling could be targeted by inhibiting the signaling pathway by two major classes of Notch inhibitors that primarily focus on the clinical development of promising agents that either block Notch receptor cleavages such as secretase inhibitors (GSIs) or interfere with the Notch ligand–receptor interaction by monoclonal antibodies directed against Notch ligand or Notch receptor or inhibit the Notch transcription complex, such as CB-103 [22].
Notch signaling is an important mechanism for direct cell–cell communication involved in cell fate determination, stem cell potential, and lineage commitment. The biological function of this pathway is critically context-dependent. The finding that Notch signaling elicits a duality of signals, involved in growth/differentiation control, and cell survival supports the inhibition of this pathway as one of the new approaches for cancer therapy [23, 24]. A therapeutic strategy involving the combined use of traditional therapies and Notch inhibitors may prove more effective in removing stem cells with drug resistance thus leading to effective cancer eradication [2]. This approach of targeting the signaling pathway of CSCs, such as the Notch pathway, represents a promising future direction for the therapeutic strategy to cure cancer. The choice of the most appropriate inhibitor for each patient relies on the best patient stratification according to efficacy and pharmacodynamic biomarkers.
4. Oxidative stress role in Notch-1 signaling activation and CSCs
4.1 Mechanisms for ROS production
Oxidative stress is the result of an excessive production of ROS or RNS and defective antioxidant mechanisms [25].
ROS and RNS are free radicals, i.e. chemical species that have at least one unpaired electron in the outer part that gives them high reactivity. ROS and RNS production is triggered by multiple cellular processes in response to exogenous and endogenous stimuli. ROS have higher reactivity than RNS. The cellular sources of ROS formation are mitochondria, plasma membranes, chloroplasts, peroxisomes, and endoplasmatic reticulum stimuli [26]. In eukaryotic cells, ROS production is mainly related to the biochemical processes associated with mitochondrial cellular respiration for oxidative ATP production [27]. The main ROS derived from molecular oxygen are superoxide and hydrogen peroxide.
During cellular respiration within mitochondria molecular oxygen is reduced to water in the electron transport chain and superoxide radical is produced. Manganese superoxide dismutase then converts the superoxide radical to hydrogen peroxide. ROS production addresses the need for the response or adaptation to bioenergetic changes in the cells.
4.2 Correlation between oxidative stress, cancer, and Notch pathway
The increase of ROS in cancer cells is known to play a role in the initiation and progression of cancer. In this regard, it is important to underline that the most significant mutagens in stem cells are potent inducers of oxidative stress. Several signaling pathways enhanced by oxidative stress affect the self-renewal ability of stem or CSCs, suggesting important roles in tumorigenesis processes. The presence of ROS can produce cellular stress and damage that may produce cell death [28]. Oxidative stress can either activate cell survival or apoptosis mechanisms, depending on the severity, and duration of exposure [29, 30]. In this regard, it has been demonstrated that ROS, at low concentrations, induces both normal cell and cancer cell proliferation, as well as cell survival [31, 32] while intermediate concentrations promote cell cycle arrest and cell differentiation [33, 34]. Higher ROS concentrations damage proteins, DNA, and RNA, and result in mutations that can promote both carcinogenesis in normal cells or resistance to drugs in cancer cells [35, 36]. Although ROS can enhance cytotoxicity and apoptosis, most cancer cells can remain viable in the presence of intrinsic oxidative stress, enabling them to avoid apoptosis and become resistant to many chemotherapeutic drugs [37, 38]. Modulating ROS to overcome multidrug resistance in cancer could be an effective strategy to fight cancer growth [3]. The fluctuation of ROS levels can activate Notch signaling via activation of Nuclear factor erythroid 2-related factor 2 (Nrf2) in cells with staminal properties [39]. The increase in ROS levels activates Nrf2 that, other than promoting antioxidant responses, triggers Notch signaling activation to maintain homeostasis balance. Nrf2 silencing downregulates both Notch ligand and receptor expression [39]. Aberrant Nrf2-Notch crosstalk can synergistically cause neoplastic proliferation in lung cancer [40].
5. Stem cells in repair of the airways and initiation of lung cancers
5.1 Stem cells in airways repair
Airway epithelium, constantly exposed to environmental hazards and oxidative stress-mediated injury, can be damaged and should be repaired to maintain homeostasis. Multiple studies indicate that cells with a stem-like behavior are present throughout the airways and play a fundamental role in repair mechanisms in response to airway injury (Fig. 2). Studies of airway repair processes in the trachea have identified two cellular populations that act as stem cells in response to injury. In this district, it is possible to identify a population of tracheal multipotent basal cells that maintain bromodeoxyuridine expression and express keratin-5 (K5) [41] and a population expressing the keratin-14 (K14) marker [42]. Excessive self-renewal, if not prevented, can lead to cancer.
Figure 2.
