Skip to main content

15-Lipoxygenase and its metabolites in the pathogenesis of breast cancer: A double-edged sword

Abstract

15-lipoxygenase is one of the key enzymes for the metabolism of unsaturated fatty acids that its manipulation has been proposed recently as a new molecular target for regulating cancer cell growth. Aberrant expression of 15-lipoxygenase enzyme seems to play an indicative role in the pathology of different cancer types, tumor progression, metastasis, or apoptosis. Based on the fact that breast cancer is one of the most common cancers that imposes a burden of mortality in women also, on the other hand, evidence in experimental models and human studies indicate the emerging role of the 15-lipoxygenase pathway in breast cancer pathogenesis, we present a review of recent findings related to the role of 15- lipoxygenase enzyme and metabolites in breast cancer growth, apoptosis, metastasis, and invasion as well as their local and circulating expression pattern in patients with breast cancer. Our review supports the emerging role of 15- lipoxygenase in molecular and cellular processes regulating breast tumor cell fate with both positive and negative effects.

Metabolism of arachidonic acid, as a long-chain polyunsaturated fatty acid

Omega-3 and omega-6 fatty acids are a family of polyunsaturated fatty acids (PUFA) that play a vital role in the body’s physiological activities due to their presence in cell membranes [1]. Dietary PUFAs include alpha-linolenic acid (ALA), as a short-chain omega-3 fatty acid, and linoleic acid (LA) as a short-chain omega-6 fatty acid, also, eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and arachidonic acid (AA) as longer chain omega-3 PUFAs [2]. AA can be produced from the essential fatty acids LA or ALA, direct intake from diet, or release from phospholipid pool by the action of phospholipase A2. The biosynthesis pathway of eicosanoids (bioactive 20-carbon lipid mediators) starts with the conversion of LA to ALA due to the catalytic activity of desaturase. Then, under the influence of the enzyme elongase, this compound is converted to AA, which is a fatty acid with 20 carbon atoms and 4 double bonds [3]. AA can produce eicosanoids through three enzymatic pathways including several oxygenases that generate a spectrum of oxygenated products. One metabolizing way is the cyclooxygenase (COX) pathway, which produces different prostaglandin (PG), such as prostaglandin E (PGE) and PGD, and thromboxane (TX), such as thromboxane A2 (TXA2) and TXA3, and prostacyclins such as prostacyclin I2 and I3 [4]. Two distinct isoforms of the COX enzyme exist, namely COX-1 and COX-2 that are constitutively expressed in most types of cells, although COX-2 is the most regulated type of the COX enzyme that its expression is induced in inflammation and proliferative diseases. Therefore, COX inhibitors appeared to be effective in the treatment of pain, inflammation, stroke, and cancer [2]. The cytochrome P450 (CYP) is the second metabolizing pathway of AA that mainly produces hydroxyeicosatetraenoic acids (HETEs) due to its ω-hydroxylase activity. However, CYP epoxygenase activity resulted in producing epoxyeicosatrienoic acids (EETs) that can further have metabolized by epoxide hydrolase and generate the corresponding diols [5]. Pieces of evidence revealed the vasodilatation effect of EETs as well as their regulatory role on the tumorigenesis process [6]. The last metabolizing way is the lipoxygenase (LOX) pathway that mainly generates leukotrienes (LT), such as LTA4, LTB4, LTC4, LTD4, and LTE4, as well as lipoxins A4, B4, C4, D4, and E4 [7]. LOX pathway is becoming dominant recently as a therapeutic target in many diseases due to its pivotal role in cellular signaling and novel mediators related to this pathway that has been discovered, therefore, the metabolic pathway of LOX is examined in more detail in the next section.

The role of the 15-LOX metabolizing pathway in the metabolism of polyunsaturated fatty acids

LOX enzymes are a family of iron (non-heme) dioxygenases that can transport oxygen molecules into the free or esterified unsaturated fatty acids. These enzymes are divided into four categories based on the position that the oxygen group enters: lipoxygenase types 5, 8, 12, and 15 [8]. The LOX enzyme uses two major substrates in cells, including AA, a polyunsaturated omega-6 fatty acid (20:4(5,8,11,14)), and LA, a polyunsaturated omega-6 fatty acid (8,2 cis-9,12). The primary product of the enzymatic function of lipoxygenase is hydro peroxy n-6 fatty acids, which are rapidly converted to hydroxy derivatives. Membrane phospholipids are converted to arachidonic acid by phospholipase A4 and phospholipase C, and then AA can be metabolized by lipoxygenase enzymes. The lipoxygenase metabolizes AA and converts it to cyclic hydroperoxides such as hydroperoxy eicosatetraenoicacid (HpETE), Hydroxyeicosatetraenoic acid (HETE), and LTA4. The active metabolites of HpETE are then reduced and converted to HETE. The next substrate for LOX is LA, which is converted to 13-Hydroxyoctadecadienoic acid (13-HODE) [9, 10]. Among all types of lipoxygenases, 15-lipoxygenase (15-LOX), is the target of this study due to its extensive and regulatory roles in cancer pathogenesis. 15-LOX, formerly known as arachidonate lipoxygenase, is made up of two types of isoenzymes: 15-lipoxygenase type 1 (15-LOX-1), and 15- lipoxygenase type 2 (15-LOX-2, 8]. 15-LOX is mainly expressed in reticulocytes, eosinophils and epithelial cells, including respiratory epithelium and macrophages [10]. Intracellular 15-LOX activity is highly regulated and the regulation of this enzyme occurs at the levels of transcription, translation, and post-translation in different cellular systems. The results of previous studies showed that 15-LOX at the transcriptional level can be induced by interleukins [11]. In human monocytes, adenocarcinoma human alveolar basal epithelial cells (A549 cell line), and human colorectal adenocarcinoma cells (Caco2 cell line), 15-LOX expression level was induced following treatment with interleukin 4 (IL4). It was shown that activation of IL4 cell surface receptor, resulted in the activation of signal transducer and activator of transcription 6 (STAT6), and transport of its phosphorylated homodimer to the nucleus to bind to STAT6-responsible agents including IL4-related genes such as 15-LOX [12]. Notably, 15-LOX-1 transcription can be activated both in a STAT6- dependent and independent manner via histone modification of the 15-LOX promotor [13]. Also, it was shown that Interleukin-13 (IL-13) induces the 15-LOX-1 expression and activity that resulted in the elevation of 15-LOX-1 metabolites, activation of peroxisome proliferator-activated receptor-gamma (PPAR-γ), initiation of apoptosis in glioblastoma cells [14]. On the other hand, the transcriptional activity of PPARγ can be facilitated by STAT6 indicating the crucial role of the IL13–15-LOX-PPARγ axis in regulating cell growth and invasion [15]. Besides, it was postulated that 15-LOX expression can be regulated by acetylation of histones in the nucleus [16]. Moreover, Nitric oxide (NO) is assumed to be another regulatory factor for 15-LOX activity, as it was revealed that incubation of 15-LOX for a short time with NO increases the length of the catalytic period of the enzyme, which can cause reversible inhibition of the enzyme [17]. Another important regulatory factor at the post-translational level is the membrane binding ability of 15-LOX in a calcium-dependent manner. When the 15-LOX enzyme was incubated with biological membranes in the presence of calcium ions, the enzyme’s ability to bind to the membrane enhanced that was associated with a 10-fold increase in the enzyme’s catalytic ability to oxidize fatty acids [18]. The 15-LOX the enzyme is involved in various cellular processes, including cell differentiation, development, mitochondrial degradation, and reticulocyte maturation [19,20,21]. Levels of 15-LOX products (15(S)- hydroperoxy eicosatetraenoic acid (15-S-HETE), 3(S)-hydroxy-9Z,11E-octadecadienoic acid (13(S)-HODE)) in tissues usually increase during inflammation [22]. However, the role of lipoxygenase enzymes in the development and spread of cancer is very complex and controversial. Evaluating the expression and activity level of lipoxygenases in epithelial cancerous and normal tissues in humans and mice shows that 15-LOX- type 1 and 2 are preferably expressed more in normal /benign tissues than cancerous tissues of the bladder [23], breast [24], colon [25], lung [26] and prostate [19]. However, conflicting evidence exists regarding the 15-LOX expression pattern in the aforementioned tissues. Based on shreds of evidence, LOX isoforms revealed both anti-tumorigenic and pro-tumorigenic effects in different tumor tissues [16, 27]. In breast cancer, both isoforms of 15- LOX are down-regulated in the cancerous epithelium than in the normal tissue, while a high level of the enzymes 12 and 5-LOX are expressed in the breast cancer sample and tissue [28]. The results of other studies show that increasing the expression level of 12-LOX and decreasing the level of 15-LOX in breast cancer patients play a prognostic role [29]. Similarly, in non-Small Cell Lung Cancer (NSCLC), the expression of 15-LOX-2 increased in well-differentiated tumors [26]. It can be concluded that the role of LOX expression in cancer growth depends on the type of tissue and the type of isoform that is expressed. To find the role of each isoform in cancer progression, it is necessary to examine the role of LOX metabolism in various aspects of cancer development, including cell growth, cell invasion, angiogenesis, and metastasis. In this review, we present recent evidence and findings indicating the role of the 15-LOX pathway in breast cancer and we also discuss the significance and importance of 15-LOX enzyme isoforms and main products on breast cancer cell growth, death, metastasis, and invasion. Identifying the role of the 15-LOX pathway, as a critical pathway in the process of lipid metabolism, in breast cancer pathogenesis may enhance our understanding of the putative mechanisms underlying breast tumor onset and progression and also may open up promising solutions for more effective treatments of this cancer.

How 15-LOX works in a breast normal and tumor cell and what is the 15-LOX putative mechanism of action?

