Next Article in Journal
A Review of the Potential Role of Human Cytomegalovirus (HCMV) Infections in Breast Cancer Carcinogenesis and Abnormal Immunity
Next Article in Special Issue
The Chalcone Lonchocarpin Inhibits Wnt/β-Catenin Signaling and Suppresses Colorectal Cancer Proliferation
Previous Article in Journal
ANLN and TLE2 in Muscle Invasive Bladder Cancer: A Functional and Clinical Evaluation Based on In Silico and In Vitro Data
Previous Article in Special Issue
Streptomyces sp. MUM256: A Source for Apoptosis Inducing and Cell Cycle-Arresting Bioactive Compounds against Colon Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reversal of Epithelial–Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids

College of Pharmacy, Dongguk University, Seoul 100-715, Korea
Cancers 2019, 11(12), 1841; https://doi.org/10.3390/cancers11121841
Submission received: 23 October 2019 / Revised: 16 November 2019 / Accepted: 19 November 2019 / Published: 21 November 2019
(This article belongs to the Special Issue Role of Natural Bioactive Compounds in the Rise and Fall of Cancers)

Abstract

:
Epithelial mesenchymal transition (EMT) is a key process in the progression of malignant cancer. Therefore, blocking the EMT can be a critical fast track for the development of anticancer drugs. In this paper, we update recent research output of EMT and we explore suppression of EMT by natural anti-inflammatory compounds and pro-resolving lipids.

1. Introduction

The epithelial–mesenchymal transition (EMT) is defined as a phenomenon that epithelial cells transform into mesenchymal cells [1]. EMT plays a key role in cancer progression and fibrosis. Many researchers and pharmaceutical companies have tried to develop novel EMT blockers due to its importance in such diseases [2,3,4].
Inflammation is one of ten in cancer hallmark [5]. It is a critical factor of tumor microenvironment affecting EMT. Several reviews have emphasized role of inflammation in EMT [6]. However, there are a few reviews that deal with the blocking of EMT by natural anti-inflammatory compounds and pro-resolving lipids.
In this review, I have dealt with ingredients derived from natural products that were not covered in the 2018 review on EMT [6]. I have also added the story of inflammasomes, which play an essential role in the early steps of inflammation, and how they are involved in EMT. Naturally derived compounds that control these inflammasome-related molecules in the EMT have been discussed. About the resolution of inflammation, the newly discovered pro-resolving lipids including RvTs are added and the receptors specifically acting on pro-resolving lipids have also been discussed. Understanding the action of natural anti-inflammatory compounds and pro-resolving lipids with anti-EMT activities might provide a new armory to suppress the progression of cancer.

2. EMT in Cancers

EMT shows the reduced expression of epithelial markers including E-cadherin and keratins and the increased expression of mesenchymal marker proteins such as vimentin and N-cadherin via actions of transcription factors including as SNAIL1 and ZEB1 (Figure 1) [6]. The molecular mechanism of EMT process has been well explained in the report (references in it) [1]. In this part, we will briefly update the concept that reflects recent achievements for EMT.
EMT occurs by a various mediators from tumor microenvironments via receptor through signal transduction. EMT-related transcription factors blocks the expression of epithelial cell-marker genes and evoke mesenchymal-marker genes. E-cadherin, keratin, ZO-1, miR-34, and miR-200 belong to the epithelial markers, and N-cadherin, vimentin, fibronectin, SNAIL, ZEB1, TWIST, Brachyury, Foxq1, Runx2, GATA, and SOX belong to the mesenchymal markers. P-cadherin is the marker of partial EMT. The dot triangle indicates the relative ratio of p-cadherin expression. Modified from Lee’s report [6].

2.1. Adaptation to New Concepts of EMT

2.1.1. Signaling Pathways in the EMT Process

A typical signaling pathway of EMT is the transforming growth factor-β1 (TGF-β1) pathway. TGF-β1 induces EMT via SMAD-dependent or non-SMAD signaling pathway [7]. Growth factors including FGF, HGF, IGF1, EGF, and PDGF via receptor tyrosine kinase can induce EMT via signaling pathway of PI3K-AKT and ERK MAPK [8,9,10,11]. Wnt signaling, hedgehog signaling, Notch signaling, hypoxia, and inflammatory tumor microenvironment also involves in EMT [5]. Recently, it has been shown that hippo signaling is also involved in EMT [12]. YAP and TAZ can enhance EMT through upregulation of EMT transcription factors such as forkhead box C2 (FOXC2), snail family zinc finger 1/2 (SNAIL1, SLUG), twist-related protein 1 (TWIST1), and ZEB1 [12,13,14,15].

2.1.2. Transcription Factors Involved in EMT

Novel players are newly recognized as regulatory transcription factor in the EMT. Brachyury, the T-box transcription factor, is a novel transcription factor implicated in the EMT of cancer cells [16]. Brachyury is known as the target gene of WNT, one of the major signaling pathways of EMT [17]. Foxq1, one of forkhead transcription factor, has also regarded as a novel transcription factor mediating the EMT of gastric cancer [6,18]. Runt-related transcription factor 2 (Runx2) belongs to the runt-related transcription factor family [19]. Runx2 plays a key role in EMT of hepatocellular carcinoma (HCC) [20]. GATA transcription factors are also implicated in the EMT of cancer cells [21]. Serine 161 and serine 187 phosphorylated GATA1 by PAK5 can promote EMT of breast cancer cells by recruiting histone deacetylase 3/4 to E-cadherin promoter [22]. Other players of EMT include SRY-box (SOX) transcription factors [23]. Sox4 acts as a master regulator in EMT of cancerous breast epithelial cells [24].

2.1.3. Partial EMT

EMT could be not defined as a dichotomous transition from epithelial status to mesenchymal one of cells [6,25].
Partial EMT with both epithelial and mesenchymal cell markers was proposed (Figure 1) [25]. Cells that have undergone with partial EMT have the capability showing collective sheet or cluster migration [26]. Partial EMTed cells have the competitiveness in that not all cells need to respond to EMT signals. Thus, they can far more efficiently execute plasticity in converting to a colonization state of metastasis via MET [25].

2.1.4. Parallelism between Cancer Stem Cell and EMT

A cancer stem cell (CSC) is a cancer cell having the ability of self-renewal and differentiation. It divides to progenitor cancer cells. It is a culprit of cancer recurrence and metastasis [27]. Several EMT transcription factors and inducers can evoke the expression of cancer stem cells markers, thereby enhancing the capability to initiate cancer, a typical characteristic of cancer stem cells [28]. TGF-β promotes the de-differentiation of human basal breast non-CSCs into CSCs via ZEB1, suggesting that the activation of EMT in cancer cells by TGF-β/ZEB1 is closely linked to the de-differentiation of cancer cells into the CSC state [29].

2.2. Focus on Chemoresistance and Immune Evasion of EMT in Cancers

In the course of EMT, epithelial cancer cells can lose contacts between cells and apicobasal polarity but gain enhanced migration and invasion [1,6]. EMT has also strong influences on several hallmarks of cancer including cancer initiation, immune evasion, proliferation, survival, and resistance to therapeutics [6,30]. We will briefly update EMT’s roles in cancer such as chemoresistance and immune evasion.

2.2.1. Chemoresistance

Chemoresistance to anticancer chemotherapeutics implies that cancer cells can survive despite the administration of an anticancer drug in a dose that can usually kill cancer cells. A significant correlation has been found between EMT-related gene expression and chemo-resistance to anticancer therapy [6,30,31]. Although the role of EMT in metastasis is disputable by some group, EMT is crucial for anticancer drug resistance [32,33]. For example, TWIST-mediated EMT is related to sorafenib resistance to advanced HCC [34]. Therefore, EMT should be understood as a predictor of chemoresistance for anticancer drugs.

2.2.2. Immune Evasion

EMT has been understood as a key mechanism of immune escape of cancer cells. When snail1, an EMT transcription factor, is ectopically expressed in MCF7 breast cancer cells, cancer cell lysis executed by TNF-α-induced CTL is curtailed [35]. When snail1 is overexpressed in B16 melanoma cells, CTL-induced lysis is reduced and maturation of dendritic cells is inhibited while inhibitory Treg-like CD4+ Foxp3+ cells are expanded [36].
Enhanced EMT properties in cancer cells (A549, MCF7, and HepG2) by the TGF-β, IFN-γ, and TNF-α may affect differentiation and death of natural killer (NK), T, and B cells [37]. Reduced miR-200 and enhanced ZEB1 expression in lung cancer cells not only can evoke EMT, but also can lead to enhanced expression of PD-L1, which is related to the exhaustion of CD8+ T lymphocytes in lung cancer tissues [38]. In contrast, activated CD8+ T cells provoke mammary epithelial tumor cells to experience EMT, thus obtaining cancer-initiating power of breast cancer stem cells [6,39].