(A) Airway epithelium, constantly exposed to oxidative stress-mediated injury, can be damaged and should be repaired to maintain homeostasis. (B) In tracheal airway epithelium, stem cells (basal cells) constantly exposed to oxidative stress-mediated injury can lead to squamous cell carcinoma. (C) In the airway epithelium of bronchioles, stem cells (club variant cells) constantly exposed to oxidative stress-mediated injury can lead to small cell carcinoma. (D) In the airway epithelium of alveoli, cells such as (Bronchiolar Club cells) are constantly exposed to oxidative stress-mediated injury can lead to adenocarcinoma.
5.2 Initiation of lung cancers
It has been established that tracheal squamous cell carcinoma originates from K5+ stem cells. This finding may be useful to promote further targeted therapy research aimed to identify and validate molecules directed against K5+ cells thus eliminating, in a selective way, only the stem cell niche of squamous carcinoma without affecting K14+ stem cells that can continue to preserve tissue homeostasis. Importantly, CSCs maintain multipotency, i.e. the ability to give rise to phenotypically different cells with different expression of markers that confer drug resistance. Cells with higher expression of anti-apoptotic markers and drug-resistant proteins survive even after chemotherapy, and this explains why despite the effect of marked reduction of tumor mass after therapy, the onset of metastasis resistant to that treatment is observed after time. The selection of chemoresistant CSCs that continue to proliferate undisturbed after therapy could explain the development of aggressive lung cancers with a high propensity to metastasize [43]. Lung tumors are classified as small-cell lung cancers (SCLC) and non-small cell lung cancers (NSCLC). Lung adenocarcinoma is the most frequent cancer among the NSCLC. Similar to other cancers, lung cancer originates from molecular alterations in airway stem cells induced by mutagenic substances. In this regard, a subtype of lung adenocarcinomas with specific molecular, pathological, and clinical phenotypes maintains the gene expression and molecular characteristics typical of basal cells (BCs) of the human airways [44]. A cluster of lung adenocarcinomas with high expression of BC signature genes has a poorer tumor grade, shorter survival, and higher metastatic potential. These lung adenocarcinomas are characterized by an increased frequency of KRAS mutations and by activation of multiple pathways regulating cell cycle, extracellular matrix organization, and differentiation processes with suppression of ciliated and exocrine bronchiolar cell-related genes [44].
Common tumors from each representative airway zone have been associated with stem cell niches differentiated by cellular expression of surface markers. The lung CSCs can be identified by markers that mainly include surface biomarkers such as CD133, CD44, and ATP-binding cassette subfamily G member 2 [45]. CD133+ cells isolated from fresh lung tumor samples can grow indefinitely in culture and this property is normally found in cell lines transformed or immortalized by viral transfections [46]. CD133+ cells can proliferate into phenotypically diverse types of tumors. Lung cancer patients with increased numbers of CD133+ cells show a worse prognosis than those with reduced numbers of this type of cells. In fact, CD133 lung CSCs exhibit increased resistance to cisplatin-based therapies and this finding should be considered for the choice of chemotherapeutic regimens in lung cancer patients [47]. With regard to the CD44 marker, it has been demonstrated that overexpression of this marker in lung cancer patients leads to poor survival due to increased metastatic potential confirmed by high CD44 expression in metastatic lymph nodes [48]. ATP-binding cassette subfamily G member 2 favors chemotherapy resistance in lung cancer. In this regard, it has been demonstrated that ATP Binding Cassette Subfamily B Member 1 (ABCB1) inhibitor restores the etopoposide activity inducing lung cancer cell apoptosis [49].
All these findings together confirm that lung CSCs have an important impact on cancer prognosis, strongly suggests that the specific targeting of lung CSCs or the pathways that are able to confer stem cell-like properties to lung cancer cells are effective therapeutic strategies possible in the future [50].
6. Role of cigarette smoke, stem cells, oxidative stress, and Notch-1 signaling pathway in the relationship between chronic obstructive pulmonary disease and lung cancer
Cigarette smoking promotes repeated damage/repair cycles that can compromise the integrity of lung tissues by exhausting the repair potential of stem cells [51]. Furthermore, cigarette smoke contains carcinogenic substances and it is known to promote the onset and progression of cancer. One of the mechanisms through which it promotes cancer progression is the activation of the Notch pathway. Dysregulation of this pathway causes a wide range of respiratory diseases including chronic obstructive pulmonary disease (COPD) and lung cancer [52].
Activation of Notch-1 signaling is promoted by hypoxia-induced microRNAs (miRNAs) and may play a role in triggering lung cancer progression by acting in the regulation of EMT and contributing to aggressive tumor growth, invasion as well as metastasis, and cancer recurrence [24]. In this regard, nutrients and oxygen deprivation induce stromal fibroblasts to release IL-6 and growth factors thus activating a paracrine loop that in turn drives both cell dedifferentiation and tumor formation/progression [53]. Hypoxia also plays a role in the induction of CD133 expression, which has been used as a stem cell biomarker [54]. In patients with COPD, hypoxia may be one of the fundamental biological phenomena that can contribute to the development and aggressiveness of lung cancer [55].