Given the breadth of 15-LOX-mediated cell death in breast tumors, the expression level of 15-LOX and its products have been surveyed in several studies that are summarized in Table 1. Evaluating the transcript level of 15-LOX in tumor tissues of patients with breast cancer showed a significant reduction of 15-LOX expression level compared to the normal breast tissues in node-positive patients [24]. The attenuated level of 15-LOX was associated with tumor stage and grade since the lowest level of 15-LOX was detected in TNM4 breast tumors. Also, the lower level of 15-LOX was detected in Lobular carcinomas compared to the ductal breast tumors [24]. Alongside with 15-LOX reduced level, over-expression of 12-LOX and cyclooxygenase-2 (COX-2) was observed in breast tumor tissues while 5-LOX level remained un-changed in breast tumor and normal tissues [28]. In accordance, it was shown that both 15-LOX isoforms (15-LOX-1 and 15-LOX-2) were expressed in breast normal epithelial cells and vascular endothelial cells while breast tumor cells showed lower protein levels of both 15-LOX isoforms compared to the normal breast cells. Also, in node-positive tumors, the 15-LOX-1 transcript was lower compared to the node-negative tumors. In addition, the lower level of 15-LOX-2: Cytokeratin 19 (CK19) ratio was detected in TNM3 tumors compared to TNM1 tumors indicating the trend of high grades tumors to down-regulate 15-LOX isoforms. Interestingly, in estrogen receptor (ER)-positive breast tumors, the expression level of 15-LOX-2 was reduced in the corresponding cells while the 15-LOX-1 expression level remained unchanged compared to the ER-negative tumors. The association of metastasis occurrence and development, tumor recurrence, and patient survival with a lower level of 15-LOX-2 and 15-LOX-1 was detected in patients with breast cancer. Based on the findings, the 15LOX1:15LOX2 ratio has prognostic value in predicting the clinical outcome of patients with breast cancer [24]. A high level of the 15-LOX-2 transcript was detected in normal prostate, breast, bladder, and skin tissues, while the expression level of 15-LOX-2 reduced remarkably in tumors of the mentioned tissues [30, 35, 36]. It might be explained by the hypothesis that 15-LOX-2 is localized in Golgi-like structures inside the normal epithelial cells, however, the mentioned structures are not found in the transformed cells. It was revealed that the 15-LOX-2 product (15-S-HETE) level was elevated in normal cells while reduced in the correspondence tumor cells. The regulatory role of PPAR-γ was demonstrated in the study of Subbarayan et al.; that based on their report, stimulation of PPAR-γ expression caused down-regulation of 15-LOX-2 in breast and lung normal epithelial cells. Accordingly, over-expression of 15-LOX-2 or 15-S-HETE in tumor cells was accompanied by reduced PPAR-γ protein level [30]. In another survey, the preoperative serum level of prostaglandin E2 (PGE2) and 6-keto-Prostaglandin F1 Alpha (6-k-PGF) in patients with malignant breast tumors was significantly higher compared to the postoperative levels of PGE2 and 6-k-PGF [31]. The eicosanoid profile of breast tumor tissues indicated that the eicosanoid metabolites except for prostaglandin F2α (PGF2α), heptadecatrienoic acid (HHT), and 15-Hydroxyeicosatetraenoic acid (15-HETE) were higher compared to the benign breast tumor or mammary reduction. A correlation was observed between tumor value of LOX products such as (15-HETE) and 13,14-dihydro-l5-keto-prostaglandin (DHKPG) with tumor diameter and fibrosis score. As a result, the eicosanoid synthesis profile showed a different pattern in mammary tumors and normal tissues [31]. In an in-vitro study, it was observed that 13(S)-HODE suppressed the growth of MCF-7 and MDA-MB-231 breast cancer cell lines in a dose/time-dependent manner. Moreover, the increasing concentration of 13(S)-HODE was accompanied by the accumulation of cells in the Sub-G1 phase of the cell cycle and the induction of early apoptosis. It was shown that the expression level of PPAR-γ was attenuated following treatment of breast cancer cells with 13(S)-HODE [37]. To clarify the expression status of 15-LOX-2 and its relationship with PPAR-γ, various normal and tumor tissues from epithelial and non-epithelial sources were enrolled in the survey. The normal epithelial cells derived from prostate, breast, lung, bladder, and skin tissues, expressed a high level of 15-LOX-2 gene and protein while low level of PPAR-γ. In contrast, the malignant tumors and the transformed epithelial cells of the prostate (PC-3 and DU145), breast (MDA453), lung (Calu I), bladder (U-9 and U-14), skin (HaCaT), and pancreas (Mia PaCa-2 and ASPC-1) exhibited a low level of 15-LOX-2 and high level of PPAR-γ. However, other epithelial cells of the same tissues including prostate (LNCaP), lung (MSK-3), breast (MCF-7 and SK-BR-3), and skin (SCC-M7 and SCC-P9), expressed a low level of 15-LOX-2 gene and protein while the level of PPAR-γ was not as high as the other malignant epithelial cells [30]. Treatment of MCF-7 cells with arachidonic acid and 15-L-(S)- hydroperoxy eicosatetraenoic acid (15-L-(S)-HPETE) induced cell toxicity in a time and dose-dependent manner and the cytotoxic effect of 15-L-(s)-HPETE, as a 15-LOX product was more remarkable [38]. It was found that 15-LOX-1 suppressed tumor growth and metastasis development in transgenic mice with mammary gland carcinoma and/ also mice with Lewis lung carcinoma [39]. In addition to studies that reported the decreased expression of the 15-LOX and its products, significant studies have shown that the LOX enzyme expression and activity increase in patients with breast cancer/ breast cancer cells (summarized in Fig. 1). In accordance, in the survey to determine the expression pattern of arachidonic acid metabolizing enzymes in a panel of human epithelial cancer cell lines, it was determined that cyclooxygenase-1 (COX-1), 5-lipoxygenase (5-LOX), and 5-lipoxygenase activating protein (FLAP) demonstrated a universal expression in all cells while the expression level of COX-2, 12-lipoxygenase (12-LOX), and 15-LOX revealed to be variable based on the type of cell line. For instance, the 15-LOX transcript was expressed in all types of colon and lung cancer cell lines while among 7 types of breast cancer cell lines (MB231, H2380, SKBR3, T47D, ZR75, MCF-WT, and MCF7-adr), only SKBR3 cells were not able to express 15-LOX also the expression of 15-LOX was not detectable in PC-3 (prostate cancer cells). Furthermore, it was shown that dual inhibition of LOX/COX enzymes (using 5,8,11,14- eicosatetraenoic acid (ETYA) and general inhibition of LOX enzymes (using Nordihydroguaiaretic acid (NDGA)) caused strong growth inhibition in SKBR3(breast cancer cell line), ZR75 (breast cancer cell line), T47D (breast cancer cell line), and COLO205 (colon cancer cell line) cells. While aspirin (ASA) as a general COX inhibitor, revealed little growth inhibitory effect on the mentioned cell lines indicating that the LOX inhibitors, unlike the COX inhibitors, were more putative in suppressing the growth of epithelial cancer cells regardless of the expression status of each enzyme [32]. To investigate the status of oxylipins in plasma of 20 patients with breast cancer and the age-matched healthy subjects, it was shown that the significant elevation 13-Hydroxyoctadecadienoic acid (13-HODE), 9-Hydroxyoctadecadienoic acid (9-HODE), 13S-hydroxy-9Z,11E,15Z-octadecatrienoic acid (13-HOTrE), 9-hydroxy-10E,12Z,15Z-octadecatrienoic acid (9-HOTrE), and 12S-hydroxy-5Z,8E,10E-heptadecatrienoic acid (12-HHTrE) was detected in the plasma of the breast cancer patients indicating that the LOX pathway metabolites from linoleic acid and linolenic acid metabolism are mainly up-regulated in breast cancer [33]. The metabolites of normal and tumor breast tissue of 27 patients were quantified using mass spectrometry. In accordance, it was shown that 13-HODE, 15-HETE, 12-Hydroxyeicosatetraenoic acid (12-HETE), 5-Hydroxyeicosatetraenoic acid (5-HETE), 5-Oxo-eicosatetraenoic acid (5-oxo-ETE), prostaglandin D2 (PGD2), and PGE2 metabolites were expressed in malignant and most of the normal breast tissues, while the only metabolite that its level was correlated with Mib1 scores, aggressive grade, and lymph node metastasis was 13-HODE. In contrast, a negative correlation was observed for PGE2 and PGD2 and aggressive features of breast tumors. To further determine the exact role of 13-HODE and 15-HETE on breast cancer cell growth, cells were exposed to different concentrations of these metabolites and it was shown that MDA-MB-231 and MCF-7 breast cancer cell number and proliferation was increased while PGD2 and PGE2 induced anti-proliferative effects in breast cancer cells. Also, no correlation was detected between the 13-HODE level and those of 15-HETE, 12-HETE, PGE2, or PGD2 in normal breast tissue, while the 13-HODE level was highly correlated with the 15-HETE level in tumors tissues of patients with 20 Mib1 scores [41]. To clarify the issue, it is worth mentioning that Mib1 is a cell proliferation marker that accounts for the tumor grading system based on summing the tumor differentiation, tumor necrosis, and the Mib1 score [42]. Taken together, activation of 15-LOX-1 and elevation of 13-HODE can stimulate breast cancer proliferation and invasion that possibly might shorten a patient’s survival rate [41]. The controversy was observed regarding the expression pattern of 15-LOX level in patients with breast cancer since the mRNA expression of 15-LOX was detected in breast cancer tumor tissues, however, the 15-LOX mRNA level was increased in tumor tissues of some patients and decreased in tumor tissues of other patients compared to the normal breast tissues [43]. Therefore, as with other aspects of tumor growth, the role of each 15-LOX isoform concerning breast cancer growth must be considered independently, given the origin of the tumor and the specific factors produced by the enzymes.

Table 1 The expression pattern of 15-LOX and its metabolites in breast cancer
Fig. 1
figure 1

The expression pattern of 15-LOX and its related metabolites in breast tumor and normal tissues, the blood of patients and healthy subjects, and breast cancer cell lines. The local and circulating level of 15-LOX enzymes and their main metabolites is different in patients with breast cancer and healthy subjects. The 15-LOX and its metabolite levels are shown in healthy subjects (The bottom right of the image) and patients with breast cancer (The bottom left of the image). The expression level of the 15-LOX and the metabolites in the serum and tissue samples of individuals is shown separately. The decreasing level of 15-LOX-1, − 2 transcripts, and 15-HETE in breast tumor tissue was accompanied by the elevated level of PPARγ [17, 21, 25]. Besides the increasing level of 15-HODE, 9-HODE, 13-HOTrE, 9-HOTrE, and 12-HHTre was apparent in breast cancer patients versus the elevated level of 15-HETE in serum of healthy subjects [26, 36, 40]. 15-LOX activation through exogenous 15-HETE and 13-HODE administration leads to breast cancer cell proliferation and invasion that is illustrated in breast cancer cells (Top left of the image) and breast normal cells (top right of the image) [25, 40]

What is the role of 15-LOX products on breast normal cell proliferation and breast tumor cell growth, metastasis, and drug resistance?