3. Induction of EMT by Mediators from the Chronic Inflammatory Tumor Microenvironment

3.1. Upgrade of Inflammation Concept: From Initiation (alpha) to Resolution (omega)

Inflammation is intrinsically a protective process via microcirculation. Local or systemic inflammatory reactions delete the causing stimuli and reboot repair and healing processes of tissue [40].
Acute inflammation has two phases: initiation (alpha) and resolution (omega). Inflammation starts by the soluble inflammatory mediators such as complement, cytokines including chemokines, free radicals, vasoactive amines, and eicosanoids (including prostaglandins) by adjacent cells of the infected or injured part in the body [6,40,41].
Inflammasome is a multiprotein oligomer responsible for the activation of inflammatory responses and consists of NLRP protein such as NLRP3, ASC, and procaspase-1 [42,43]. The inflammasome can promote the maturation and secretion of interleukin 1β (IL-1β) and IL-18 [44].
Inflammasomes are involved in the EMT of cancer or other epithelial cells. For example, knockdown of NLRP3 alleviates high glucose or TGF-β1-induced EMT in human renal tubular cells [45]. NLRP3 regulates cellular proliferation and metastasis via EMT and the PTEN/AKT signaling pathway [46]. NLRP3 inhibition can attenuate silica-induced EMT in human bronchial epithelial cells [47]. NLRP3 also participates in the regulation of EMT in bleomycin-induced pulmonary fibrosis [48]. Uric acid can activate NLRP3 inflammasome in the EMT in the kidney of rats [49]. NLRP3 appears to be important for EMT since inflammasome-independent NLRP3 is enough to EMT in colon cancer cells [50].
The resolution (omega) phase of inflammation releases specialized lipid mediators that can actively prevent further progress of inflammation and enhance resolution of inflammation [51]. A new specialized group of lipids that can actively terminate inflammation has been found by Serhan et al. [52]. These kinds of lipids include lipoxins (Lx), resolvins (Rvs), protectins (PDs), and maresins (MaRs; Figure 2). They exhibit inflammation-suppressing action with pro-resolving effect that can promote efferocytosis [41,52].
13-series resolvins (RvT1-4) derived from docosapentaenoic acid (DPA) have been newly discovered [53]. Briefly describing their production, human platelets pre-treated with aspirin or atorvastatin convert omega-3 DPA (DPAn-3) to a 13S-hydroperoxy intermediate via aspirin-treated or atorvastatin-treated COX-2. This intermediate is converted into four RvTs via ALOX5 enzyme activity exerted on the nearby neutrophils [53]: RvT1 (7,13R,20-trihydroxy-DPAn-3); RvT2 (7,8,13R-trihydroxy-DPAn-3); RvT3 (7,12,13R-trihydroxy-8Z,10E,14E,16Z,19Z-DPAn-3); and RvT4 (7,13R-dihydroxy-DPAn-3). Four RvTs are also formed by a mixture of human neutrophils and vascular endothelium cells, which are found in infected rodent and human tissues. [52].

3.2. EMT Inducers from Chronic Inflammatory Tumor Microenvironments

The tumor microenvironment mainly influences the progression of cancers via secretion of various factors that cause EMT [54,55]. Cancer-related chronic inflammation is described as a chaotic state where both pro-inflammatory and anti-inflammatory signals are present to permit tumor growth and immune evasion [6]. Besides, the tumor microenvironment contributes to the cancer heterogeneity. Therefore, the tumor microenvironment has been regarded as a promising target for the cure of cancer. Thus, Vanneman and Dranoff have demonstrated a novel way of curing cancer by re-educating the tumor microenvironment [56].
Here, we will briefly introduce EMT inducers from chronic inflammatory tumor microenvironment (Figure 3).
TNF-α is a critical determiner of inflammatory responses [57]. Serum concentration of TNF-α was determined as 1.47 pg/mL in invasive breast cancer patients and 0.98 ± 0.37 pg/mL in the control cohort [6,58]. TNF-α produced by macrophages can accelerate TGF- 1-induced EMT [59].
IL-6 serves as either a pro-inflammatory or anti-inflammatory cytokine [6]. The mean serum concentration of IL-6 was observed as 31.7 pg/mL in patients with breast cancer and 3.3 pg/mL in the normal cohort [6,60]. IL-6 induces EMT of human breast cancer cells [61]. IL-6/STAT3-induced expression of lncTCF7 can promote EMT of liver cancer cells [62]. The IL-6 pathway induces EMT in biliary tract cancer via cross-talking to the SMAD4 in the TGF- 1 pathway [63].
IL-8 is a chemokine mainly secreted by macrophages [64]. The mean serum concentration of IL-8 was found as 40.1 pg/mL in patients with breast cancer and 5.3 pg/mL in the normal group [60]. IL-8 expression is highly increased in TGF- 1-induced EMT in colon carcinoma and nasopharyngeal carcinoma [65]. IL-8 is also involved in mast cell-induced EMT of human lung and thyroid cancer cells [66,67]. JAK2/STAT3/Snail pathway is involved in the IL-8-induced EMT of HCC cells [68]. Brachyury-induced EMT of the tumor is mediated by IL-8/IL-8R signaling pathway [69].
IL-17 is a pro-inflammatory cytokine and mainly released from Th17 cells and macrophages [70]. IL-17 induces EMT of prostate cancers via MMP7 [71]. IL-17 induces EMT through STAT3 in the lung adenocarcinoma [72]. IL-17 can evoke self-renewal of CD133+ cancer cells in ovarian cancer [73].
High-mobility group box 1 (HMGB1) is a nuclear DNA-binding protein and released to the outside from macrophages, NK cells, dendritic cells, necrotic cells, and apoptotic cells according to infection, injury, and inflammation [74]. The mean serum HMGB1 level was 4.64 ng/mL in patients with malignant breast cancer, which was remarkably higher than in patients with benign breast cancer (1.32 ng/mL) or in healthy subjects (1.36 ng/mL) [75]. HMGB1 (2 μg/mL) induces EMT of colorectal and prostate cancer cells via the RAGE/NF-κB pathway [76,77].
IL-10 is a potent anti-inflammatory cytokine that suppresses T cell/macrophage cytokine synthesis and blocks their antigen-presenting capacity [78]. In vitro generated M1- and M2-macrophages both can induce EMT of pancreatic cancer cells via the IL-10 signaling pathway [79].
TGF- 1 is a potent anti-inflammatory cytokines [80]. Plasma TGF- 1 levels were significantly higher in stage IIIB/IV breast cancer patients (2.40 ng/mL) than those in healthy controls (1.30 ng/mL) [81]. It is a typical EMT inducer involved in cancer progression [6]. Please read other review for a detailed information about TGF- 1-induced EMT [6,82]. TGF- 1 promotes the production of IL-10 in macrophages from mouse cancer [83]. HMGB1 promotes expression of TGF- 1 via RAGE pathway to mediate TGF-1-induced EMT [84].
PGE2 is biosynthesized from arachidonic acids (Figure 2). The mean serum level of PGE2 was 6.324 pg/mL in patients with brain cancer and 1.677 pg/mL in the compared normal cohort [85]. PGE2 acts through G protein-coupled receptors such as EP1-EP4 [86]. PGE2 (2–10 μg/mL) works in an autocrine or paracrine manner, leading to stimulation of EMT through the expression of SNAIL and ZEB1 [87]. In turn, SNAIL induces blocking of degradation of PGE2 by repressing prostaglandin dehydrogenase, generating a positive loop that promotes cancer progression [88]. PGE2 (5 μM) enhances invasion of HCC cells via EP1-mediated expression of YB-1, which induces TGF- 1-induced EMT by AKT activation [89]. PGE2 (0.01–1 μM) inhibits fibroblast chemotaxis but stimulates chemotaxis of epithelial cells in the airway [90]. PGE2′s inhibitory action against TGF-1-induced EMT seems to be via the EP2 pathway since EP2 agonist can block TGF- 1-induced EMT [91]. As such, the effect of PGE2 on EMT varies depending on concentration and in cell types.
Leukotriene B4 (LTB4) can be produced via the 5-lipoxygenase pathway. Significantly higher levels of LTB4 have been found in the whole blood of lung cancer patients than those in the control group (44.1 vs. 17.9 pg/mL) [92]. LTB4 acts via two distinct GPCR called BLT1 and BLT2. BLT2 is involved in keratin phosphorylation and perinuclear reorganization, which is a prelude of EMT [93]. Accordingly, BLT2 is involved in the ras-promoted TGF-1-induced EMT [94]. As expected, LTB4 (100 nM) can induce EMT leading to vimentin expression through the BLT2/ERK2 activation [95].
Cysteinyl leukotrienes (CysLTs) include LTC4, LTD4, and LTE4 [96]. Mean serum levels of LTD4 found in HCC patients and healthy groups were 174.95 and 10.75 pg/mL, respectively [97]. Actions of CysLTs are mediated via GPCRs such as CysLT1 and CysLT2 [98]. LTD4 (100 nM) can suppress E-cadherin expression in cancer cells through enhanced translocation of -catenin to the nucleus while montelukast (0.1 mM) suppresses eosinophil-induced EMT in bronchial epithelial cells [99]. Recently, exosomes and cells from ascites in lung cancer patients can convert LTC4 to LTD4 to promote cancer cell migration and invasion via CysLT1 [100].