6.1 Impact of cigarette smoke on Notch signaling in non-tumoral cells
Notch-1 is a fundamental signaling system involved in cellular machinery that relies on direct cell–cell contact [56]. In bronchial epithelial cells, activation of Notch-1 by Jag-1 increases Ki-67 and cell proliferation [57]. Exposure to cigarette smoke extract (CSE) in non-tumor bronchial epithelial cells acts negatively on Notch-1 signaling, reducing both the expression of the Notch-1 ligand, Jag-1, and the translocation of the Notch-1 activated form to the nuclear region [57]. Furthermore, although CSE increases the expression of PCNA, Ki-67, and p21, reducing cell proliferation and repair processes are observed.
It has also been demonstrated that CSE exposure promotes a persistent increase in ROS and reduces antioxidant responses [58]. In this way, it could compromise the physiological repair process of the airway epithelium. In this regard, acute exposure to arsenite increases the oxidative stress mechanism and inhibits the proliferation of keratinocyte cells through upregulation of the p21 oncogene and through downregulation of the Notch-1 molecule [59].
The harmful effects of CSE on airway cellular homeostasis could also be due to the regulation of alarmins, such as IL-33. IL-33 belongs to the IL-1 superfamily of cytokines and is produced by endothelial and epithelial cells. IL-33 can translocate into the nucleus and can act as a transcription factor regulating the expression of some important target genes [60]. IL-33 can also be released in the extracellular district and therefore, after binding with its receptor expressed on immune cells, plays a role in the activation of allergic inflammation inducing the release of the cytokines IL-5 and IL-13 [61] and of mucus from the metaplastic epithelium [62]. IL-33 is known to contribute to cellular homeostasis in the airways in response to injury [63] and induces a significant decrease in the expression of the Jag1 molecule [64]. A previous study demonstrates that although CSE increases the presence of IL-33 at the intracellular level, it reduces its release [65].
6.2 Impact of cigarette smoke on Notch signaling in cancer cells
The Notch-1 activation pathway is known to be involved in lung cancer by promoting CSCs maintenance [66]. A histological study on surgical specimens from patients with lung adenocarcinoma has been demonstrated that the lung parenchyma of smokers has an increased expression of Notch-1 but not of CD133 and Jag-1 compared to the lung parenchyma of non-smokers [67]. In a previous study, increased expression of Notch-3 is demonstrated in smokers with lung adenocarcinoma [68]. Furthermore, lung adenocarcinoma cells show constitutive basal activation of Notch-1 signaling as they constitutively express both Notch-1 and Jag-1 and also nuclear NICD. In this regard, the A549 cell line (lung adenocarcinoma) exposed to CSE showed an increase in the expression of the Notch-1 and Hes-1 genes as well as the protein localized in the nuclei without modified Jag-1 gene expression [67].
These results further confirm that the regulation of the Notch pathway can also occur at the transcriptional level, through the activation of molecules, such as Hes-1, that play a role as downstream effectors of the pathway [69]. It has been demonstrated in chronic lymphocytic leukemia that Hes-1 binds to the PTEN promoter region reducing its expression and in turn promoting cell proliferation and inhibiting cell apoptosis [70].
Resistance to the cellular apoptosis mechanism as well as increased cell proliferation are both fundamental events in the progression of cancer. In previous studies, it has been demonstrated that exposure to CSE in A549 induces both the expression of Ki-67 and survivin and that DAPT, a Notch pathway inhibitor, counteracts the CSE effect inducing a downregulation of Ki-67 and survivin [67]. Furthermore, CSE increases the expression of survivin in adenocarcinoma cells also reducing the expression of FoxO3 [71, 72], a transcription factor that negatively controls survivin gene expression [73]. Another study confirms that Notch signaling also promotes cancer progression in other cancer types such as breast cancer by inducing tumor cell survival, proliferation, and apoptotic resistance through increased survivin expression [74]. Survivin promotes cancer progression by exercising a dual role of apoptosis inhibition and mitotic effector, thus protecting tumor cells also from apoptosis induced by drug treatment [75]. Ki-67 promotes cell proliferation and the evaluation of its expression is used as a marker for estimating the degree of histological malignancy and for the prognosis of NSCLC [76]. In fact, it has been demonstrated that in smokers affected by lung adenocarcinoma, the expression of survivin and Ki-67 is increased especially in the initial stages [77]. Another study demonstrates that an increase in survivin expression can also be found in the proximal airways of smoking subjects not affected by lung cancer and this is also confirmed in vitro in bronchial epithelial cells exposed to CSE [78]. Furthermore, it has been observed that nicotine present in cigarette smoke plays a role in increasing survivin in human NSCLC cell lines [79].