Tumor cell growth is not only dependent on increased cell proliferation but also decreased cell death or apoptosis [44]. In cancer cells, the onset of apoptosis depends on the imbalance between pro-apoptosis and anti-apoptosis proteins and the downstream signaling pathways, which often involve caspase or mitochondrial cascades [45]. Evidence from previous studies suggests that 15-LOX may be effective in inducing and promoting cell death in tumors. In accordance, it was shown that in a doxorubicin-resistant breast cell line, 15-LOX-1 was down-regulated remarkably at both mRNA and protein levels compared to the doxorubicin-sensitive cells. Interestingly, the level of 13(S)-HODE showed to be lower in doxorubicin-resistant cells [46]. Based on the results it seems that following acquired resistance to doxorubicin, the 15-LOX-1 mRNA became less stable and no change in a copy number of 15-LOX-1 has occurred. Moreover, inhibition of 15-LOX-1 by (pSUPER-shALOX) shRNA vector in drug-sensitive parental cells were not able to induce doxorubicin-resistant phenotype indicating that other signaling pathways besides 15-LOX-1 activity might be involved in the induction of drug resistance phenotype. Although 15-LOX-1 over-expression enhanced drug accumulation, cell motility, subG1 arrest and apoptosis induction and, caspase 3/7 increased activity in doxorubicin-resistant MCF-7 cells, the aforementioned results were not obtained following 15-LOX-1 over-expression in HeLa doxorubicin-resistant cells [46]. It seems that 15-LOX-1 can interfere with the cell fate and induce apoptosis in a cell-dependent manner. Interestingly, exogenous 13(S)-HODE induced apoptosis and cell cycle arrest in doxorubicin-resistant MCF-7 cells that might be due to the activation of PPAR-γ. The lack of PPAR-γ expression in HeLa doxorubicin-resistant cells might be involved in a poor response of these cells to the cytotoxic effect of 13(S)-HODE [46]. Tavakoli-Yaraki et al.; reported that Trichostatin A (TSA) induced cell death and apoptosis in MCF-7 and MDA-MB-231 cells which were accompanied by the induction of apoptosis, cell cycle arrest, and increase in the level of 13(S)-HODE. However, 15-LOX-1 inhibition using PD146176 as a specific, non-competitive 15-LOX inhibitor, attenuated the pro-apoptotic effect of TSA in breast cancer cells also it was shown that 13(S)-HODE synergized with TSA to inhibit breast cancer cell growth and apoptosis. Therefore, based on evidence, it is indicated that the pro-apoptotic effect of TSA on breast cancer cell growth might occur through activation of 15-LOX-1 and its metabolite [16]. Most studies on the molecular mechanisms underlying 15-LOX-1-induced apoptosis have been implemented on colorectal cancer and the results show that 15-LOX-1 expression level is decreased in patients with colorectal cancer [25]. Also, the results of experiments on colorectal tumor cells show that the promoter of the 15-LOX-1 gene is under the strict control of several processes, including epigenetic processes [47, 48]. The potential regulatory role of epigenetic processes in controlling 15-LOX expression in breast cancer as well as colorectal cancer has been investigated. In accordance, it was shown that sodium butyrate as a short-chain fatty acid that is suggested to have histone deacetylase inhibitory effects, can regulate the growth of some types of cancer cells such as breast and colon [49]. Based on the evidence provided by Tavakoli-Yaraki et al.; sodium butyrate stimulated the activity of 15-LOX-1 through elevation of 13(S)-HODE level and 15-LOX-1 transcript. Also, it was reported that the rate of apoptosis and the percentages of apoptotic cells induced by sodium butyrate in MCF-7 and MDA-MB-468 cells was abrogated following pretreatment of the cells by PD146176. The simultaneous exposure of MCF-7 and MDA-MB-468 cells to the 13(S)-HODE and sodium butyrate was more effective in the induction of breast cancer cell death [50]. Table 2 and Fig. 2 summarize the involvement of 15-LOX in breast cancer cell growth …

Table 2 The anti-proliferative effects of 15-LOXand its metabolites in breast cancer cells
Fig. 2
figure 2

The pro-apoptotic effects of 15-LOX 1 in breast cancer cells. 15-LOX may mediate the regulation of breast cancer cell death by inducing apoptosis. Activation of 15-LOX and exogenous 13-HODE administration induce apoptosis and cell cycle arrest in doxorubicin-resistant MCF-7 cells [32]. Also, 15-LOX-1 mediates pro-apoptotic effects of Trichostatin A in MCF-7 and MDA-MB-231 cells possibly through elevation of 13-HODE [9]. sodium butyrate induced apoptosis in breast cancer cells through activation of 15-LOX-1 activity and transcript level [43]

What is the interaction between COX and LOX enzymes with particular reference to 15-LOX?

As it is well established, improper regulation of the balance between cell proliferation and cell death results in tumor formation and is a hallmark of cancer [44]. Many cellular pathways interfere with the process of cell stagnation, apoptosis, or cell proliferation and it was revealed that LOX products cooperate with different growth factor signaling cascades to stimulate the growth of tumor cells [51]. It was reported that in nude mice under a high-fat diet enriched with linoleic acid, administration of indomethacin (A cyclooxygenase inhibitor) resulted in a higher concentration of prostaglandin E (PGE), 5-HETE, 12-HETE, and 15-HETE, which in tumors from the control group under high-fat diet enriched with linoleic acid (LA-enriched diet) than in those under diet contains low linoleic acid that was associated with reducing the risk of breast cancer progression in nude mice fed by LA-enriched diet (summarized in Fig. 3) [53]. In support of this evidence, it was shown that the LA-enriched diet can stimulate the MDA-MB-435 cells to produce COX and LOX products such as PGE2, 12-HETE, and 15-HETE which was associated with cell invasion. While, inhibition of 12-LOX, but not COX, can abolish eicosanoid metabolites secretion and cell invasion also stimulate the activity of metalloproteinase-9. It can be postulated that the effect of an LA-enriched diet on breast cancer cell growth can be at least partly due to the role of the lipoxygenase enzymes specially 12-LOX [54]. In addition to studies evaluating the role of 15-LOX in the animal model, several studies have examined the above mechanisms at the cellular level, which are demonstrated in Fig. 4. It was shown that inhibition of 15-HETE synthesis using docosahexaenoic acid administration was associated with reduced tumor cell growth and cell proliferation, increased apoptotic cells, and inhibition of tumor-induced angiogenesis [55] (Fig. 4). Also, it was shown that LA isomers suppress the uptake of linoleic acid, the content of cyclic adenosine monophosphate (cAMP) and the activity of extracellular signal-regulated protein kinase 1/2 (Erk1/2), and production of 13-HODE in steroid receptor-negative (SR2) MCF-7 cells possibly in a receptor-mediated manner. In addition, a small amount of 15(S)-HETE was produced in tumor tissue of MCF-7 (SR2) human breast xenografts. The combination of 13-HODE with linoleic acid isomers resulted in the elevation of cAMP level and emphasized the impact of 13-HODE on cAMP level (through activation of adenylate cyclase or suppression of phosphodiesterase) at a pharmacological level however the underlying mechanism needs to be clarified [56] (Fig. 4). It was shown that treatment of MDA-MB-231 cells with conjugated linoleic acid (CLA) (t10, c12-CLA), reduced the level of 15-HETE and 5-HETE. Also, incubation of proteins that were extracted from MDA-MB-231 cells with arachidonic acid, resulted in the production of 15- HETE, 12- HETE, and 5-HETE while simultaneous incubation of proteins with arachidonic acid and c9, t11-CLA or t10, c12-CLA did not affect the level of metabolite production. So, it can postulate that 5-, 12- and 15-LOX-2 enzymes have minimum effect on metabolizing c9, t11-CLA or t10, c12-CLA in breast cancer. Also, based on the evidence of this study 15-LOX-1 enzyme was not able to metabolize the aforementioned isomers while the linoleic acid is metabolized by 15-LOX-1 and produces 13-HODE [52]. In a study to investigate the impact of CLA supplements on arachidonic acid metabolites (15-, 12-, 5-HETE) and linoleic acid (13-, 9-HODE) levels during induction of breast tumors in rats, it was shown that the 5-HETE, 9-HODE, 12-HETE and 13-HODE levels were enhanced while the highest level belonged to the 5-HETE and 15-HETE in 7,12-dimethylbenz [a] anthracene (DMBA)-induced tumor group. While the 13-HODE level was dominated in the oil-DMBA-induced tumor group, however; its level was lower in the group treated with CLA- DMBA [57, 58]. It was shown that a high CLA diet in pregnant and breastfeeding female rats was associated with a lower risk of mammary tumor induced by chemicals in their offspring. It seems that the concentration of LOX metabolites was effective in this regard. Receiving the CLA supplements was associated with a higher concentration of 15-HETE in serum indicating that the LOX metabolites of arachidonic acid (15-, 12-, 5-HETE) and linoleic acid (HODE) were higher in mammary tumors in groups under the CLA diet. In accordance, there is a competition between CLA and PUFAs that might affect the PUFAs and their LOX metabolites concentrations [59]. It was shown that treatment of MDA-MB-435 breast cancer cells with exogenous arachidonic acid resulted in the production of 15(S)-HETE. While in the presence of NDGA (a LOX inhibitor), the exogenous arachidonic acid failed to produce 15(S)-HETE, and the phosphorylation of p38 and mitogen-activated protein kinase (MAPK) was inhibited (Fig. 4). Interestingly the protein level of 15-LOX-2 but not 15-LOX-1 was increased in MDA-MB-435 cells suggesting that 15-LOX-2 was responsible to metabolize exogenous arachidonic acid to 15(S)-HETE. Also, treatment of MDA-MB-435 cells with exogenous 15(S)-HETE resulted in an activation of p38 and MAPK pathway and cell adhesion to type IV collagen indicating the pushing role of 15(S)-HETE produced by 15-LOX-2 in activation of signaling pathways related to the cell adhesion to the extracellular matrix [60]. Interestingly it was shown that the circulating level of eicosapentaenoic acid (EPA) inhibits the proliferation of human MCF-7 xenografts that might be mediated by reduction of linoleic acid uptake and 13-HODE formation. The lower level of 13-HODE was accompanied by a decrease in cAMP and ERK1/2 phosphorylation in MCF-7 xenografts. In the presence of exogenous 13-HODE to the EPA-containing arterial blood, the uptake of 13-HODE by tumors enhanced, the tumor DNA content increased, and the phosphorylated ERK1/2 was restored [61]. In a study on MCF-7 human breast xenografts, a perfusion system was designed and the tumors were perfused with rat blood that was supplemented with melatonin (MLT), EPA, and CLA promptly and the tumor uptake of linoleic acid and its conversion to 13-HODE and the subsequent release to the blood circulation were monitored. It was revealed that the linoleic acid uptake and 13-HODE generation was suppressed and the phosphorylation of ERK1/2 was significantly reduced following receiving anticancer agents suggesting the association of 13-HODE release rate and ERK1/2 phosphorylation [62]. Also, it was revealed that the peanut oil-enriched diet which is rich in ω-3 and ω9 PUFAs caused elevation of membrane arachidonic acid content and reduced activity of 15-LOX-2 and 15-LOX-1 that was accompanied by increased apoptosis and decrease mitosis in murine mammary gland adenocarcinoma (Fig. 3). The reduced tumor volume, number of metastasis, and longer survival were detected in the mice group under a peanut oil-enriched diet. In support of this, it was proposed that the reduced severe features of tumors were related to the suppression of LOXs activity and decrease of pro-tumorigenic eicosanoids such as 15 (S)-HETE [63]. Tables 3 and 4 summarize the tumorigenic effect of 15-LOX and its products in breast cancer tissue and cells.