4. Reversal of EMT by Anti-inflammatory and Pro-Resolving Natural Compounds

The 2018 review by us did not cover the regulation of EMT by natural products [6]. In this part, we describe anti-inflammatory natural compounds and pro-resolving lipids that can prohibit EMT stimulated by mediators released in the tumor microenvironment.

4.1. Reversal of EMT by Anti-Inflammatory Natural Compounds

Diverse natural products can inhibit EMT of cancer cells or epithelial cells. We focused on EMT inhibitors found from natural compounds that can inhibit the EMT of cancer cells caused by EMT inducers from the chronic inflammatory tumor microenvironment due to limited space (Figure 4, Table 1).
Arctigenin from Asteraceae has anti-inflammatory effects [101]. Arctigenin (12–50 μM) can suppress TGF--induced EMT of human lung cancer cells, thus blocking invasion [102]. Arctigenin (0.25–1 μM) can inhibit the expression of MCP-1 and subsequent EMT induced by ROS-dependent ERK/NF-κB pathway of renal tubular epithelial cells [103].
Baicalin found in Scutellaria baicalensis and Scutellaria lateriflora has well-known anti-inflammatory effects [104]. Baicalin (2 μM) can suppress TGF-1-mediated EMT in MCF10A cells by reducing the expression of slug [105]. Baicalin (12.5–25 μM) can inhibit the expression of TGF-1-induced EMT-related transcription factors in osteosarcoma cells and inhibit the aggressive metastasis of breast cancer by blocking EMT via inhibiting the activation of -catenin [106,107]
Berberine found in Berberis can reduce the secretion of IL-1 and TNF-α [108,109]. Berberine (5–20 μM) can reverse EMT in uterine cancer, leading to suppression of cancer metastasis [110]. Berberine can inhibit the metastatic ability of prostate cancer cells by suppressing EMT-associated genes [111]. Berberine (50 μM) can make nasopharyngeal carcinoma cells sensitive to radiation through EMT inhibition [112].
Betanin from beets can decrease the production of superoxide anion and cytokines TNF-α and IL-1 [113,114].
Betanin (25–50 μM) can inhibit high glucose-induced EMT of renal proximal tubular cells [115]. However, activity of betanin for EMT of cancer cells has not been reported yet.
Brusatol from the seeds of Brucea sumatrana can inhibit the response of cultured beta-cells to pro-inflammatory cytokines in vitro [116]. Brusatol (2 μM) can inhibit the EMT of pancreatic cancer cells [117].
Cardamonin, one of major component of Alpinia katsumadai has anti-tumor, anti-inflammatory, anti-nociceptive, and anti-itching activities [118,119,120]. Cardamonin (1–10 μM) can suppress TGF-1-stimulated EMT of A549 cells by restoring protein phosphatase 2A expression [120]. Cardamonin (5–20 μM) can block the invasiveness of human triple negative breast cancer cell by downregulation of Wnt/-catenin signaling pathway and induce the reversal of EMT [121]. Cardamonin (5–25 μM) also inhibits transglutaminase-2, one players in EMT, leading to JNK activation and NF-κB pathway [122].
Carnosol, found in Rosmarinus officinalis and Salvia pachyphylla, can block UV-induced inflammation through inhibition of STAT3 [123,124,125]. For a more in-depth look at various anti-inflammatory effects of carnosol, please refer to the 2017 review [126]. Carnosol (0.1–10 μM) controls the human glioblastoma stemness features by modulating EMT and inducing cancer stem cell apoptosis [127]. Carnosol (5–10 μM)-mediated SIRT1 activation inhibits the enhancer of zeste homolog 2 to attenuate liver fibrosis [128].
Celastrol (0.1–1 μM), from the root extracts of Tripterygium wilfordii and Celastrus regelii, can suppress experimental autoimmune encephalomyelitis [129]. Celastrol can inhibit the expression of snail and increased the expression of E-cadherin in the lung cancer cells [130]. Many studies have reported the effect of celastrol on EMT of diseases other than cancer (please refer the ref 128 by Kashyap et al.) [131].
Codonolactone, a major component of Atractylodes lancea, exhibits anti-allergic activity, anti-inflammatory, anticancer, gastroprotective, and neuroprotective activities [132,133]. Codonolactone (10–40 μM) can inhibit EMT in breast cancer cells by downregulating the transcriptional activity of Runx2 [134].
Cordycepin from the fungus Cordyceps militaris can suppress LPS-induced cytokine production by increasing heme oxygenase-1 expression [135,136]. Cordycepin (25–100 μM) can inhibit cancer stemness of TGF- induced chemo-resistant ovarian cancer cells [137]. Metronomic cordycepin therapy (25 mg/kg and 50 mg/kg) can prolong the survival of oral cancer-bearing mice and it (50 μM) inhibit EMT [138]. Cordycepin (100–200 μM) suppresses integrin/FAK signaling and EMT in HCC [139].
Cryptotanshinone, obtained from the root of Salvia miltiorrhiza, can protect against IL-1-induced inflammation in human osteoarthritis chondrocytes [140]. Cryptotanshinone (5–10 μM) targets tumor-initiating cells through down-regulation of stemness genes expression [141].
Curcumin, a phenolic compound found in Zingiberaceae turmeric, has strong anti-inflammatory, antioxidant, and antitumor properties [142]. A more extensive and detailed review of curcumin’s EMT has been reported recently [143]. Curcumin (25–50 μM) can inhibit metastasis in human papillary thyroid carcinoma cells by negatively regulating TGF-1-mediated Smad2/3 signaling pathway [142]. Curcumin (15 μM) can inhibit TNF-α-induced EMT in melanoma [144]. It can decrease EMT in cervical cancer cells by a pirin-dependent mechanism [145]. Pirin is a coregulatory of NF-κB involved in EMT [146]. Curcumin (30 μM) can suppress paraquat-induced EMT by blocking TGF- in A549 cells [147]. It (8 μM) can reverse oxaliplatin resistance caused by EMT in colorectal cancer through inhibition of the TGF-/Smad2/3 pathway [148].
Dioscin from roots of wild yam (Dioscorea villosa) shows potent anti-inflammatory effects via suppression of TNF-α-induced NF-κB-mediated VCAM-1, and ICAM-1 expression [149,150]. Dioscin (3 μM) can suppress TGF-1-induced EMT in A549 and HepG2 cells [149,151]. Dioscin (1–10 μM) also reverses HMT-induced EMT by down-regulating mdm2 and vimentin [152]. Diosgenin (400 μM), an aglycone of dioscin inhibits breast cancer cells with stem cell like properties by attenuation of the Wnt-catenin signaling [153].
Delphinidin, an anthocyanin, can reduce levels of inflammatory mediators including IL-6 and TNF-α induced by LPS [154]. Delphinidin (10–50 μM) inhibits TGF-1-induced EMT through a TGF- 1/Smad2 signaling pathway in glioblastoma cells and EGF-induced EMT in HCC cells [155,156].
Epigallocatechin-3-gallate found in green tea can regulates anti-inflammatory action through laminin receptor-mediated tollip signaling induction in LPS-stimulated human intestinal epithelial cells [157]. Epigallocatechin-3-gallate (25–50 μM) can also suppress nicotine-induced migration and invasion by blocking angiogenesis and EMT of non-small cell lung carcinoma (NSCLC) cells [158]. It (10–60 μM) also suppresses EMT and invasion in anaplastic thyroid carcinoma cells by blocking TGF-1/Smad pathways [159].