All these events are involved in mechanisms promoting cancer development and progression. Indeed, blocking Notch-1 signaling through the inhibitor DAPT or a specific siRNA has been shown to reduce the growth potential of lung CSCs [80].
Taken together, all of this evidence supports a novel mechanism by which exposure to cigarette smoke could promote tumorigenesis by impairing repair processes within the airways due to alterations in redox balance and signaling activation. Nocth-1 and downstream molecular events (Fig. 3).
Figure 3.
(A) Cigarette smoke, induces oxidative stress, affects the self-renewal ability of stem cells impairing tissue repair, and promotes mutagenic events and dedifferentiation processes leading to cancer stem cells. (B) Notch-1 pathway activation affects cell proliferation, survival, and angiogenesis suggesting important roles in the tumorigenesis process. (D) Notch-1 pathway activation confers metastatic properties and resistance to therapy contributing to the progression of cancer including cancer metastasis.
7. Effects of natural compounds
Natural compounds have been widely recognized to have the potential for the prevention of cancer progression and as complementary or standalone treatments for cancer patients [81]. Dietary factors may modulate the impact of adverse environmental exposures or genetic predisposition within the lung. Changes in diet over the past few decades, with decreased consumption of fruits, vegetables, whole grains, and fish, and increased consumption of processed and refined foods have a negative impact on human health increasing disability due to chronic diseases such as COPD or increasing lethality due to cancer onset and progression. It has now emerging evidence that dietary patterns rather than individual nutrients exert a protective effect suggesting that synergistic interactions between multiple nutrients contained in dietary patterns are responsible for the observed benefits [82]. Targeting oxidative stress with antioxidants or boosting endogenous levels of antioxidants can be an effective strategy in the management of COPD and in preventing cancer onset and progression. Adequate fruit and vegetable consumption is associated with lower markers of oxidative stress and inflammation, and higher levels of antioxidant markers [83].
Polyphenols, a class of natural compounds largely present in fruits and vegetables, have been shown to act as anti-inflammatory agents. Phenolic acids, flavonoids, and stilbenes (resveratrol), all belonging to polyphenols, have been reportedly associated with the prevention of chronic diseases and cancer [84]. The most relevant mechanisms by which polyphenols are beneficial are related to the modulation of the Nrf2 pathway [81]. As previously mentioned aberrant Nrf2-Notch crosstalk exerts a relevant role in lung cancer [40] and accordingly several experimental studies demonstrate that polyphenol-mediated cancer cell death is linked to Nrf2 downregulation [81]. Luteolin, a flavonoid suppresses Nrf2 activity by increasing Nrf2 mRNA turnover and sensitizes NSCLC A549 cells to therapeutic drugs. Resveratrol reduces the migratory and invasive abilities of A549 by inhibiting heme oxygenase (HO). Apigenin, a natural dietary flavonoid, is an effective small molecule inhibitor against Nrf2 and has displayed anticancer activity in various cancers [85]. It has been demonstrated that apigenin in A549 increases the production of ROS, with augmented cell death and apoptosis [86]. The toxic effects of apigenin on lung cancer cells are maintained also in the presence of pro-survival events such as leptin pathway activation or exposure to pleural fluids from patients with lung cancer [86]. Moreover, a systematic review and meta-analysis performed on 8799 cases of lung cancer and 17 072 non-tumoral cases demonstrates that the dietary patterns containing fish intake are associated with a decreased risk of lung cancer [87]. In this regard, the protective effects of fish can be associated with the high content of omega-3 that are potent modulators of oxidative stress [88].
Omega 3 metabolites are substrates for oxidants inside the cell into cell membranes and promote the formation of nonenzymatic lipid peroxidation products whose biological activity is context and dose-dependent. In fact, they have also been reported to be cytoprotective and anti-inflammatory in macrophages [89] but are highly toxic in cancer cells [90]. The levels of these toxic compounds are higher in cancer cells than in normal cells since tumor cells contain higher levels of ROS compared to normal cells, principally due to their accelerated metabolism needed to maintain their high proliferation rate. It has been reported that electrophilic keto-derivative of the omega-3 fatty acid docosahexaenoic acid (DHA), endogenously generated by cyclooxygenase-2 and cellular dehydrogenase, promotes anti-lung cancer effects. This electrophilic keto-derivative of the omega-3 s, alone and more when added to gemcitabine, reduces proliferation and increases cell apoptosis on a panel of five histologically different human NSCLC cell lines [91].
8. Conclusions
This review offers a comprehensive view of the effects of oxidative stress in Notch-1 signaling activation in CSCs and adult stem cells (ASCs) in order to better understand both initiation and progression processes in lung cancer. Several mechanisms have been elucidated and potential new therapeutic targets have been identified to contrast cancer growth. Moreover, in COPD patients, targeting oxidative stress with natural compounds can be an easy way to stunt tumor onset, growth, and progression. The role exerted by the complex relationship among Notch, cigarette smoke, and stemness in lung cancer biology deserves further investigation.