Fig. 3
figure 3

The effect of Linoleic acid-rich diet on 15-LOX activity and breast tumor behavior (In-vivo evidence). Changes in the activity of enzymes involved in fat metabolism can affect the fate of tumor cells. LA-enriched diet supplemented with indomethacin-induced 15-LOX metabolites and reduced breast tumor size in nude mice [45]. The peanut oil-enriched diet enriched by ω-3 and ω9 PUFAs caused elevation of membrane arachidonic acid (AA) content and reduced activity of 15-LOX-2 and 15-LOX-1 that was accompanied by increased apoptosis and decrease mitosis in murine mammary gland adenocarcinoma [52]. The conjugated linoleic acid (CLA) in pregnant and breastfeeding female rats was associated with reduced LOX metabolites and enzyme activity lower risk of mammary tumor size induced by chemicals in their offspring [51, 53]

Fig. 4
figure 4

The effect of exogenous lipid metabolism modulation on breast cancer cell behavior and 15-LOX metabolites (In-vitro evidence). Various compounds that target the metabolism of fatty acids could be effective in the growth and invasion of breast tumor cells through lipoxygenase metabolites. Docosahexaenoic acid reduced tumor cell growth, cell proliferation, tumor-induced angiogenesis through inhibition of 15-HETE synthesis [47]. The cAMP content, activity of Erk1/2 and 13-HODE synthesis were suppressed following linoleic acid isomers administration [48]. The conjugated linoleic acid (CLA) reduced the level of 15-HETE and 5-HETE while exogenous arachidonic acid stimulated production of 15(S)-HETE and phosphorylation of p38 MAPK [54]. The linoleic acid-rich supplement stimulates the production of PGE2, 12-HETE, and 15-HETE which was associated with tumor cell invasion [46]

Table 3 The involvement of 15-LOX and its metabolites in the pro-tumorigenic effects of various conjugated lipid-based diets in breast cancer; in vivo evidences
Table 4 The involvement of 15-LOX and its metabolites in the pro-tumorigenic effects of various conjugated lipid-based diets in breast cancer; in vitro evidences

How anti-cancer drugs act on 15-LOX

Although it may seem a bit off-topic, interesting studies have been found on the relationship between melatonin, and 13-HODE in breast cancer. It was shown that in human MCF-7 breast cancer xenografts, exposure to constant light stimulates tumor tissue growth. Interestingly, the LA uptake and 13-HODE production in the group that was constantly exposed to the light was increased compared to the control group. The constant light exposure-induced tumor growth can be explained by the suppression of melatonin (MLT) synthesis, increased linoleic acid uptake, and production of 13-HODE [64]. Based on multiple lines of evidence, neurohormone MLT suppresses several human tumor cell growth and metabolism. It seems that the anticancer effect of melatonin occurs in a receptor-mediated manner through inhibition of linoleic acid uptake by tumor cells. It is well documented that linoleic acid is a fatty acid that promotes tumor growth and converts to the 13-HODE which has a mitogenic effect. The perfusion of MCF-7 human breast xenografts by melatonin in different time schedules resulted in suppression of linoleic acid uptake and 13-HODE generation by tumors and subsequently decrease the ERK1/2 phosphorylation [62]. Following inhibition of melatonin in dim light at night, the balance among various preventive signaling pathways in cancer is disrupted and resulted in hyperglycemia, hyperinsulinemia, and proliferative activity in the tumor. In addition, during exposure to the light the fatty acid, and linoleic acid uptake, 13-HODE formation, cAMP level, glucose uptake and lactate production increased that was abrogated when exposed to the dark. It was revealed that the control of tumor cell metabolism, aerobic glycolysis, and growth was dependent on the melatonin-induced inhibition of 13-HODE formation and subsequently the activation of protein kinase B (AKT) by 13-HODE. Interestingly, melatonin itself was able to down-regulate phospho-Akt (Ser473) (pAKTs473) and reduce the activity of AKT. The observed effects were diminished in the presence of MT1/MT2 melatonin receptor antagonist S20928. It was concluded that melatonin suppresses linoleic acid uptake, 13-HODE formation, and the Warburg effect in human breast cancer xenografts and 13-HODE play signaling like to activate Warburg effect by AKT activation [65].

How interaction between breast tumor cell-normal cell-tumor microenvironment is influenced by 15-LOX? (The consequent impact on breast tumor cell growth)

Studies of the last two decades have shown that there are several factors involved in the onset and progression of breast cancer [40]. A considerable number of breast cancer patients experience tumor metastasis and tumor recurrence that leads to premature death. Therefore, recognizing the factors involved in initiating tumor metastasis and the underlying mechanism is helpful in clinical decision-making [66]. Regarding the relevance of the 15-LOX pathway in breast cancer metastasis, it was revealed that the 15-LOX-1 gene induced in MCF-7 cell spheroids when compared with endothelial monolayers. Following enzyme activity inhibition using NDGA, the MCF-7 spheroid–induced circular defects in lymphatic endothelial cell monolayers were remarkably decreased in a dose-dependent manner (Fig. 5). In addition, knockdown of 15-LOX-1 activity using shRNA caused suppression of circular defects formation as well as metastasis to the lymph node in breast xenografted tumors. Based on this study, 15-LOX-1 is involved in mediating tumor cell invasion and lymph node metastasis in breast carcinoma [68]. In support of this, simultaneous inhibition of 15-LOX-1 using baicalein and nuclear factor kappa B (NF-κB) using Bay11–7082 (an inhibitor of κB kinase (IKK)) caused inhibition of circular chemorepellent-induced defects’ (CCID) formation in MCF-7 cell spheroids. While attaching to the lymph-endothelial cell monolayers, CCID is considered as a tumor intravasation process and it seems that generation of 12(S)-HETE by 15-LOX-1 under NF-κB regulation can facilitate CCID and attachment of breast cancer cells to the lymph-endothelial cells [67]. In addition, exposure of breast cancer cells to the EPA resulted in an increased expression level of E-cadherin, while treatment of the cells with gamma linoleic acid (GLA) did not affect E-cadherin level. However, both GLA or EPA treatments caused elevation of 15(S)-HETE and 13(S)-HODE level in MCF-7 cells, and a higher level of 13(S)-HODE was observed when compared to 15(S)-HETE although the reduced expression of E-cadherin in breast cells seemed to be dependent to the ratio of 15-LOX-1 metabolites [69]. The proteome study in myeloid-derived suppressor cells (MDSCs) in response to metastatic breast tumors, demonstrated that in MDSCs the selective expression of lipid metabolic pathway occurs. It was found that in MDSCs from metastatic 4 T1 tumors, the 15-LOX-1 was a positive activator of cAMP Response Element-Binding Protein (CREB) which is a transcription factor and is involved in the biological process in MDSCs [70]. In another study, it was shown that while bovine mammary endothelial cells (BMEC) were exposed to Streptococcus uberis no effect was detected on 15-LOX-1 metabolite levels, while exposure of bovine monocytes to Streptococcus uberis resulted in the production of 13-Hydroperoxyoctadeca-cis-9, trans-11-dienoic acid (13-HPODE) and 13-HODE during mastitis. Interestingly, treatment of BMEC with 13-HPODE reduced endothelial barrier integrity and accelerate apoptosis while co-treatment with antioxidants, reverses the previously seen effects [71]. It was found that 15-LOX-1 suppression using novel purine-pyrazole hybrids which contain thiazoles, thiazolidinones and rhodanines reduced the rate of viable MCF-7 cells and proposed to act as an anticancer compound in breast cancer [34, 72, 73]. It was shown that the exogenous arachidonic acid was metabolized in TMT-081 rat mammary tumor cell line and the range of eicosanoids metabolites including lipoxygenase and cyclooxygenase metabolites were produced. In accordance, it was observed that the DNA synthesis was stimulated in response to the 15-HETE and LOX inhibition using NDGA and esculetin that resulted in TMT-081 cell growth suppression [74]. It was shown that incubation of BT-20 cells (triple-negative breast cancer cells) with Transforming growth factor-alpha (TGF-α) and A23187, a mobile ion-carrier, caused 13-HODE formation which was dependent on the Epidermal growth factor (EGF) / TGF-α. While by suppression LOX activity, the TGF-α-dependent generation of 13-HODE and stimulation of DNA synthesis was abrogated. The findings indicated that LOX activity and 13-HODE production are involved in transferring the mitogenic signals that transferred from EGF/TGF-α in the cell surface to the cell nucleus in breast cancer [75]. The relation of 15-LOX and its products with breast cancer metastasis and invasion is summarized in Table 5.

Fig. 5
figure 5

The effect of 15-LOX and its mediators on breast cancer cell metastasis and invasion. The 15-LOX activity can affect breast cancer cell metastasis and invasion through various pathways. Inhibition of 15-LOX activity leads to reduced MCF7 spheroid formation and metastasis to the lymph node [61]. The MCF-7 cell exposure to EPA and GLA induced the production of 15(S)-HETE and 13(S)-HODE [63]. 15-LOX-1accounts as a positive activator of CREB that mediates breast cancer cell metastasis [64]. 15-LOX-1 suppression using novel purine-pyrazole hybrids induced MCF-7 cell death, however; 15-LOX inhibition using NDGA caused TMT-081 cell growth suppression, reduced DNA synthesis, and 13-HODE formation [40, 64, 67]. The attachment of breast cancer cells to the lymph-endothelial cells facilitates using 12(S)-HETE generation by 15-LOX-1 [61, 62]

Table 5 The effects of 15-LOX and its metabolites in breast cancer metastasis and invasion

How are 15-LOX and oxidative stress connected to regulate breast cancer cell growth?