Eupatolide from the Inula britannica is used to treat bronchitis, disorders of the digestive system and inflammation [160]. Eupatolide can prohibit LPS-stimulated COX-2 and iNOS expression of RAW264.7 cells by evoking proteasomal degradation of TRAF6 [161]. Eupatolide (3–10 μM) can inhibit TGF-1-induced migration of breast cancer cells via down-regulation of SMAD3 phosphorylation and transcriptional repression of ALK5 [160].
Gallic acid is a trihydroxybenzoic acid found in gallnuts, sumac, witch hazel, and tea leaves [162]. For various anti-inflammatory effects of gallic acid, please refer to the 2017 review [163]. Gallic acid-coated sliver nanoparticle (50 μg/mL) can alter the expression of radiation-induced EMT in NSCLC [164]. Black tea polyphenols (10–40 μM) can also reverse EMT and inhibit invasion of human oral cancer cells [165].
Gambogic acid from the brownish or orange resin of Garcinia hanburyi can enhance the expression of heme oxygenase-1 through Nrf2 pathway and inhibit NF-κB and MAPK activation to mitigate inflammation in LPS-activated RAW264.7 cells [166]. Gambogic acid (0.5–1 μM) can suppress cancer invasion and migration by inhibiting TGF-1-induced EMT [167]. It (2 μM) can induce cleavage of vimentin in HeLa cells [168].
Gedunin, one of the main chemical compounds in the neem tree, can protect TLR-mediated inflammation by suppression of inflammasome activation and cytokine production [169,170]. Gedunin (15 μM) suppresses EMT of pancreatic cancer by inhibiting sonic hedgehog signaling pathway [171].
Genistein first isolated from Genista tinctoria can suppress psoriasis-related inflammation through a STAT3/NF-κB-dependent mechanism in keratinocytes [172,173]. For more detailed information about genistein’s anti-inflammatory action or anti-EMT, please refer to a previous review by Spagnulo et al. and Lee et al. [174,175]. Genistein (200 μM) can induce apoptosis of colon cancer cells by reversal of EMT via a notch1/NF-κB/slug/E-cadherin pathway [176]. miR-223 inhibitor and genistein (20 μM) can synergistically reverse in EMT of gemcitabine-resistant pancreatic cancer cells [177].
Geraniin found in geraniums can ameliorate experimental acute reflux esophagitis via NF-κB-regulated anti-inflammatory activities in rats [178,179]. It (15–20 μM) can also suppress TGF-1-induced EMT, migration, invasion and anoikis resistance in A549 lung cancer cells [180].
Gigantol from the Cymbidium goeringii can suppress LPS-stimulated iNOS and COX-2 expression through NF-κB inactivation in RAW 264.7 cells [181]. It (5–20 μM) can attenuate cancer stem cell-like phenotypes and induce anoikis in human lung cancer H460 cells [182,183].
Ginkgolic acid from Ginkgo biloba can significantly inhibit the production of NO, PGE2, and pro-inflammatory cytokines in ox-LDL-stimulated HUVECs cells [184]
Ginkgolic acid (100 μM) can inhibit TGF-1-induced EMT of lung cancer cells through PI3K/AKT/mTOR inactivation [185]. PPAR-γ might be involved in the suppression of EMT since ginkgolic acid is a PPAR-γ modulator.
Ginsenosides Rh1, Rg3, Rb1, Rg5, and Rg1 from ginseng can block inflammatory responses by inhibiting the activation of NLRP3, NLRP1, and AIM [186]. Rg3 (25–100 μM) can suppress EMT and invasion in lung cancer cells by reducing expression of FUT4 [187]. It (75 μg/mL) can also sensitize hypoxic lung cancer cells to cisplatin via blocking of NF-κB mediated EMT [188]. Rb1 (160 μg/mL) can inhibit hypoxia-induced EMT in ovarian cancer cells by regulating miR-25 [189]. Downregulation of HDAC3 by Rg3 (25 and 50 μg/mL) can inhibit EMT of cutaneous squamous cell carcinoma through c-Jun acetylation [190]. Rg3 (75 μg/mL) can inhibit growth and EMT of human oral squamous carcinoma cells by down-regulating miR-221 [191].
Glycyrrhizin (50–200 μM) from the roots of Glycyrrhiza glabra (Licorice) has anti-inflammatory and antiviral activities. It is also a novel pharmacological inhibitor of HMGB1 [192]. Glycyrrhizin can attenuate the EMT of prostate cancer cells by suppressing HMGB1-involved signaling pathway [192].
Honokiol from the Magnolia possesses anti-inflammatory activity by blocking downstream signaling of MEKK-1 in NF-κB activation pathway [193,194]. It (30 μM) can also inhibit EMT-mediated migration of human NSCLC cells in vitro by targeting c-FLIP and EMT of breast cancer cells by targeting STAT3/Zeb1/E-cadherin axis [195,196]. Honokiol (20 μM) can inhibit the metastasis of renal cancer cells by blocking EMT through regulating miR-141/ZEB2 pathway [197]. It (5–20 μM) can also inhibit the invasion of U87MG human glioblastoma cell via regulation of EMT [198].
Hypaconitine from the root of Aconitum species can suppress 0.1% histamine-induced acute inflammation without showing an ulcerogenic effect [199]. Hypaconitine (8 μM) can inhibit TGF-1-evoked EMT of A549 lung cancer cells possibly by blocking NF-κB activation [200].
Jatrophone from Jatropha isabellei has anti-nociceptive and anti-inflammatory activities [201]. Jatrophone (2 μM) can interfere with Wnt/-catenin signaling and reverses EMT of human triple-negative breast cancer [202].
Ligustrazine found in nattō and in fermented cocoa beans can significantly decrease CCL3, CCL19, CCl21, IL-4, IL-5, and IL-17A in bronchoalveolar lavage fluid of ovalalbumin-induced mice [203,204]. There are many reports of anti-inflammatory effects of ligustrazine [205]. Ligustrazine (100 μM) can suppress EMT of renal cell carcinoma cells by blocking MMP9 and TGF-1 [206].
Luteolin is a natural flavonoid that possesses anti-inflammatory and anti-cancer activities [207]. Luteolin (40 μM) inhibits TGF-1-induced EMT of A549 lung cancer cells through interfering with PI3K/AKT/NF-κB/Snail pathway [207]. Luteolin (10 μM) can suppress EMT and negatively regulating -catenin expression in breast cancer cells [208,209]. It (5–20 μM) can inhibit metastasis of melanoma cells by decreasing HIF-1α/VEGF signaling-mediated EMT [210]. It (5 μM) can also inhibit EMT of colorectal cancer cell by suppressing CREB1 expression [211]. Luteolin (20 μM) can inhibit the invasion of cervical cancer by blocking EMT signaling [212]. It (30 μM) can block gastric cancer progression by reversing EMT through inhibition of the notch signaling [213]. It (15.6–31.3 μM) can also inhibit EMT in paclitaxel-resistant ovarian cancer cells [214]. Luteolin (20–160 μM) can also block IL-6-induced EMT in pancreatic cancer cells by inhibiting STAT3 signaling [215].
Nimbolide, from the neem tree (Azadirachta indica), can inhibit NF-κB pathway in intestinal epithelial cells and macrophages, resulting in alleviation of experimental colitis in mice [216]. Nimbolide (5 μM) can suppress pancreatic cancer growth and metastasis through inhibition of EMT [217]. It (1–6 μM) can also suppress NSCLC cell invasion and migration via manipulation of DUSP4 expression and ERK1/2 signaling [218].