Contributor Information
Giuseppina Chiappara, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Palermo, via Ugo La Malfa 153, 90146, Italy.
Serena Di Vincenzo, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Palermo, via Ugo La Malfa 153, 90146, Italy.
Caterina Cascio, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Palermo, via Ugo La Malfa 153, 90146, Italy.
Elisabetta Pace, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Palermo, via Ugo La Malfa 153, 90146, Italy.
Author contributions
Giuseppina Chiappara contributed to the conceptualization and wrote the original draft of the review, Serena Di Vincenzo and Caterina Cascio reviewed the manuscript and Elisabetta Pace is responsible for conceptualization, supervision, and for the final revision of the manuscript. All authors have read, reviewed, and approved the final manuscript.
Conflict of interest statement: All the authors declare that there is no conflict of interest regarding the publication of this paper.
Funding
This work was supported by the Italian National Research Council (FOE 2022-DSB.AD006.361.028) and by the project “One Health Basic and Translational Research Actions Addressing Unmet Needs on Emerging Infectious Diseases (IN-FACT) Proj n. PE00000007 CUP B53C20040570005.
Data availability
Data is contained within the article.
References
- 1. Zhou B, Lin W, Long Y. et al. Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022;7:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Venkatesh V, Nataraj R, Thangaraj GS. et al. Targeting Notch signalling pathway of cancer stem cells. Stem Cell Investig 2018;5:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Dayem AA, Choi H-Y, Kim J-H. et al. Role of oxidative stress in stem, cancer, and cancer stem cells. Cancers 2010;2:859–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Leeman KT, Fillmore CM, Kim CF.. Lung stem and progenitor cells in tissue homeostasis and disease. Curr Top Dev Biol 2014;107:207–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. de Morree A, Rando TA.. Regulation of adult stem cell quiescence and its functions in the maintenance of tissue integrity. Nat Rev Mol Cell Biol 2023;24:334–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Schultz MB, Sinclair DA.. When stem cells grow old: phenotypes and mechanisms of stem cell aging. Development 2016;143:3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Allen NC, Reyes NS, Lee JY. et al. Intersection of inflammation and senescence in the aging lung stem cell niche. Front Cell Dev Biol 2022;10:932723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Abdel-Daim MM, Moustafa YM, Umezawa M. et al. Applications of antioxidants in ameliorating drugs and xenobiotics toxicity: mechanistic approach. Hindawi 2017;2017:4565127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Chen W-J, Ho C-C, Chang Y-L. et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat Commun 2014;5:3472. [DOI] [PubMed] [Google Scholar]
- 10. Aramini B, Masciale V, Grisendi G. et al. Dissecting tumor growth: the role of cancer stem cells in drug resistance and recurrence. Cancers (Basel) 2022;14:976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Phi LTH, Sari IN, Yang Y-G. et al. Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int 2018;2018:5416923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Arnold CR, Mangesius J, Skvortsova I-I. et al. The role of cancer stem cells in radiation resistance. Front Oncol 2020;10:164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Hicks MR, Pyle AD.. The emergence of the stem cell niche. Trends Cell Biol 2023;33:112–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Farahzadi R, Valipour B, Montazersaheb S. et al. Targeting the stem cell niche micro-environment as therapeutic strategies in aging. Front Cell Dev Biol 2023;11:1162136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Sharif A, Shaji A, Chammaa M. et al. Notch transduction in non-small cell lung cancer. Int J Mol Sci 2020;21:5691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kopan R, Ilagan MXG.. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell 2009;137:216–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Guo D, Ye J, Li L. et al. Down-regulation of Notch-1 increases co-cultured Jurkat cell sensitivity to chemotherapy. Leuk Lymphoma 2009;50:270–8. [DOI] [PubMed] [Google Scholar]
- 18. Riya PA, Basu B, Surya S. et al. HES1 promoter activation dynamics reveal the plasticity, stemness and heterogeneity in neuroblastoma cancer stem cells. J Cell Sci 2022;135:jcs260157. [DOI] [PubMed] [Google Scholar]
- 19. Dongre A, Weinberg RA.. New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019;20:69–84. [DOI] [PubMed] [Google Scholar]