It is taken for granted that increased lipid peroxidation and oxidative stress account as crucial risk factors for breast cancer incidence, development, and progression [76]. On the other hand, tumor cell fate, growth, invasion and, death are influenced by lipid metabolism, peroxidation, and level of saturation [77]. As a consequence of aerobic metabolism, reactive oxygen species (ROS) are continually generated and transformed in the normal and tumor cells that are involved in various cell functions including cell proliferation and death. Therefore, maintaining redox balance by increasing antioxidant capacity and reducing the production of ROS is essential for the survival of tumor cells [78]. In support of this, the catalytic activity of LOX enzymes leads to the production of lipid peroxides that are involved in different cell signaling events regulating tumor cell growth or death. Notably, overproduction of lipid peroxides alongside iron accumulation can trigger ferroptosis which accounts for a type of programmed cell death [79]. It was shown that upon ROS stress, activation of 12-LOX induced ferroptosis in tumor cells through overproduction of lipid peroxides [7]. The regulatory role of 15-LOX in inducing the above cell death seems more important and interesting. It was shown that glutathione peroxidase 4 (GPX4) as a protective enzyme against lipid peroxidation as well as 15-LOX can induce nuclear factor erythroid-2 like 2 (NFE2L2) (Nrf2) as a regulator of antioxidant response in tumor cells that suppress ferroptosis and contributes to tumor cell chemoresistance [79]. Although the role of 15-LOX and its metabolites in stimulating various intracellular pathways leading to the regulation of oxidative stress and tumor cell fate can be extensive, the evidence in breast cancer is very limited. In the most relevant study, Xinghan Wu et all demonstrated that inhibition of Glycogen synthase kinase-3β (GSK-3β) resulted in decreased ROS and malondialdehyde (MDA) level through activation of GPX4 and inhibition of 15-LOX that blocked ferroptosis in MCF-7, MDA-MB-231 breast cancer cells. It was shown that GSK-3β is a positive regulatory enzyme to promote cancer cell proliferation and survival and based on shreds of evidence, 15-LOX might be served by GSK-3β as a downstream mediator to regulate feroptosis in breast tumor cells [80]. On the other hand, it was shown that sodium butyrate as a short-chain fatty acid-induced 15-LOX gene expression and activity in breast cancer cells through elevation of 13-HODE [50]. The ability of sodium butyrate to trigger cell cycle arrest and apoptosis through the generation of ROS and reduced mitochondrial membrane potential (Δψm) in breast cancer cells [81] and its positive regulatory effect on 15-LOX may suggest that 15-LOX is contributed to the production of ROS in sodium butyrate-elicited apoptosis (Data is illustrated in Fig. 6). However, the significance of 15-LOX in the context of oxidative stress and redox biology in breast cancer pathogenesis is still emerging and needs to be clarified by more mechanistic studies.

Fig. 6
figure 6

The relevance of 15-LOX and the oxidative stress in breast cancer cell growth. The reduction in the expression level of 15-LOX-1 is associated with a decrease in the level of ROS and reduced breast cancer cell death [74]. Also, 15-LOX-1 activation-induced breast cancer cell apoptosis might occur through the generation of ROS [75]

What are the clinical implications of 15-LOX inhibition or activation in breast cancer treatment?

The use of small molecular inhibitors that can inhibit key enzymes in the metabolic pathway is one of the main and important strategies in targeted cancer therapy [82]. Notably, interesting studies have been performed on small molecular compounds capable of inhibiting the 15-LOX, which can be considered as potent chemopreventive compounds in cancer prevention and treatment [83]. One of the well-documented ones is quercetin, which is a natural flavone with the ability to regulate cell cycle distribution, apoptosis, angiogenesis, and 15-LOX inhibitory activity [84]. It is demonstrated that quercetin sensitizes breast cancer cells to death and inhibits angiogenesis and EMT process, however, the efficiency of the quercetin-rich diet in breast cancer treatment needs to be clarified by further clinical trials [85]. Moreover, it was revealed that rhein and aloe-emodin, as polymethoxylated flavonoids which are extracted from orange peel inhibited soybean 15-LOX potently as well as breast cancer angiogenesis and growth [86]. On the other hand, Δ9 –tetrahydrocannabinol (Δ9-THC) [87], as an active component of Cannabis Sativa (C. sativa) and a member of phytocannabinoid family, can directly inhibit 15-LOX and suppress breast cancer cell proliferation [88]. The selective inhibitory effect of other derivatives of phytocannabinoids such as cannabidiol (CBD) on 15-LOX activity has been proven previously and in-vivo evidence revealed the potency of CBD to reduce breast cancer growth [88, 89]. The ortho-hydroxy analogs of pyrazoles have been shown to have an inhibitory effect on 15-LOX also the potential to inhibit breast cancer cell proliferation [90]. Taken together, the mentioned 15-LOX inhibitors showed anti-proliferative and cytotoxic effects on breast cancer cells through suppression of 15-LOX activity, however, the therapeutic use of these compounds in humans has not yet been addressed and requires further investigation at the clinical trial level. Evidence also has shown that 15-LOX-1 over-expression as well as exposure to the exogenous 13(S)-HODE enhanced drug accumulation, cell motility, subG1 arrest, apoptosis induction and, caspase 3/7 increased activity in doxorubicin-resistant MCF-7 cells, indicating the possibility of 15-LOX pathway involvement in the response of breast cancer cells to the doxorubicin treatment [46].

Conclusion

This review attempts to highlight the expression pattern of 15-LOX enzymes and their metabolites in breast tumors and normal tissues as well as their significance in breast cancer cell growth, death, invasion, and metastasis. Studies show the dual role of the LOX pathway in the pathogenesis of breast cancer. Both decreased and increased expression of LOX enzymes were shown in breast tumor tissue compared to healthy breast tissues. Among the studies performed, it seems that 15-LOX-2 and 15-S-HETE showed a significant reduction in breast tumor tissue, which was adjusted by PPAR-γ activity. Based on the study of Hong et al., the expression pattern of 15-LOX isomers and metabolites in different types of breast cancer cells can be different which can explain the differential expression of these enzymes in different patients with breast cancer. Since in the patients studied more attention was paid to the degree of tumor severity, and disease severity and the molecular nature of breast tumor cells in these patients was not considered. On the other hand, the LOX enzymatic pathway is an important metabolic pathway for the normal life of healthy cells that can be affected by different conditions of growth, nutrition, and overall homeostasis of cells. Most of the results regarding the role of 15-LOX in inducing apoptosis in breast cancer cells indicated the mediating role of this enzyme or the synergistic effect of the metabolites of 15-LOX in inducing death pathways by other compounds. However, there was stronger evidence for the tumorigenic role of the 15-LOX pathway in breast tumor progression. It can be postulated that 15-LOX metabolites regulate breast tumor cell growth through elevation of cAMP level, phosphorylation of p38- MAPK, phosphorylation of ERK1/2, and increased tumor DNA content; however, the type of fat diet that the cells were exposed to, was decisive in the type of response they exhibited. It seems that 15-LOX-1 and its metabolites (15(S)-HETE 13(S)-HODE) were effective in breast cancer cell spheroids formation, metastasis to the lymph node, CREB activation, and transferring the mitogenic signals of TGF-α. Taken together, the relevance of 15-LOX enzymes and metabolites in breast tumor cell growth has been proven broadly; however, the exact role of this pathway on breast cancer pathogenesis remains to be determined, and future studies are warranted.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

ALA:

Alpha-linolenic acid

LA:

Linoleic acid

DHA:

Docosahexaenoic acid

AA:

Arachidonic acid (AA)

COX:

Cyclooxygenase

PG:

Prostaglandin

PGE:

Prostaglandin E

TX:

Thromboxane

TXA2:

Thromboxane A2

CYP:

Cytochrome P450

EETs:

Epoxyeicosatrienoic acids

LT:

Leukotrienes

LOX:

lipoxygenase

HpETE:

Hydroperoxyeicosatetraenoic acid

HETE:

Hydroxyeicosatetraenoic acid

13-HODE:

13-Hydroxyoctadecadienoic acid

15-LOX:

15-lipoxygenase-15

15-LOX-1:

15-lipoxygenase type 1

15-LOX-2:

15- lipoxygenase type 2

A549 cell line:

Adenocarcinoma human alveolar basal epithelial cells

Caco2 cell line:

Human colorectal adenocarcinoma cells

IL4:

Interleukin 4

IL13:

Interleukin-13

STAT6:

Signal transducer and activator of transcription 6

NO:

Nitric oxide

15-S-HETE:

15(S)-hydroperoxyeicosatetraenoic acid

13(S)-HODE:

3(S)-hydroxy-9Z,11E-octadecadienoic acid

NSCLC:

Non-Small Cell Lung Cancer

COX-2:

Cyclooxygenase-2

CK19:

Cytokeratin 19

ER:

Estrogen receptor

PPAR-γ:

Peroxisome proliferator- activated receptor gamma

PGE2:

Prostaglandin E2

6-k-PGF1α:

6-keto-Prostaglandin F1 Alpha

PGF :

prostaglandin F2α

HHT:

Heptadecatrienoic acid

15-HETE:

15-Hydroxyeicosatetraenoic acid

DHKPG:

13,14-dihydro-l5-keto-prostaglandin

15-L-(S)-HPETE:

15-L-(S)-hydroperoxyeicosatetraenoic acid

COX-1:

Cyclooxygenase-

5-LOX:

5-lipoxygenase

FLAP:

5-lipoxygenase activating protein

12-LOX:

12-lipoxygenase

ETYA:

5,8,11,14-eicosatetraynoic acid

NDGA:

Nordihydroguaiaretic acid

ASA:

Aspirin

9-HODE:

9-Hydroxyoctadecadienoic acid

13-HOTrE:

13S-hydroxy-9Z,11E,15Z-octadecatrienoic acid

9-HOTrE:

9-hydroxy-10E,12Z,15Z-octadecatrienoic acid

12-HHTrE:

12S-hydroxy-5Z,8E,10E-heptadecatrienoic acid

12-HETE:

12-Hydroxyeicosatetraenoic acid

5-HETE:

5-Hydroxyeicosatetraenoic acid

5-oxo-ETE:

5-Oxo-eicosatetraenoic acid

TSA:

Trichostatin A

LA-enriched diet:

High-fat diet enriched with linoleic acid

cAMP:

Cyclic adenosine monophosphate

Erk1/2:

Extracellular signal-regulated protein kinase 1/2

SR2:

Steroid receptor negative

CLA:

Conjugated linoleic acid

DMBA:

7,12-dimethylbenz [a]anthracene

PUFAs:

Polyunsaturated fatty acids

MAPK:

Mitogen-activated protein kinase

EPA:

Eicosapentaenoic acid

MLT:

Melatonin

AKT:

Protein kinase B

pAKTs473:

Phospho-Akt (Ser473)

NF-κB:

Nuclear factor kappa B

CCID:

Chemo repellent-induced defects

GLA:

Gamma linoleic acid

MDSCs:

Myeloid derived suppressor cells

CREB:

cAMP Response Element-Binding Protein

BMEC:

Bovine mammary endothelial cells

13-HPODE:

13-Hydroperoxyoctadeca-cis-9, trans-11-dienoic acid

TGF-α:

Transforming growth factor alpha

EGF:

Epidermal growth factor

ROS:

Reactive oxygen species

GPX4:

Glutathione peroxidase 4 (GPX4)

NFE2L2:

nuclear factor erythroid-2 like 2

GSK-3β:

Glycogen synthase kinase-3β

MDA:

Malondialdehyde

Δψm:

mitochondrial membrane potential

Δ9-THC:

Δ9 –tetrahydrocannabinol

C. sativa :

Cannabis Sativa

CBD:

Cannabidiol

References

  1. Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, et al. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther. 2021;6(1):94. https://doi.org/10.1038/s41392-020-00443-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Dubois RN, Abramson SB, Crofford L, Gupta RA, Simon LS, Van De Putte LB, et al. Cyclooxygenase in biology and disease. FASEB J. 1998;12(12):1063–73. https://doi.org/10.1096/fasebj.12.12.1063.