Nitidine chloride, a pentacyclic alkaloid isolated from the root of Zanthoxylum nitidium, can suppress LPS-induced interleukin production via MAPK and NF-κB in RAW 264.7 cells [219]. Hedgehog pathway is implicated in nitidine chloride (2.5 μM)-induced blocking of EMT of breast cancer cells [220]. Nitidine chloride (5 μM) can also inhibit EMT of osteosarcoma cell via Akt/GSK-3/snail signaling pathway [221].
Osthole, the major natural coumarin from Cnidium monnieri (L.) Cuss, exerts anti-inflammatory effects by blocking of the activation of the NF-κB and MAPK/p38 pathways [222]. Osthole (20 μM) can suppress HGF-induced EMT via repression of the c-Met/Akt/mTOR pathway in human breast cancer cells [223]. Osthole (20–40 μM) can also inhibit IGF-1-induced EMT by inhibiting PI3K/Akt signaling pathway in human brain cancer cells [224]. By inhibiting snail signaling and miR-23a-3p, osthole (20–80 μM) can suppress EMT-mediated metastatic ability in prostate cancer [225]. Osthole (5–20 μM) an also inhibit TGF -induced EMT by suppressing NF-κB mediated snail activation in A549 cells [226].
Oxymatrine, the active component from Radix Sophorae flavescentis, is well known for its anti-inflammatory activity [227]. Oxymatrine (1.5–6 μM) can reverse EMT of breast cancer cells by depressing αⅤ3 integrin/FAK/PI3K/Akt signaling activation [228]. It (0.25–0.75 μM) can inhibit EMT of colorectal cancer cells by suppressing NF-κB signaling [229]. Chronic oxymatrine treatment can induce resistance and EMT of colorectal cancer cells [230].
Paeoniflorin from Paeonia lactiflora has anti-inflammatory effects [231,232]. Paeoniflorin (5–10 μM) can suppress EMT of human colorectal cancer cells and glioblastoma cells and prevent hypoxia-induced EMT of human breast cancer cells [233,234].
Paeonol found in Paeonia suffruticosa (moutan cortex) can suppress LPS-induced HMGB1 translocation from the nucleus to the cytoplasm in RAW264.7 cells [235,236]. It (60–120 μM) can attenuate aging of MRC-5 cells and inhibit EMT of HaCaT cells induced by aging MRC-5 cell-conditioned medium [237].
Parthenolide from Tanacetum parthenium has well-known anti-inflammatory activities [238]. Parthenolide (5 μM) can inhibit TGF-1-induced EMT of colorectal cancer cells [239]. Parthenolide (10–20 μM) can suppress HIF-1α signaling and hypoxia-induced EMT in colorectal cancer [240]. Parthenolide binds Gly-Leu-Ser/Lys- “co-adaptation pocket” to inhibit EMT of lung cancer cell [241].
Plectranthoic acid isolated from Ficus microcarpa, can alleviate the symptoms of type 2 diabetes mellitus by inhibiting dipeptidyl peptidase 4 [242]. Plectranthoic acid is a novel activator of AMPK can induce apoptotic death in prostate cancer cells [243]. Plectranthoic acid (20–40 μM) can suppress EMT of prostate cancer [244].
Piperlongumine, a constituent of the fruit of the long pepper (Piper longum) can inhibit neuroinflammation via regulating NF-κB signaling pathways in LPS-stimulated BV2 microglia cells [245,246]. Piperlongumine (1–5 μM) inhibits TGF--induced EMT by modulating the expression of E-cadherin, Snail1, and Twist1 [247].
Plumbargin from Plumbago genus can attenuate the expression of inflammatory cytokine in LPS-activated BV-2 cells [248]. Plumbagin (0.5–1 μM) can inhibit EMT of human cervical carcinoma cells and inhibit tumor invasion of endocrine-resistant breast cancer through EMT [249]. It (0.1–0.5 μM) suppresses EMT via inhibiting Nrf2-mediated signaling pathway in human tongue squamous cell carcinoma cells [250]. Plumbagin (1–5 μM) can inhibit PI3K/Akt/mTOR-mediated EMT in human pancreatic cancer cells [251]. Plumbagin (1–5 μM) shows differential proteomic responses to EMT of PC-3 and DU145 prostate cancer cells [252].
Polyphyllin I, a component in the Rhizoma of Paris, can improve collagen-induced arthritis by blocking the inflammation response in macrophages through the NF-κB Pathway [253]. Polyphyllin I (0.3 μM) can overcome EMT-associated resistance to erlotinib in lung cancer cells via IL-6/STAT3 pathway inhibition [254].
Pterostilbene (5–10 μM) from blueberries can effectively suppress the generation of cancer stem cells and metastatic potential under the influence of M2 TAMs by modulating EMT associated signaling pathways, specifically the NF-κB/miR488 circuit [255]. Pterostilbene (10 μM) can also inhibit triple-negative breast cancer metastasis by inducing miR205 expression and negatively modulates EMT [256]. Long non-coding RNAs such as MEG3, TUG1, H19, and DICER1-AS1 contribute to the inhibitory effect of pterostilbene (1–50 μM) on proliferation and EMT of human breast cancer cells [257].
Resveratrol, a constituent of grapes and various other plants, is an activator of PPAR and SIRT1 [258]. Resveratrol (12 μM) can suppress TGF-1-induced EMT in colorectal cancer through the TGF-1/SMADs signaling pathway [259]. Resveratrol (20 μM) can inhibit TGF-1-induced EMT and suppress lung cancer invasion and metastasis [260]. Resveratrol (50 μM) can inhibit the EMT of pancreatic cancer cells through suppression of the PI3K/Akt/NF-κB pathway [261]. Resveratrol might inhibit EMT via downregulation of COX and SIRT1 activation, although roles of SIRT1 in EMT of cancer cells show conflicting results [262,263].
Salvianolic acid, an active compound present in Salvia miltiorrhiza, can suppress CCL-20 expression in TNF-α-treated macrophages [264]. Salvianolic acid B (1–100 μM)-induced expression of miR-106b-25 can suppress EMT of HK-2 cells [265]. Salvianolic acid B (1–10 μM) can prevent EMT through the TGF-1 signal transduction pathway [266].
α−Solanine is a glycoalkaloid poison found in species of the nightshade family within the genus Solanum, such as the potato and the eggplant [267]. A chloroform fraction of Solanum nigrum can suppresses nitric oxide and TNF-α in LPS-stimulated mouse peritoneal macrophages through inhibition of p38, JNK and ERK1/2 [268]. α-Solanine (4–12 μM) can suppress the invasion of human prostate cancer cell by inhibiting EMT and MMPs expression [269].
Sulforaphane from cruciferous vegetables may directly impair the formation of NLRP3 inflammasome by inhibiting ASC or caspase-1 [270]. Sulforaphane (1–5 μM) can also inhibit the EMT and metastasis of human lung cancer through miR616-5p-involved GSK3/-catenin pathways [271]. Sulforaphane (20–40 μM) can suppress TGF-1-induced EMT of HCC cells via the ROS-dependent pathway [272]. It (5–20 μM) can block the EMT of human bladder cancer cells via COX-2/MMP2,9/SNAIL, ZEB1, and miR200c/ZEB1 pathways [273].
Tannic acid is a type of polyphenol inhibits NLRP3 inflammasome-mediated IL-1 production via blocking NF-κB signaling in macrophages [274]. Tannic acid (25 μM) attenuates TGF-1-induced EMT in lung epithelial cells [275].
Withaferin A from the Solanaceae family attenuates bleomycin-induced scleroderma by targeting FoxO3a and NF-κB signaling [276]. Withaferin A (0.5 μM) can inhibit the EMT of NSCLC cells [277]. It (2 μM) can also inhibit the EMT of MCF10A cells and suppress vimentin expression in breast tumors [278]. Extracts of root in Withania somnifera can suppress mammary EMT and cancer metastasis [279].