- 20. Kar R, Jha NK, Jha SK. et al. A “NOTCH” deeper into the Epithelial-To-Mesenchymal Transition (EMT) Program in breast cancer. Genes (Basel) 2019;10:961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Wang H, Unternaehrer JJ.. Epithelial–mesenchymal transition and cancer stem cells: at the crossroads of differentiation and dedifferentiation. Dev Dyn 2019;248:10–20. [DOI] [PubMed] [Google Scholar]
- 22. Allen F, Maillard I.. Therapeutic targeting of notch signaling: from cancer to inflammatory disorders. Front Cell Dev Biol 2021;9:649205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Dotto GP. Notch tumor suppressor function. Oncogene 2008;27:5115–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Espinoza I, Pochampally R, Xing F. et al. Notch signaling: targeting cancer stem cells and epithelial-to-mesenchymal transition. Onco Targets Ther 2013;6:1249–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Pizzino G, Irrera N, Bitto A. et al. Oxidative stress: harms and benefits for human health. Oxid Med Cell Longev 2017;2017:6341671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Jomova K, Raptova R, Alomar SY. et al. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol 2023;97:2499–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Snezhkina AV, Kudryavtseva AV, Kardymon OL. et al. ROS generation and antioxidant defense systems in normal and malignant cells. Oxid Med Cell Longev 2019;2019:6175804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Nita M, Grzybowski A.. The role of the reactive oxygen species and oxidative stress in the pathomechanism of the age-related ocular diseases and other pathologies of the anterior and posterior eye segments in adults. Oxid Med Cell Longev 2016;2016:3164734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Clerkin J, Naughton R, Quiney C. et al. Mechanisms of ROS modulated cell survival during carcinogenesis. Cancer Lett 2008;266:30–6. [DOI] [PubMed] [Google Scholar]
- 30. Zafar A, Singh S, Satija YK. et al. Deciphering the molecular mechanism underlying anticancer activity of coumestrol in triple-negative breast cancer cells. Toxicol In Vitro 2018;46:19–28. [DOI] [PubMed] [Google Scholar]
- 31. Schieber M, Chandel NS.. ROS function in redox signaling and oxidative stress. Curr Biol 2014;24:R453–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Gill JG, Piskounova E, Morrison SJ.. Cancer, oxidative stress, and metastasis. Cold Spring Harb Symp Quant Biol 2016;81:163–75. [DOI] [PubMed] [Google Scholar]
- 33. Boonstra J, Post JA.. Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene 2004;337:1–13. [DOI] [PubMed] [Google Scholar]
- 34. Verbon EH, Post JA, Boonstra J.. The influence of reactive oxygen species on cell cycle progression in mammalian cells. Gene 2012;511:1–6. [DOI] [PubMed] [Google Scholar]
- 35. Speijer D. Oxygen radicals shaping evolution: why fatty acid catabolism leads to peroxisomes while neurons do without it: FADH2/NADH flux ratios determining mitochondrial radical formation were crucial for the eukaryotic invention of peroxisomes and catabolic tissue differentiation. Bioessays 2011;33:88–94. [DOI] [PubMed] [Google Scholar]
- 36. Bridge G, Rashid S, Martin SA.. DNA mismatch repair and oxidative DNA damage: implications for cancer biology and treatment. Cancers (Basel) 2014;6:1597–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Dharmaraja AT. Role of reactive oxygen species (ROS) in therapeutics and drug resistance in cancer and bacteria. J Med Chem 2017;60:3221–40. [DOI] [PubMed] [Google Scholar]
- 38. Robinett ZN, Bathla G, Wu A. et al. Persistent oxidative stress in vestibular schwannomas after stereotactic radiation therapy. Otol Neurotol 2018;39:1184–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Paul MK, Bisht B, Darmawan DO. et al. Dynamic changes in intracellular ROS levels regulate airway basal stem cell homeostasis through Nrf2-dependent Notch signaling. Cell Stem Cell 2014;15:199–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Sparaneo A, Fabrizio FP, Muscarella LA.. Nrf2 and notch signaling in lung cancer: near the crossroad. Oxid Med Cell Longev 2016;2016:7316492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Borthwick DW, Shahbazian M, Krantz QT. et al. Evidence for stem-cell niches in the tracheal epithelium. Am J Respir Cell Mol Biol 2001;24:662–70. [DOI] [PubMed] [Google Scholar]
- 42. Cole BB, Smith RW, Jenkins KM. et al. Tracheal basal cells: a facultative progenitor cell pool. Am J Pathol 2010;177:362–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Mulvihill MS, Kratz JR, Pham P. et al. The role of stem cells in airway repair: implications for the origins of lung cancer. Chin J Cancer 2013;32:71–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Fukui T, Shaykhiev R, Agosto-Perez F. et al. Lung adenocarcinoma subtypes based on expression of human airway basal cell genes. Eur Respir J 2013;42:1332–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Zheng Y, Wang L, Yin L. et al. Lung cancer stem cell markers as therapeutic targets: an update on signaling pathways and therapies. Front Oncol 2022;12:873994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Wang S, Xu ZY, Wang LF. et al. CD133+ cancer stem cells in lung cancer. Front Biosci 2013;18:53. [DOI] [PubMed] [Google Scholar]