    Article  CAS  PubMed  Google Scholar 

  3. Samuelsson B. Prostaglandins, thromboxanes, and leukotrienes: formation and biological roles. Harvey Lect. 1979;75:1–40.

    PubMed  Google Scholar 

  4. Rizos AL, Sargenti CJ, Jeste DV. Psychotropic drug interactions in the patient with late-onset depression or psychosis. Part 2. Psychiatr Clin North Am. 1988;11(1):253–77. https://doi.org/10.1016/S0193-953X(18)30527-6.

    Article  CAS  PubMed  Google Scholar 

  5. Capdevila J, Marnett LJ, Chacos N, Prough RA, Estabrook RW. Cytochrome P-450-dependent oxygenation of arachidonic acid to hydroxyicosatetraenoic acids. Proc Natl Acad Sci U S A. 1982;79(3):767–70. https://doi.org/10.1073/pnas.79.3.767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Xu X, Zhang XA, Wang DW. The roles of CYP450 epoxygenases and metabolites, epoxyeicosatrienoic acids, in cardiovascular and malignant diseases. Adv Drug Deliv Rev. 2011;63(8):597–609. https://doi.org/10.1016/j.addr.2011.03.006.

    Article  CAS  PubMed  Google Scholar 

  7. Zheng Z, Li Y, Jin G, Huang T, Zou M, Duan S. The biological role of arachidonic acid 12-lipoxygenase (ALOX12) in various human diseases. Biomed Pharmacother. 2020;129:110354. https://doi.org/10.1016/j.biopha.2020.110354.

    Article  CAS  PubMed  Google Scholar 

  8. Andreou A, Feussner I. Lipoxygenases - Structure and reaction mechanism. Phytochemistry. 2009;70(13-14):1504–10. https://doi.org/10.1016/j.phytochem.2009.05.008.

    Article  CAS  PubMed  Google Scholar 

  9. Liavonchanka A, Feussner I. Lipoxygenases: occurrence, functions and catalysis. J Plant Physiol. 2006;163(3):348–57. https://doi.org/10.1016/j.jplph.2005.11.006.

    Article  CAS  PubMed  Google Scholar 

  10. Feussner I, Wasternack C. The lipoxygenase pathway. Annu Rev Plant Biol. 2002;53(1):275–97. https://doi.org/10.1146/annurev.arplant.53.100301.135248.

    Article  CAS  PubMed  Google Scholar 

  11. Brinckmann R, Topp MS, Zalán I, Heydeck D, Ludwig P, Kühn H, et al. Regulation of 15-lipoxygenase expression in lung epithelial cells by interleukin-4. Biochem J. 1996;318(Pt 1):305–12. https://doi.org/10.1042/bj3180305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Heydeck D, Thomas L, Schnurr K, Trebus F, Thierfelder WE, Ihle JN, et al. Interleukin-4 and -13 induce upregulation of the murine macrophage 12/15-lipoxygenase activity: evidence for the involvement of transcription factor STAT6. Blood. 1998;92(7):2503–10. https://doi.org/10.1182/blood.V92.7.2503.

    Article  CAS  PubMed  Google Scholar 

  13. Zuo X, Morris JS, Shureiqi I. Chromatin modification requirements for 15-lipoxygenase-1 transcriptional reactivation in colon cancer cells. J Biol Chem. 2008;283(46):31341–7. https://doi.org/10.1074/jbc.M803729200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hsi LC, Kundu S, Palomo J, Xu B, Ficco R, Vogelbaum MA, et al. Silencing IL-13Rα2 promotes glioblastoma cell death via endogenous signaling. Mol Cancer Ther. 2011;10(7):1149–60. https://doi.org/10.1158/1535-7163.MCT-10-1064.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dhabal S, Das P, Biswas P, Kumari P, Yakubenko VP, Kundu S, et al. Regulation of monoamine oxidase A (MAO-A) expression, activity, and function in IL-13-stimulated monocytes and A549 lung carcinoma cells. J Biol Chem. 2018;293(36):14040–64. https://doi.org/10.1074/jbc.RA118.002321.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tavakoli-Yaraki M, Karami-Tehrani F, Salimi V, Sirati-Sabet M. Induction of apoptosis by Trichostatin A in human breast cancer cell lines: involvement of 15-Lox-1. Tumour Biol. 2013;34(1):241–9. https://doi.org/10.1007/s13277-012-0544-7.

    Article  CAS  PubMed  Google Scholar 

  17. O'Donnell VB, Taylor KB, Parthasarathy S, Kühn H, Koesling D, Friebe A, et al. 15-Lipoxygenase catalytically consumes nitric oxide and impairs activation of guanylate cyclase. J Biol Chem. 1999;274(29):20083–91. https://doi.org/10.1074/jbc.274.29.20083.

    Article  CAS  PubMed  Google Scholar 

  18. Brinckmann R, Schnurr K, Heydeck D, Rosenbach T, Kolde G, Kühn H. Membrane translocation of 15-lipoxygenase in hematopoietic cells is calcium-dependent and activates the oxygenase activity of the enzyme. Blood. 1998;91(1):64–74. https://doi.org/10.1182/blood.V91.1.64.

    Article  CAS  PubMed  Google Scholar 

  19. Tang DG, Bhatia B, Tang S, Schneider-Broussard R. 15-lipoxygenase 2 (15-LOX2) is a functional tumor suppressor that regulates human prostate epithelial cell differentiation, senescence, and growth (size). Prostaglandins Other Lipid Mediat. 2007;82(1-4):135–46. https://doi.org/10.1016/j.prostaglandins.2006.05.022.

    Article  CAS  PubMed  Google Scholar 

  20. Roy B, Cathcart MK. Induction of 15-lipoxygenase expression by IL-13 requires tyrosine phosphorylation of Jak2 and Tyk2 in human monocytes. J Biol Chem. 1998;273(48):32023–9. https://doi.org/10.1074/jbc.273.48.32023.

    Article  CAS  PubMed  Google Scholar 

  21. Rademacher M, Kuhn H, Borchert A. Systemic deficiency of mouse arachidonate 15-lipoxygenase induces defective erythropoiesis and transgenic expression of the human enzyme rescues this phenotype. FASEB J. 2020;34(11):14318–35. https://doi.org/10.1096/fj.202000408RR.

    Article  CAS  PubMed  Google Scholar 

  22. Uderhardt S, Krönke G. 12/15-lipoxygenase during the regulation of inflammation, immunity, and self-tolerance. J Mol Med (Berl). 2012;90(11):1247–56. https://doi.org/10.1007/s00109-012-0954-4.

    Article  CAS  Google Scholar 

  23. Philips BJ, Dhir R, Hutzley J, Sen M, Kelavkar UP. Polyunsaturated fatty acid metabolizing 15-Lipoxygenase-1 (15-LO-1) expression in normal and tumorigenic human bladder tissues. Appl Immunohistochem Mol Morphol. 2008;16(2):159–64. https://doi.org/10.1097/PAI.0b013e31805baa41.

    Article  CAS  PubMed  Google Scholar 

  24. Jiang WG, Watkins G, Douglas-Jones A, Mansel RE. Reduction of isoforms of 15-lipoxygenase (15-LOX)-1 and 15-LOX-2 in human breast cancer. Prostaglandins Leukot Essent Fatty Acids. 2006;74(4):235–45. https://doi.org/10.1016/j.plefa.2006.01.009.

    Article  CAS  PubMed  Google Scholar 

  25. Il Lee S, Zuo X, Shureiqi I. 15-Lipoxygenase-1 as a tumor suppressor gene in colon cancer: is the verdict in? Cancer Metastasis Rev. 2011;30(3-4):481–91. https://doi.org/10.1007/s10555-011-9321-0.

    Article  CAS  PubMed  Google Scholar 

  26. Gonzalez AL, Roberts RL, Massion PP, Olson SJ, Shyr Y, Shappell SB. 15-Lipoxygenase-2 expression in benign and neoplastic lung: an immunohistochemical study and correlation with tumor grade and proliferation. Hum Pathol. 2004;35(7):840–9. https://doi.org/10.1016/j.humpath.2004.04.001.

    Article  CAS  PubMed  Google Scholar 

  27. Hennig R, Kehl T, Noor S, Ding XZ, Rao SM, Bergmann F, et al. 15-lipoxygenase-1 production is lost in pancreatic cancer and overexpression of the gene inhibits tumor cell growth. Neoplasia. 2007;9(11):917–26. https://doi.org/10.1593/neo.07565.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang WG, Douglas-Jones A, Mansel RE. Levels of expression of lipoxygenases and cyclooxygenase-2 in human breast cancer. Prostaglandins Leukot Essent Fatty Acids. 2003;69(4):275–81. https://doi.org/10.1016/S0952-3278(03)00110-8.

    Article  CAS  PubMed  Google Scholar 

  29. Nie D. Cyclooxygenases and lipoxygenases in prostate and breast cancers. Front Biosci. 2007;12(1):1574–85. https://doi.org/10.2741/2170.

    Article  CAS  PubMed  Google Scholar 

  30. Subbarayan V, Xu XC, Kim J, Yang P, Hoque A, Sabichi AL, et al. Inverse relationship between 15-lipoxygenase-2 and PPAR-gamma gene expression in normal epithelia compared with tumor epithelia. Neoplasia. 2005;7(3):280–93. https://doi.org/10.1593/neo.04457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kort WJ, Bijma AM, van Dam JJ, van der Ham AC, Hekking JM, van der Ingh HF, et al. Eicosanoids in breast cancer patients before and after mastectomy. Prostaglandins Leukot Essent Fatty Acids. 1992;45(4):319–27. https://doi.org/10.1016/0952-3278(92)90090-6.