4.2. Reversal of EMT by Natural Pro-resolving Lipids

Pro-resolving lipids can be useful agent against cancer and EMT of cancer has been studied since cancer is understood as a non-resolving disease [6,280]
Here, we briefly introduced the effect of pro-resolving lipids on EMT of cancer cells and emphasized natural sources of pro-resolving lipids (Table 2).
Pro-resolving lipids are generally known to act via GPCR receptors such as BLT1, CMKLR1, FPRL1 (ALX/FPR2), GPR18, and GPR3 [41,281]. CMKLR1 is a receptor with high affinity for RvE1, as measured via radioligand-binding assay (Kd = 11.3–5.4 nM) [282,283]. BLT1 is also a receptor with low affinity for RvE1, as measured via radioligand-binding assay (Kd = 45 nM) [284]. CMKLR1 has not been reported to be associated with EMT in cancer cells but seems to be associated with EMT in diabetic nephropathy [285]. FPRL1 (ALX/FPR2) not only possesses an annexin A1 [286] protein but also LxA4 (Kd = 1.7 nM, radioligand binding) [287], AT-RvD1 (EC50 = 1.8 × 10−10, -arrestin receptor system [288], RvD1 (Kd = 0.17 ± 0.06 nM, radioligand assay; EC50 = 4.5 × 10–11, -arrestin receptor system) [288], RvD3 (activation of FPRL1 at 100 nM) [289] and other similar proteins as a ligand. FPRL1 mediates EMT inhibition by LxA4 and RvD1 [290,291].
GRP18 has been reported as a receptor for RvD2 on performing GPCR--arrestin-based screening (Kd of 9.6 ± 0.9 nM, radioligand binding) [292]. GPR32 is known as the receptor of RvD1 (EC50 = 3.6 × 10−12, -arrestin receptor system) [293] and RvD5 (activation of GPR32 at the range of 10−13–10−9 M) [294]. GPR32 is also activated by RvD3 and AT-RvD3 (0.1 pM–10 nM) [295]; it mediates EMT Inhibition by RvD1 [291]. RvD1 also promotes wound healing in pulmonary epithelial cells and mediates EMT inhibition [296].
LxA4 (0.1–0.8 μM) can suppress TGF-1 signaling in pancreatic cancer cells, reverse mesenchymal features and block invasion and migration via a FPR2 [297]. LxA4 (0.01–0.1 μM) can also suppress estrogen-induced EMT via LxA4 receptor-dependent manner in endometriosis [298]. LxA4 (0.2 μM) and its analogue can suppress EMT, migration and metastasis of HCC by regulating integrin-linked kinase axis [299].
100 nM of RvD1 and RvD2 can inhibit TGF-1-induced EMT of A549 lung cancer cells through FPR2/ALXR and GPR32 [291]. GPR32 can recognize RvD1 (EC50 = 8.8 pM from -arrestin receptor system) as an endogenous ligand [293]. Aspirin-triggered RvD1 (10 ng/mL) can block TGF-1-induced EMT of A549 lung cancer cells via suppression of the mTOR pathway by reducing the expression of pyruvate kinase M2 [300]. RvD1 (400 nM) prevents EMT of HCC cells by inhibiting paracrine of cancer-associated fibroblast-derived cartilage oligomeric matrix protein [301].
MaR1 (0.1–10 ng/mice), a docosahexaenoic acid-derived pro-resolution lipid, can protect skin from inflammation and oxidative stress caused by UVB irradiation [302]. Receptor for MaR1 is yet unknown. The effect of MaR1 on EMT of cancer cells has not been reported yet except one study has shown that incubating DHA with A549 lung cancer cells can produce MaR1 (1.58 ng/mL) and PD1 (1.67 ng/mL) [303]. MaR1 (10 nM) can inhibit TGF-1-induced proliferation, migration, and differentiation in human lung fibroblasts [304].
PD1 has anti-inflammatory and survival effects on neuronal diseases such as Alzheimer’s disease and retinal degenerations [305]. PDX (1–100 nM), one of PD1 derivatives, can suppress bleomycin-induced lung fibrosis through blocking EMT [6,306]. However, it is hard to find reports about the effect of PDs on the EMT of cancer cells [6,306]. However, it is hard to find reports about the effect of PDs on the EMT of cancer cells.
Pro-resolving lipid classes are mainly originated from ω-3 fatty acids which constitute a group of essential fats that humans cannot synthesize endogenously [307]. Several pro-resolving lipids can be obtained by total synthesis. Studies on derivatives are in progress.
It is possible to produce pro-resolving lipids by biological methods. EPA and DHA are the major long chain ω-3 fatty acids in the diet. Algae are the major producers of EPA and DHA in the ecosystem. Therefore, fish that consumes algae contains a lot of EPA and DHA [307]. Accordingly, algae or fish might be a starting point for isolating or producing pro-resolving lipids in industrial scale. Brain cells of rainbow trout (Oncorhynchus mykiss) can produce novel DHA-derived Rvs and PDs (Table 2) [308]. However, baking reduces proportions of PG, Rv, and hydroxy-fatty acid in farm-raised Atlantic salmon (Salmo salar; Table 2) [309].
Infectious organisms can produce pro-resolving lipids to control host inflammation. Thus, supraphysiological levels of LxA4 are generated during infection by Toxoplasma gondii, which in turn reduces IL-12 production by dendritic cells, thus dampening Th1-type cell-mediated immune responses (Table 2) [310].
Candida albicans can modulate host defense by biosynthesizing the pro-resolving lipid RvE1 [311]. C. albicans can biosynthesize nanogram quantities of RvE1 from EPA without collaboration of other cellular partners. It can also biosynthesize PDs (Table 2) [311]. Trypanosoma cruzi is a protozoan parasite that causes Chagas disease and produces the RvD1, RvD5, and RvE2 (Table 2) [312]. These reports suggested that algae, fish, and some infectious organisms might be applied to produce pro-resolving lipids.

5. Perspectives

Several natural compounds have anti-inflammatory activities and/or anti-EMT activities. However, few reports have explained their anti-EMT activity by anti-inflammatory or pro-resolving mechanisms. Therefore, examining anti-EMT activities of natural compounds based on their anti-inflammatory or pro-resolving activities and assuring their anti-EMT activities in vivo might be important in the future. Especially, it might be a reasonable way to study the anti-EMT activity of natural products through interaction with the tumor microenvironment [255].
The flux of inflammatory or pro-resolving lipids from arachidonic acid related pathway (Figure 2) could change if one pathway is blocked by natural anti-inflammatory or pro-resolving compounds. Therefore, prevention of the production of these lipids might influence levels of other inflammatory and pro-resolving lipids. Thus, further studies on effects of anti-inflammatory compounds or pro-resolving lipids on EMT of cancer might require entire profiles of lipid metabolites affecting the EMT.

6. Conclusions

Expansion of studies about other pro-resolving lipids and nature-derived pro-resolving compounds to inhibit EMT are needed. To do this, various types of pro-resolving lipids and new compounds from natural sources should be procured and diffused to researcher without barrier. Thus, collaboration of natural chemistry researchers with other fields is also required.

Funding

This study was supported by grants (NRF-2017R1A2A1A05000878 and NRF-2018R1A5A2023127) of the Basic Science Research Program, through the National Research Foundation (NRF) of Korea.