- 47. Ma Z, Zhang C, Liu X. et al. Characterisation of a subpopulation of CD133+ cancer stem cells from Chinese patients with oral squamous cell carcinoma. Sci Rep 2020;10:8875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Li G, Gao Y, Cui Y. et al. Overexpression of CD44 is associated with the occurrence and migration of non-small cell lung cancer. Mol Med Rep 2016;14:3159–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Omori M, Noro R, Seike M. et al. Inhibitors of ABCB1 and ABCG2 overcame resistance to topoisomerase inhibitors in small cell lung cancer. Thorac Cancer 2022;13:2142–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Varghese B, Ling Z, Ren X.. Reconstructing the pulmonary niche with stem cells: a lung story. Stem Cell Res Ther 2022;13:161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Deeb RS, Walters MS, Strulovici-Barel Y. et al. Smoking-associated disordering of the airway basal stem/progenitor cell metabotype. Am J Respir Cell Mol Biol 2016;54:231–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Kiyokawa H, Morimoto M.. Notch signaling in the mammalian respiratory system, specifically the trachea and lungs, in development, homeostasis, regeneration, and disease. Dev Growth Differ 2020;62:67–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Rodrigues CF et al. Stroma-derived IL-6, G-CSF and Activin-A mediated dedifferentiation of lung carcinoma cells into cancer stem cells. Sci Rep 2018;8:11573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Li Z. CD133: a stem cell biomarker and beyond. Exp Hematol Oncol 2013;2:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Bao B, Azmi AS, Ali S. et al. The biological kinship of hypoxia with CSC and EMT and their relationship with deregulated expression of miRNAs and tumor aggressiveness. Biochim Biophys Acta 2012;1826:272–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zou B, Zhou X-L, Lai S-Q. et al. Notch signaling and non-small cell lung cancer. Oncol Lett 2018;15:3415–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Di Sano C, D'Anna C, Ferraro M. et al. Impaired activation of Notch-1 signaling hinders repair processes of bronchial epithelial cells exposed to cigarette smoke. Toxicol Lett 2020;326:61–9. [DOI] [PubMed] [Google Scholar]
- 58. Pace E, Ferraro M, Siena L. et al. Carbocysteine regulates innate immune responses and senescence processes in cigarette smoke stimulated bronchial epithelial cells. Toxicol Lett 2013;223:198–204. [DOI] [PubMed] [Google Scholar]
- 59. Cialfi S, Palermo R, Manca S. et al. Loss of Notch1-dependent p21(Waf1/Cip1) expression influences the Notch1 outcome in tumorigenesis. Cell Cycle 2014;13:2046–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Chan BCL, Lam CWK, Tam LS. et al. IL33: roles in allergic inflammation and therapeutic perspectives. Front Immunol 2019;10:394368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Takatori H, Makita S, Ito T. et al. Regulatory mechanisms of IL-33-ST2-mediated allergic inflammation. Front Immunol 2018;9:401513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Han M, Hong JY, Jaipalli S. et al. IFN-γ blocks development of an asthma phenotype in rhinovirus-infected baby mice by inhibiting type 2 innate lymphoid cells. Am J Respir Cell Mol Biol 2017;56:242–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Michaudel C, Mackowiak C, Maillet I. et al. Ozone exposure induces respiratory barrier biphasic injury and inflammation controlled by IL-33. J Allergy Clin Immunol 2018;142:942–58. [DOI] [PubMed] [Google Scholar]
- 64. Imaeda H, Andoh A, Aomatsu T. et al. Interleukin-33 suppresses Notch ligand expression and prevents goblet cell depletion in dextran sulfate sodium-induced colitis. Int J Mol Med 2011;28:573–8. [DOI] [PubMed] [Google Scholar]
- 65. Pace E, Di Sano C, Sciarrino S. et al. Cigarette smoke alters IL-33 expression and release in airway epithelial cells. Biochim Biophys Acta 2014;1842:1630–7. [DOI] [PubMed] [Google Scholar]
- 66. Hassan KA, Wang L, Korkaya H. et al. Notch pathway activity identifies cells with cancer stem cell-like properties and correlates with worse survival in lung adenocarcinoma. Clin Cancer Res 2013;19:1972–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Chiappara G, Di Vincenzo S, Sangiorgi C. et al. Cigarette smoke upregulates Notch-1 signaling pathway and promotes lung adenocarcinoma progression. Toxicol Lett 2022;355:31–40. [DOI] [PubMed] [Google Scholar]