    Article  CAS  PubMed  Google Scholar 

  32. Hong SH, Avis I, Vos MD, Martínez A, Treston AM, Mulshine JL. Relationship of arachidonic acid metabolizing enzyme expression in epithelial cancer cell lines to the growth effect of selective biochemical inhibitors. Cancer Res. 1999;59(9):2223–8.

    CAS  PubMed  Google Scholar 

  33. Chocholoušková M, Jirásko R, Vrána D, Gatěk J, Melichar B, Holčapek M. Reversed phase UHPLC/ESI-MS determination of oxylipins in human plasma: a case study of female breast cancer. Anal Bioanal Chem. 2019;411(6):1239–51. https://doi.org/10.1007/s00216-018-1556-y.

    Article  CAS  PubMed  Google Scholar 

  34. Afifi OS, Shaaban OG, Abd El Razik HA, SEA SE-D, Ashour FA, El-Tombary AA, et al. Synthesis and biological evaluation of purine-pyrazole hybrids incorporating thiazole, thiazolidinone or rhodanine moiety as 15-LOX inhibitors endowed with anticancer and antioxidant potential. Bioorg Chem. 2019;87:821–37. https://doi.org/10.1016/j.bioorg.2019.03.076.

    Article  CAS  PubMed  Google Scholar 

  35. Tang S, Bhatia B, Maldonado CJ, Yang P, Newman RA, Liu J, et al. Evidence that arachidonate 15-lipoxygenase 2 is a negative cell cycle regulator in normal prostate epithelial cells. J Biol Chem. 2002;277(18):16189–201. https://doi.org/10.1074/jbc.M111936200.

    Article  CAS  PubMed  Google Scholar 

  36. Bhatia B, Maldonado CJ, Tang S, Chandra D, Klein RD, Chopra D, et al. Subcellular localization and tumor-suppressive functions of 15-lipoxygenase 2 (15-LOX2) and its splice variants. J Biol Chem. 2003;278(27):25091–100. https://doi.org/10.1074/jbc.M301920200.

    Article  CAS  PubMed  Google Scholar 

  37. Tavakoli Yaraki M, Karami Tehrani F. Apoptosis Induced by 13-S-hydroxyoctadecadienoic acid in the Breast Cancer Cell Lines, MCF-7 and MDA-MB-231. Iran J Basic Med Sci. 2013;16(4):653–9.

    PubMed  PubMed Central  Google Scholar 

  38. Najid A, Beneytout JL, Tixier M. Cytotoxicity of arachidonic acid and of its lipoxygenase metabolite 15-hydroperoxyeicosatetraenoic acid on human breast cancer MCF-7 cells in culture. Cancer Lett. 1989;46(2):137–41. https://doi.org/10.1016/0304-3835(89)90021-9.

    Article  CAS  PubMed  Google Scholar 

  39. Harats D, Ben-Shushan D, Cohen H, Gonen A, Barshack I, Goldberg I, et al. Inhibition of carcinogenesis in transgenic mouse models over-expressing 15-lipoxygenase in the vascular wall under the control of murine preproendothelin-1 promoter. Cancer Lett. 2005;229(1):127–34. https://doi.org/10.1016/j.canlet.2005.02.017.

    Article  CAS  PubMed  Google Scholar 

  40. Byler S, Goldgar S, Heerboth S, Leary M, Housman G, Moulton K, et al. Genetic and epigenetic aspects of breast cancer progression and therapy. Anticancer Res. 2014;34(3):1071–7.

    CAS  PubMed  Google Scholar 

  41. O'Flaherty JT, Wooten RE, Samuel MP, Thomas MJ, Levine EA, Case LD, et al. Fatty acid metabolites in rapidly proliferating breast cancer. PLoS One. 2013;8(5):e63076. https://doi.org/10.1371/journal.pone.0063076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hasegawa T, Yamamoto S, Matsuno Y. Quantitative immunohistochemical evaluation of MIB-1 labeling index in adult soft-tissue sarcomas by computer-assisted image analysis. Pathol Int. 2002;52(7):433–7. https://doi.org/10.1046/j.1440-1827.2002.01378.x.

    Article  CAS  PubMed  Google Scholar 

  43. Natarajan R, Esworthy R, Bai W, Gu JL, Wilczynski S, Nadler J. Increased 12-lipoxygenase expression in breast cancer tissues and cells. Regulation by epidermal growth factor. J Clin Endocrinol Metab. 1997;82(6):1790–8. https://doi.org/10.1210/jcem.82.6.3990.

    Article  CAS  PubMed  Google Scholar 

  44. Strasser A, Vaux DL. Cell Death in the Origin and Treatment of Cancer. Mol Cell. 2020;78(6):1045–54. https://doi.org/10.1016/j.molcel.2020.05.014.

    Article  CAS  PubMed  Google Scholar 

  45. Shen J, Tower J. Programmed cell death and apoptosis in aging and life span regulation. Discov Med. 2009;8(43):223–6.

    PubMed  Google Scholar 

  46. Kazan HH, Urfali-Mamatoglu C, Yalcin GD, Bulut O, Sezer A, Banerjee S, et al. 15-LOX-1 has diverse roles in the resensitization of resistant cancer cell lines to doxorubicin. J Cell Physiol. 2020;235(5):4965–78. https://doi.org/10.1002/jcp.29375.

    Article  CAS  PubMed  Google Scholar 

  47. Zuo X, Shen L, Issa JP, Moy O, Morris JS, Lippman SM, et al. 15-Lipoxygenase-1 transcriptional silencing by DNA methyltransferase-1 independently of DNA methylation. FASEB J. 2008;22(6):1981–92. https://doi.org/10.1096/fj.07-098301.

    Article  CAS  PubMed  Google Scholar 

  48. Hsi LC, Xi X, Lotan R, Shureiqi I, Lippman SM. The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces apoptosis via induction of 15-lipoxygenase-1 in colorectal cancer cells. Cancer Res. 2004;64(23):8778–81. https://doi.org/10.1158/0008-5472.CAN-04-1867.

    Article  CAS  PubMed  Google Scholar 

  49. Hague A, Manning AM, Hanlon KA, Huschtscha LI, Hart D, Paraskeva C. Sodium butyrate induces apoptosis in human colonic tumour cell lines in a p53-independent pathway: implications for the possible role of dietary fibre in the prevention of large-bowel cancer. Int J Cancer. 1993;55(3):498–505. https://doi.org/10.1002/ijc.2910550329.

    Article  CAS  PubMed  Google Scholar 

  50. Salimi V, Shabani M, Nourbakhsh M, Tavakoli-Yaraki M. Involvement of 15-lipoxygenase-1 in the regulation of breast cancer cell death induced by sodium butyrate. Cytotechnology. 2016;68(6):2519–28. https://doi.org/10.1007/s10616-016-9972-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Çolakoğlu M, Tunçer S, Banerjee S. Emerging cellular functions of the lipid metabolizing enzyme 15-Lipoxygenase-1. Cell Prolif. 2018;51(5):e12472. https://doi.org/10.1111/cpr.12472.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kim JH, Hubbard NE, Ziboh V, Erickson KL. Attenuation of breast tumor cell growth by conjugated linoleic acid via inhibition of 5-lipoxygenase activating protein. Biochim Biophys Acta. 1736;2005(3):244–50. https://doi.org/10.1016/j.bbalip.2005.08.015.

    Article  CAS  Google Scholar 

  53. Connolly JM, Liu XH, Rose DP. Dietary linoleic acid-stimulated human breast cancer cell growth and metastasis in nude mice and their suppression by indomethacin, a cyclooxygenase inhibitor. Nutr Cancer. 1996;25(3):231–40. https://doi.org/10.1080/01635589609514447.

    Article  CAS  PubMed  Google Scholar 

  54. Liu XH, Connolly JM, Rose DP. Eicosanoids as mediators of linoleic acid-stimulated invasion and type IV collagenase production by a metastatic human breast cancer cell line. Clin Exp Metastasis. 1996;14(2):145–52. https://doi.org/10.1007/BF00121211.

    Article  CAS  PubMed  Google Scholar 

  55. Rose DP, Connolly JM. Antiangiogenicity of docosahexaenoic acid and its role in the suppression of breast cancer cell growth in nude mice. Int J Oncol. 1999;15(5):1011–5. https://doi.org/10.3892/ijo.15.5.1011.

    Article  CAS  PubMed  Google Scholar 

  56. Dauchy RT, Dauchy EM, Sauer LA, Blask DE, Davidson LK, Krause JA, et al. Differential inhibition of fatty acid transport in tissue-isolated steroid receptor negative human breast cancer xenografts perfused in situ with isomers of conjugated linoleic acid. Cancer Lett. 2004;209(1):7–15. https://doi.org/10.1016/j.canlet.2003.12.012.

    Article  CAS  PubMed  Google Scholar 

  57. Jelińska M, Białek A, Gielecińska I, Mojska H, Tokarz A. Impact of conjugated linoleic acid administered to rats prior and after carcinogenic agent on arachidonic and linoleic acid metabolites in serum and tumors. Prostaglandins Leukot Essent Fatty Acids. 2017;126:1–8. https://doi.org/10.1016/j.plefa.2017.08.013.

    Article  CAS  PubMed  Google Scholar 

  58. Jelińska M, Białek A, Mojska H, Gielecińska I, Tokarz A. Effect of conjugated linoleic acid mixture supplemented daily after carcinogen application on linoleic and arachidonic acid metabolites in rat serum and induced tumours. Biochim Biophys Acta. 1842;2014(11):2230–6. https://doi.org/10.1016/j.bbadis.2014.08.013.

    Article  CAS  Google Scholar 

  59. Białek A, Jelińska M, Tokarz A. Influence of maternal diet enrichment with conjugated linoleic acids on lipoxygenase metabolites of polyunsaturated fatty acids in serum of their offspring with 7,12-dimethylbenz [a] anthracene induced mammary tumors. Prostaglandins Other Lipid Mediat. 2015;116-117:10–8. https://doi.org/10.1016/j.prostaglandins.2014.10.001.

    Article  CAS  PubMed  Google Scholar 

  60. Nony PA, Kennett SB, Glasgow WC, Olden K, Roberts JD. 15S-Lipoxygenase-2 mediates arachidonic acid-stimulated adhesion of human breast carcinoma cells through the activation of TAK1, MKK6, and p38 MAPK. J Biol Chem. 2005;280(36):31413–9. https://doi.org/10.1074/jbc.M500418200.