Acknowledgments

The author apologizes to researchers in the EMT field whose work we are not able to cite here due to space limits.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Brabletz, T.; Kalluri, R.; Nieto, M.A.; Weinberg, R.A. EMT in cancer. Nat. Rev. Cancer 2018, 18, 128–134. [Google Scholar] [CrossRef] [PubMed]
  2. Marcucci, F.; Stassi, G.; De Maria, R. Epithelial-mesenchymal transition: A new target in anticancer drug discovery. Nat. Rev. Drug Discov. 2016, 15, 311–325. [Google Scholar] [CrossRef] [PubMed]
  3. Reka, A.K.; Kuick, R.; Kurapati, H.; Standiford, T.J.; Omenn, G.S.; Keshamouni, V.G. Identifying inhibitors of epithelial-mesenchymal transition by connectivity map-based systems approach. J. Thorac. Oncol. 2011, 6, 1784–1792. [Google Scholar] [CrossRef] [PubMed]
  4. David, J.M.; Dominguez, C.; Palena, C. Pharmacological and immunological targeting of tumor mesenchymalization. Pharmacol. Ther. 2017, 170, 212–225. [Google Scholar] [CrossRef]
  5. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  6. Lee, C.H. Epithelial-mesenchymal transition: Initiation by cues from chronic inflammatory tumor microenvironment and termination by anti-inflammatory compounds and specialized pro-resolving lipids. Biochem. Pharmacol. 2018, 158, 261–273. [Google Scholar] [CrossRef]
  7. Xu, J.; Lamouille, S.; Derynck, R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 2009, 19, 156–172. [Google Scholar] [CrossRef]
  8. Smith, C.L.; Baek, S.T.; Sung, C.Y.; Tallquist, M.D. Epicardial-derived cell epithelial-to-mesenchymal transition and fate specification require PDGF receptor signaling. Circ. Res. 2011, 108, e15–e26. [Google Scholar] [CrossRef]
  9. Strutz, F.; Zeisberg, M.; Ziyadeh, F.N.; Yang, C.Q.; Kalluri, R.; Muller, G.A.; Neilson, E.G. Role of basic fibroblast growth factor-2 in epithelial-mesenchymal transformation. Kidney Int. 2002, 61, 1714–1728. [Google Scholar] [CrossRef]
  10. Farrell, J.; Kelly, C.; Rauch, J.; Kida, K.; Garcia-Munoz, A.; Monsefi, N.; Turriziani, B.; Doherty, C.; Mehta, J.P.; Matallanas, D.; et al. HGF induces epithelial-to-mesenchymal transition by modulating the mammalian hippo/MST2 and ISG15 pathways. J. Proteome Res. 2014, 13, 2874–2886. [Google Scholar] [CrossRef]
  11. Liao, G.; Wang, M.; Ou, Y.; Zhao, Y. IGF-1-induced epithelial-mesenchymal transition in MCF-7 cells is mediated by MUC1. Cell Signal 2014, 26, 2131–2137. [Google Scholar] [CrossRef] [PubMed]
  12. Janse van Rensburg, H.J.; Yang, X. The roles of the Hippo pathway in cancer metastasis. Cell Signal 2016, 28, 1761–1772. [Google Scholar] [CrossRef] [PubMed]
  13. Hollier, B.G.; Tinnirello, A.A.; Werden, S.J.; Evans, K.W.; Taube, J.H.; Sarkar, T.R.; Sphyris, N.; Shariati, M.; Kumar, S.V.; Battula, V.L.; et al. FOXC2 expression links epithelial-mesenchymal transition and stem cell properties in breast cancer. Cancer Res. 2013, 73, 1981–1992. [Google Scholar] [CrossRef] [PubMed]
  14. Tang, Y.; Weiss, S.J. Snail/Slug-YAP/TAZ complexes cooperatively regulate mesenchymal stem cell function and bone formation. Cell Cycle 2017, 16, 399–405. [Google Scholar] [CrossRef] [PubMed]
  15. Lehmann, W.; Mossmann, D.; Kleemann, J.; Mock, K.; Meisinger, C.; Brummer, T.; Herr, R.; Brabletz, S.; Stemmler, M.P.; Brabletz, T. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat. Commun. 2016, 7, 10498. [Google Scholar] [CrossRef]
  16. Roselli, M.; Fernando, R.I.; Guadagni, F.; Spila, A.; Alessandroni, J.; Palmirotta, R.; Costarelli, L.; Litzinger, M.; Hamilton, D.; Huang, B.; et al. Brachyury, a driver of the epithelial-mesenchymal transition, is overexpressed in human lung tumors: An opportunity for novel interventions against lung cancer. Clin. Cancer Res. 2012, 18, 3868–3879. [Google Scholar] [CrossRef]
  17. Arnold, S.J.; Stappert, J.; Bauer, A.; Kispert, A.; Herrmann, B.G.; Kemler, R. Brachyury is a target gene of the Wnt/beta-catenin signaling pathway. Mech. Dev. 2000, 91, 249–258. [Google Scholar] [CrossRef]
  18. Zhang, J.; Liu, Y.; Zhang, J.; Cui, X.; Li, G.; Wang, J.; Ren, H.; Zhang, Y. FOXQ1 promotes gastric cancer metastasis through upregulation of Snail. Oncol. Rep. 2016, 35, 3607–3613. [Google Scholar] [CrossRef]
  19. Fujita, T.; Azuma, Y.; Fukuyama, R.; Hattori, Y.; Yoshida, C.; Koida, M.; Ogita, K.; Komori, T. Runx2 induces osteoblast and chondrocyte differentiation and enhances their migration by coupling with PI3K-Akt signaling. J. Cell Biol. 2004, 166, 85–95. [Google Scholar] [CrossRef]
  20. Cao, Z.; Sun, B.; Zhao, X.; Zhang, Y.; Gu, Q.; Liang, X.; Dong, X.; Zhao, N. The Expression and Functional Significance of Runx2 in Hepatocellular Carcinoma: Its Role in Vasculogenic Mimicry and Epithelial-Mesenchymal Transition. Int. J. Mol. Sci. 2017, 18, 500. [Google Scholar] [CrossRef]
  21. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [PubMed]
  22. Li, Y.; Ke, Q.; Shao, Y.; Zhu, G.; Li, Y.; Geng, N.; Jin, F.; Li, F. GATA1 induces epithelial-mesenchymal transition in breast cancer cells through PAK5 oncogenic signaling. Oncotarget 2015, 6, 4345–4356. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, X.; Xin, Z.; Zou, Z.; Lu, C.; Yu, Z.; Feng, S.; Pan, P.; Hao, G.; Dong, Y.; Yang, Y. SRY-related high-mobility-group box 4: Crucial regulators of the EMT in cancer. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef] [PubMed]
  24. Tiwari, N.; Tiwari, V.K.; Waldmeier, L.; Balwierz, P.J.; Arnold, P.; Pachkov, M.; Meyer-Schaller, N.; Schubeler, D.; van Nimwegen, E.; Christofori, G. Sox4 is a master regulator of epithelial-mesenchymal transition by controlling Ezh2 expression and epigenetic reprogramming. Cancer Cell 2013, 23, 768–783. [Google Scholar] [CrossRef]
  25. Jolly, M.K.; Boareto, M.; Huang, B.; Jia, D.; Lu, M.; Ben-Jacob, E.; Onuchic, J.N.; Levine, H. Implications of the Hybrid Epithelial/Mesenchymal Phenotype in Metastasis. Front. Oncol. 2015, 5, 155. [Google Scholar] [CrossRef]
  26. Elisha, Y.; Kalchenko, V.; Kuznetsov, Y.; Geiger, B. Dual role of E-cadherin in the regulation of invasive collective migration of mammary carcinoma cells. Sci. Rep. 2018, 8, 4986. [Google Scholar] [CrossRef]
  27. Park, S.Y.; Choi, J.H.; Nam, J.S. Targeting Cancer Stem Cells in Triple-Negative Breast Cancer. Cancers (Basel) 2019, 11, 965. [Google Scholar] [CrossRef]
  28. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
  29. Chaffer, C.L.; Marjanovic, N.D.; Lee, T.; Bell, G.; Kleer, C.G.; Reinhardt, F.; D’Alessio, A.C.; Young, R.A.; Weinberg, R.A. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 2013, 154, 61–74. [Google Scholar] [CrossRef]
  30. Nieto, M.A.; Huang, R.Y.-J.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef]
  31. Du, B.; Shim, J.S. Targeting Epithelial-Mesenchymal Transition (EMT) to Overcome Drug Resistance in Cancer. Molecules 2016, 21, 965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.; Choi, H.; El Rayes, T.; Ryu, S.; Troeger, J.; et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef] [PubMed]
  33. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
  34. Hu, B.; Cheng, J.W.; Hu, J.W.; Li, H.; Ma, X.L.; Tang, W.G.; Sun, Y.F.; Guo, W.; Huang, A.; Zhou, K.Q.; et al. KPNA3 Confers Sorafenib Resistance to Advanced Hepatocellular Carcinoma via TWIST Regulated Epithelial-Mesenchymal Transition. J. Cancer 2019, 10, 3914–3925. [Google Scholar] [CrossRef] [PubMed]
  35. Akalay, I.; Janji, B.; Hasmim, M.; Noman, M.Z.; Andre, F.; De Cremoux, P.; Bertheau, P.; Badoual, C.; Vielh, P.; Larsen, A.K.; et al. Epithelial-to-mesenchymal transition and autophagy induction in breast carcinoma promote escape from T-cell-mediated lysis. Cancer Res. 2013, 73, 2418–2427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kudo-Saito, C.; Shirako, H.; Takeuchi, T.; Kawakami, Y. Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 2009, 15, 195–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ricciardi, M.; Zanotto, M.; Malpeli, G.; Bassi, G.; Perbellini, O.; Chilosi, M.; Bifari, F.; Krampera, M. Epithelial-to-mesenchymal transition (EMT) induced by inflammatory priming elicits mesenchymal stromal cell-like immune-modulatory properties in cancer cells. Br. J. Cancer 2015, 112, 1067–1075. [Google Scholar] [CrossRef]
  38. Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat. Commun. 2014, 5, 5241. [Google Scholar] [CrossRef]