- 68. Cheng Z, Tan Q, Tan W. et al. Cigarette smoke induces the expression of Notch3, not Notch1, protein in lung adenocarcinoma. Oncol Lett 2015;10:641–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Meisel CT, Porcheri C, Mitsiadis TA.. Cancer stem cells, quo vadis? The Notch signaling pathway in tumor initiation and progression. Cells 2020;9:1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Zhang Q, Zhu Z, Guan J. et al. Hes1 controls proliferation and apoptosis in chronic lymphoblastic leukemia cells by modulating PTEN expression. Mol Biotechnol 2022;64:1419–30. [DOI] [PubMed] [Google Scholar]
- 71. Di Vincenzo S, Sangiorgi C, Ferraro M. et al. Cigarette smoke extract reduces FOXO3a promoting tumor progression and cell migration in lung cancer. Toxicology 2021;454:152751. [DOI] [PubMed] [Google Scholar]
- 72. Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C. et al. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci 2020;21:9739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Pace E, Di Vincenzo S, Ferraro M. et al. Carbocysteine counteracts the effects of cigarette smoke on cell growth and on the SIRT1/FoxO3 axis in bronchial epithelial cells. Exp Gerontol 2016;81:119–28. [DOI] [PubMed] [Google Scholar]
- 74. Chen L, Zhang J, Lyu Z. et al. Positive feedback loop between mitochondrial fission and Notch signaling promotes survivin-mediated survival of TNBC cells. Cell Death Dis 2018;9:1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Li D, Hu C, Li H.. Survivin as a novel target protein for reducing the proliferation of cancer cells. Biomed Rep 2018;8:399–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Nguyen VN, Mirejovský P, Mirejovský T. et al. Expression of cyclin D1, Ki-67 and PCNA in non-small cell lung cancer: prognostic significance and comparison with p53 and bcl-2. Acta Histochem 2000;102:323–38. [DOI] [PubMed] [Google Scholar]
- 77. Hirano H, Maeda H, Yamaguchi T. et al. Survivin expression in lung cancer: association with smoking, histological types and pathological stages. Oncol Lett 2015;10:1456–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Chiappara G, Gjomarkaj M, Virzì A. et al. The role of p21 Waf1/Cip1 in large airway epithelium in smokers with and without COPD. Biochim Biophys Acta 2013;1832:1473–81. [DOI] [PubMed] [Google Scholar]
- 79. Dasgupta P, Kinkade R, Joshi B. et al. Nicotine inhibits apoptosis induced by chemotherapeutic drugs by up-regulating XIAP and survivin. Proc Natl Acad Sci USA 2006;103:6332–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Cai H, Lu W, Zhang Y. et al. Specific inhibition of Notch1 signaling suppresses properties of lung cancer stem cells. J Cancer Res Ther 2019;15:1547–52. [DOI] [PubMed] [Google Scholar]
- 81. Ghareghomi S, Moosavi-Movahedi F, Saso L. et al. Modulation of Nrf2/HO-1 by natural compounds in lung cancer. Antioxidants (Basel, Switzerland) 2023;12:735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Scoditti E, Massaro M, Garbarino S. et al. Role of diet in chronic obstructive pulmonary disease prevention and treatment. Nutrients 2019;11:1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Holt EM, Steffen LM, Moran A. et al. Fruit and vegetable consumption and its relation to markers of inflammation and oxidative stress in adolescents. J Am Diet Assoc 2009;109:414–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Chu M, Zheng C, Chen C. et al. Targeting cancer stem cells by nutraceuticals for cancer therapy. Semin Cancer Biol 2022;85:234–45. [DOI] [PubMed] [Google Scholar]
- 85. Paredes‐Gonzalez X, Fuentes F, Jeffery S. et al. Induction of NRF2‐mediated gene expression by dietary phytochemical flavones apigenin and luteolin. Biopharm Drug Dispos 2015;36:440–51. [DOI] [PubMed] [Google Scholar]
- 86. Bruno A, Siena L, Gerbino S. et al. Apigenin affects leptin/leptin receptor pathway and induces cell apoptosis in lung adenocarcinoma cell line. Eur J Cancer 2011;47:2042–51. [DOI] [PubMed] [Google Scholar]
- 87. Song J, Su H, Wang B-L. et al. Fish consumption and lung cancer risk: systematic review and meta-analysis. Nutr Cancer 2014;66:539–49. [DOI] [PubMed] [Google Scholar]
- 88. D’Eliseo D, Velotti F.. Omega-3 fatty acids and cancer cell cytotoxicity: implications for multi-targeted cancer therapy. J Clin Med 2016;5:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Gutiérrez S, Svahn SL, Johansson ME.. Effects of omega-3 fatty acids on immune cells. Int J Mol Sci 2019;20:5028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Siddiqui RA, Harvey K, Stillwell W.. Anticancer properties of oxidation products of docosahexaenoic acid. Chem Phys Lipids 2008;153:47–56. [DOI] [PubMed] [Google Scholar]
- 91. Siena L, Cipollina C, Di Vincenzo S. et al. Electrophilic derivatives of omega-3 fatty acids counteract lung cancer cell growth. Cancer Chemother Pharmacol 2018;81:705–16. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data is contained within the article.