    Article  CAS  PubMed  Google Scholar 

  61. Sauer LA, Dauchy RT, Blask DE, Krause JA, Davidson LK, Dauchy EM. Eicosapentaenoic acid suppresses cell proliferation in MCF-7 human breast cancer xenografts in nude rats via a pertussis toxin-sensitive signal transduction pathway. J Nutr. 2005;135(9):2124–9. https://doi.org/10.1093/jn/135.9.2124.

    Article  CAS  PubMed  Google Scholar 

  62. Dauchy EM, Dauchy RT, Davidson LK, Lynch DT, Krause JA, Blue LM, et al. Human cancer xenograft perfusion in situ in rats: a new perfusion system that minimizes delivery time and maintains normal tissue physiology and responsiveness to growth-inhibitory agents. J Am Assoc Lab Anim Sci. 2006;45(3):38–44.

    CAS  PubMed  Google Scholar 

  63. Comba A, Maestri DM, Berra MA, Garcia CP, Das UN, Eynard AR, et al. Effect of ω-3 and ω-9 fatty acid rich oils on lipoxygenases and cyclooxygenases enzymes and on the growth of a mammary adenocarcinoma model. Lipids Health Dis. 2010;9(1):112. https://doi.org/10.1186/1476-511X-9-112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Blask DE, Dauchy RT, Sauer LA, Krause JA, Brainard GC. Growth and fatty acid metabolism of human breast cancer (MCF-7) xenografts in nude rats: impact of constant light-induced nocturnal melatonin suppression. Breast Cancer Res Treat. 2003;79(3):313–20. https://doi.org/10.1023/A:1024030518065.

    Article  CAS  PubMed  Google Scholar 

  65. Blask DE, Dauchy RT, Dauchy EM, Mao L, Hill SM, Greene MW, et al. Light exposure at night disrupts host/cancer circadian regulatory dynamics: impact on the Warburg effect, lipid signaling and tumor growth prevention. PLoS One. 2014;9(8):e102776. https://doi.org/10.1371/journal.pone.0102776.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kozłowski J, Kozłowska A, Kocki J. Breast cancer metastasis - insight into selected molecular mechanisms of the phenomenon. Postepy Hig Med Dosw. 2015;69:447–51. https://doi.org/10.5604/17322693.1148710.

    Article  Google Scholar 

  67. Viola K, Kopf S, Huttary N, Vonach C, Kretschy N, Teichmann M, et al. Bay11-7082 inhibits the disintegration of the lymphendothelial barrier triggered by MCF-7 breast cancer spheroids; the role of ICAM-1 and adhesion. Br J Cancer. 2013;108(3):564–9. https://doi.org/10.1038/bjc.2012.485.

    Article  CAS  PubMed  Google Scholar 

  68. Kerjaschki D, Bago-Horvath Z, Rudas M, Sexl V, Schneckenleithner C, Wolbank S, et al. Lipoxygenase mediates invasion of intrametastatic lymphatic vessels and propagates lymph node metastasis of human mammary carcinoma xenografts in mouse. J Clin Invest. 2011;121(5):2000–12. https://doi.org/10.1172/JCI44751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Pasqualini ME, Heyd VL, Manzo P, Eynard AR. Association between E-cadherin expression by human colon, bladder and breast cancer cells and the 13-HODE:15-HETE ratio. A possible role of their metastatic potential. Prostaglandins Leukot Essent Fatty Acids. 2003;68(1):9–16. https://doi.org/10.1016/S0952-3278(02)00230-2.

    Article  CAS  PubMed  Google Scholar 

  70. Boutté AM, McDonald WH, Shyr Y, Yang L, Lin PC. Characterization of the MDSC proteome associated with metastatic murine mammary tumors using label-free mass spectrometry and shotgun proteomics. PLoS One. 2011;6(8):e22446. https://doi.org/10.1371/journal.pone.0022446.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ryman VE, Packiriswamy N, Sordillo LM. Apoptosis of Endothelial Cells by 13-HPODE Contributes to Impairment of Endothelial Barrier Integrity. Mediat Inflamm. 2016;2016:9867138–13. https://doi.org/10.1155/2016/9867138.

    Article  CAS  Google Scholar 

  72. Blecha JE, Anderson MO, Chow JM, Guevarra CC, Pender C, Penaranda C, et al. Inhibition of IGF-1R and lipoxygenase by nordihydroguaiaretic acid (NDGA) analogs. Bioorg Med Chem Lett. 2007;17(14):4026–9. https://doi.org/10.1016/j.bmcl.2007.04.092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Somvanshi RK, Singh AK, Saxena M, Mishra B, Dey S. Development of novel peptide inhibitor of Lipoxygenase based on biochemical and BIAcore evidences. Biochim Biophys Acta. 1784;2008(11):1812–7. https://doi.org/10.1016/j.bbapap.2008.07.004.

    Article  CAS  Google Scholar 

  74. Lee PP, Ip MM. Regulation of proliferation of rat mammary tumor cells by inhibitors of cyclooxygenase and lipoxygenase. Prostaglandins Leukot Essent Fatty Acids. 1992;45(1):21–31. https://doi.org/10.1016/0952-3278(92)90098-4.

    Article  PubMed  Google Scholar 

  75. Reddy N, Everhart A, Eling T, Glasgow W. Characterization of a 15-lipoxygenase in human breast carcinoma BT-20 cells: stimulation of 13-HODE formation by TGF alpha/EGF. Biochem Biophys Res Commun. 1997;231(1):111–6. https://doi.org/10.1006/bbrc.1997.6048.

    Article  CAS  PubMed  Google Scholar 

  76. Röhrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016;16(11):732–49. https://doi.org/10.1038/nrc.2016.89.

    Article  CAS  PubMed  Google Scholar 

  77. Nagarajan SR, Butler LM, Hoy AJ. The diversity and breadth of cancer cell fatty acid metabolism. Cancer Metab. 2021;9(1):2. https://doi.org/10.1186/s40170-020-00237-2.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Koundouros N, Poulogiannis G. Reprogramming of fatty acid metabolism in cancer. Br J Cancer. 2020;122(1):4–22. https://doi.org/10.1038/s41416-019-0650-z.

    Article  CAS  PubMed  Google Scholar 

  79. Li D, Li Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct Target Ther. 2020;5(1):108. https://doi.org/10.1038/s41392-020-00216-5.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Wu X, Liu C, Li Z, Gai C, Ding D, Chen W, et al. Regulation of GSK3β/Nrf2 signaling pathway modulated erastin-induced ferroptosis in breast cancer. Mol Cell Biochem. 2020;473(1-2):217–28. https://doi.org/10.1007/s11010-020-03821-8.

    Article  CAS  PubMed  Google Scholar 

  81. Salimi V, Shahsavari Z, Safizadeh B, Hosseini A, Khademian N, Tavakoli-Yaraki M. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis. 2017;16(1):208. https://doi.org/10.1186/s12944-017-0593-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Han J, Liu Y, Yang S, Wu X, Li H, Wang Q. MEK inhibitors for the treatment of non-small cell lung cancer. J Hematol Oncol. 2021;14(1):1. https://doi.org/10.1186/s13045-020-01025-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Orafaie A, Matin MM, Sadeghian H. The importance of 15-lipoxygenase inhibitors in cancer treatment. Cancer Metastasis Rev. 2018;37(2-3):397–408. https://doi.org/10.1007/s10555-018-9738-9.

    Article  CAS  PubMed  Google Scholar 

  84. Kashyap D, Mittal S, Sak K, Singhal P, Tuli HS. Molecular mechanisms of action of quercetin in cancer: recent advances. Tumor Biol. 2016;37(10):12927–39. https://doi.org/10.1007/s13277-016-5184-x.

    Article  CAS  Google Scholar 

  85. Ezzati M, Yousefi B, Velaei K, Safa A. A review on anti-cancer properties of Quercetin in breast cancer. Life Sci. 2020;248:117463. https://doi.org/10.1016/j.lfs.2020.117463.

    Article  CAS  PubMed  Google Scholar 

  86. Fernand VE, Losso JN, Truax RE, Villar EE, Bwambok DK, Fakayode SO, et al. Rhein inhibits angiogenesis and the viability of hormone-dependent and -independent cancer cells under normoxic or hypoxic conditions in vitro. Chem Biol Interact. 2011;192(3):220–32. https://doi.org/10.1016/j.cbi.2011.03.013.

    Article  CAS  PubMed  Google Scholar 

  87. Tahamtan A, Tavakoli-Yaraki M, Shadab A, Rezaei F, Marashi SM, Shokri F, et al. The Role of Cannabinoid Receptor 1 in the Immunopathology of Respiratory Syncytial Virus. Viral Immunol. 2018;31(4):292–8. https://doi.org/10.1089/vim.2017.0098.

    Article  CAS  PubMed  Google Scholar 

  88. Caffarel MM, Sarrió D, Palacios J, Guzmán M, Sánchez C. Delta9-tetrahydrocannabinol inhibits cell cycle progression in human breast cancer cells through Cdc2 regulation. Cancer Res. 2006;66(13):6615–21. https://doi.org/10.1158/0008-5472.CAN-05-4566.

    Article  CAS  PubMed  Google Scholar 

  89. Sadeghian H, Jabbari A. 15-Lipoxygenase inhibitors: a patent review. Expert Opin Ther Pat. 2016;26(1):65–88. https://doi.org/10.1517/13543776.2016.1113259.

    Article  CAS  PubMed  Google Scholar 

  90. Armstrong MM, Freedman CJ, Jung JE, Zheng Y, Kalyanaraman C, Jacobson MP, et al. A potent and selective inhibitor targeting human and murine 12/15-LOX. Bioorg Med Chem. 2016;24(6):1183–90. https://doi.org/10.1016/j.bmc.2016.01.042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was financially supported by Iran University of Medical Sciences (Grant Number: 93–02–30-24469). The funding body played no role in the design of the study, collection, interpretation and in writing a manuscript.

Author information

Authors and Affiliations

Authors

Contributions

M.A.V performed data collection and investigation and contributed in preparing tables and writing - original draft, B. S and A.R.E, contributed in data collection and investigation, S.S.GH and M. R prepared figures, V. S performed conceptualization and validate data and M.T.Y designed the study, supervised the project and prepared the manuscript.All authors read and approved the final manuscript.

Corresponding author

Correspondence to Masoumeh Tavakoli-Yaraki.

Ethics declarations

Consent for publication

Not applicable.

Competing interests

The authors declare that there is no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vaezi, M.A., Safizadeh, B., Eghtedari, A.R. et al. 15-Lipoxygenase and its metabolites in the pathogenesis of breast cancer: A double-edged sword. Lipids Health Dis 20, 169 (2021). https://doi.org/10.1186/s12944-021-01599-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12944-021-01599-2

Keywords