  39. Reiman, J.M.; Knutson, K.L.; Radisky, D.C. Immune promotion of epithelial-mesenchymal transition and generation of breast cancer stem cells. Cancer Res. 2010, 70, 3005–3008. [Google Scholar] [CrossRef] [Green Version]
  40. Fullerton, J.N.; Gilroy, D.W. Resolution of inflammation: A new therapeutic frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567. [Google Scholar] [CrossRef]
  41. Lee, C.H. Resolvins as new fascinating drug candidates for inflammatory diseases. Arch. Pharm. Res. 2012, 35, 3–7. [Google Scholar] [CrossRef] [PubMed]
  42. Mariathasan, S.; Newton, K.; Monack, D.M.; Vucic, D.; French, D.M.; Lee, W.P.; Roose-Girma, M.; Erickson, S.; Dixit, V.M. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 2004, 430, 213–218. [Google Scholar] [CrossRef] [PubMed]
  43. Guo, H.; Callaway, J.B.; Ting, J.P. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015, 21, 677–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Martinon, F.; Burns, K.; Tschopp, J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  45. Song, S.; Qiu, D.; Luo, F.; Wei, J.; Wu, M.; Wu, H.; Du, C.; Du, Y.; Ren, Y.; Chen, N.; et al. Knockdown of NLRP3 alleviates high glucose or TGFB1-induced EMT in human renal tubular cells. J. Mol. Endocrinol. 2018, 61, 101–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Yin, X.F.; Zhang, Q.; Chen, Z.Y.; Wang, H.F.; Li, X.; Wang, H.X.; Li, H.X.; Kang, C.M.; Chu, S.; Li, K.F.; et al. NLRP3 in human glioma is correlated with increased WHO grade, and regulates cellular proliferation, apoptosis and metastasis via epithelial-mesenchymal transition and the PTEN/AKT signaling pathway. Int. J. Oncol. 2018, 53, 973–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Li, X.; Yan, X.; Wang, Y.; Wang, J.; Zhou, F.; Wang, H.; Xie, W.; Kong, H. NLRP3 inflammasome inhibition attenuates silica-induced epithelial to mesenchymal transition (EMT) in human bronchial epithelial cells. Exp. Cell Res. 2018, 362, 489–497. [Google Scholar] [CrossRef]
  48. Tian, R.; Zhu, Y.; Yao, J.; Meng, X.; Wang, J.; Xie, H.; Wang, R. NLRP3 participates in the regulation of EMT in bleomycin-induced pulmonary fibrosis. Exp. Cell Res. 2017, 357, 328–334. [Google Scholar] [CrossRef]
  49. Romero, C.A.; Remor, A.; Latini, A.; De Paul, A.L.; Torres, A.I.; Mukdsi, J.H. Uric acid activates NRLP3 inflammasome in an in-vivo model of epithelial to mesenchymal transition in the kidney. J. Mol. Histol. 2017, 48, 209–218. [Google Scholar] [CrossRef]
  50. Wang, H.; Wang, Y.; Du, Q.; Lu, P.; Fan, H.; Lu, J.; Hu, R. Inflammasome-independent NLRP3 is required for epithelial-mesenchymal transition in colon cancer cells. Exp. Cell Res. 2016, 342, 184–192. [Google Scholar] [CrossRef]
  51. Serhan, C.N.; Chiang, N.; Dalli, J. The resolution code of acute inflammation: Novel pro-resolving lipid mediators in resolution. Semin. Immunol. 2015, 27, 200–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Serhan, C.N.; Chiang, N.; Dalli, J. New pro-resolving n-3 mediators bridge resolution of infectious inflammation to tissue regeneration. Mol. Aspects Med. 2018, 64, 1–17. [Google Scholar] [CrossRef] [PubMed]
  53. Dalli, J.; Chiang, N.; Serhan, C.N. Elucidation of novel 13-series resolvins that increase with atorvastatin and clear infections. Nat. Med. 2015, 21, 1071–1075. [Google Scholar] [CrossRef] [PubMed]
  54. Dominguez, C.; David, J.M.; Palena, C. Epithelial-mesenchymal transition and inflammation at the site of the primary tumor. Semin. Cancer Biol. 2017, 47, 177–184. [Google Scholar] [CrossRef] [PubMed]
  55. Lou, Y.; Diao, L.; Cuentas, E.R.; Denning, W.L.; Chen, L.; Fan, Y.H.; Byers, L.A.; Wang, J.; Papadimitrakopoulou, V.A.; Behrens, C.; et al. Epithelial-Mesenchymal Transition Is Associated with a Distinct Tumor Microenvironment Including Elevation of Inflammatory Signals and Multiple Immune Checkpoints in Lung Adenocarcinoma. Clin. Cancer Res. 2016, 22, 3630–3642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Bradley, J.R. TNF-mediated inflammatory disease. J. Pathol. 2008, 214, 149–160. [Google Scholar] [CrossRef]
  58. Sheen-Chen, S.M.; Chen, W.J.; Eng, H.L.; Chou, F.F. Serum concentration of tumor necrosis factor in patients with breast cancer. Breast Cancer Res. Treat. 1997, 43, 211–215. [Google Scholar] [CrossRef]
  59. Bates, R.C.; Mercurio, A.M. Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids. Mol. Biol. Cell 2003, 14, 1790–1800. [Google Scholar] [CrossRef] [Green Version]
  60. Kozlowski, L.; Zakrzewska, I.; Tokajuk, P.; Wojtukiewicz, M.Z. Concentration of interleukin-6 (IL-6), interleukin-8 (IL-8) and interleukin-10 (IL-10) in blood serum of breast cancer patients. Rocz. Akad. Med. Bialymst. 2003, 48, 82–84. [Google Scholar]
  61. Sullivan, N.J.; Sasser, A.K.; Axel, A.E.; Vesuna, F.; Raman, V.; Ramirez, N.; Oberyszyn, T.M.; Hall, B.M. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene 2009, 28, 2940–2947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Wu, J.; Zhang, J.; Shen, B.; Yin, K.; Xu, J.; Gao, W.; Zhang, L. Long noncoding RNA lncTCF7, induced by IL-6/STAT3 transactivation, promotes hepatocellular carcinoma aggressiveness through epithelial-mesenchymal transition. J. Exp. Clin. Cancer Res. 2015, 34, 116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yamada, D.; Kobayashi, S.; Wada, H.; Kawamoto, K.; Marubashi, S.; Eguchi, H.; Ishii, H.; Nagano, H.; Doki, Y.; Mori, M. Role of crosstalk between interleukin-6 and transforming growth factor-beta 1 in epithelial-mesenchymal transition and chemoresistance in biliary tract cancer. Eur. J. Cancer 2013, 49, 1725–1740. [Google Scholar] [CrossRef] [PubMed]
  64. Bates, R.C.; DeLeo, M.J., 3rd; Mercurio, A.M. The epithelial-mesenchymal transition of colon carcinoma involves expression of IL-8 and CXCR-1-mediated chemotaxis. Exp. Cell Res. 2004, 299, 315–324. [Google Scholar] [CrossRef] [PubMed]
  65. Li, X.-J.; Peng, L.-X.; Shao, J.-Y.; Lu, W.-H.; Zhang, J.-X.; Chen, S.; Chen, Z.-Y.; Xiang, Y.-Q.; Bao, Y.-N.; Zheng, F.-J. As an independent unfavorable prognostic factor, IL-8 promotes metastasis of nasopharyngeal carcinoma through induction of epithelial–mesenchymal transition and activation of AKT signaling. Carcinogenesis 2012, 33, 1302–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Visciano, C.; Liotti, F.; Prevete, N.; Franco, R.; Collina, F.; De Paulis, A.; Marone, G.; Santoro, M.; Melillo, R. Mast cells induce epithelial-to-mesenchymal transition and stem cell features in human thyroid cancer cells through an IL-8-Akt-Slug pathway. Oncogene 2015, 34, 5175–5186. [Google Scholar] [CrossRef]
  67. Qu, J.; Cheng, T.; Liu, L.; Heng, J.; Liu, X.; Sun, Z.; Wang, W.; Li, K.; Yang, N. Mast cells induce epithelial-to-mesenchymal transition and migration in non-small cell lung cancer through IL-8/Wnt/beta-catenin pathway. J. Cancer 2019, 10, 3830–3841. [Google Scholar] [CrossRef]
  68. Fu, X.T.; Dai, Z.; Song, K.; Zhang, Z.J.; Zhou, Z.J.; Zhou, S.L.; Zhao, Y.M.; Xiao, Y.S.; Sun, Q.M.; Ding, Z.B.; et al. Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway. Int. J. Oncol. 2015, 46, 587–596. [Google Scholar] [CrossRef] [Green Version]
  69. Palena, C.; Hamilton, D.H.; Fernando, R.I. Influence of IL-8 on the epithelial-mesenchymal transition and the tumor microenvironment. Future Oncol. 2012, 8, 713–722. [Google Scholar] [CrossRef] [Green Version]
  70. Rouvier, E.; Luciani, M.F.; Mattei, M.G.; Denizot, F.; Golstein, P. CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to a herpesvirus saimiri gene. J. Immunol. 1993, 150, 5445–5456. [Google Scholar]
  71. Zhang, Q.; Liu, S.; Parajuli, K.; Zhang, W.; Zhang, K.; Mo, Z.; Liu, J.; Chen, Z.; Yang, S.; Wang, A. Interleukin-17 promotes prostate cancer via MMP7-induced epithelial-to-mesenchymal transition. Oncogene 2017, 36, 687–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Huang, Q.; Han, J.; Fan, J.; Duan, L.; Guo, M.; Lv, Z.; Hu, G.; Chen, L.; Wu, F.; Tao, X.; et al. IL-17 induces EMT via Stat3 in lung adenocarcinoma. Am. J. Cancer Res. 2016, 6, 440–451. [Google Scholar] [PubMed]
  73. Xiang, T.; Long, H.; He, L.; Han, X.; Lin, K.; Liang, Z.; Zhuo, W.; Xie, R.; Zhu, B. Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene 2015, 34, 165–176. [Google Scholar] [CrossRef] [PubMed]
  74. Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Balasubramaniam, V.; Othman, I.; Shaikh, M.F. Enlightening the role of high mobility group box 1 (HMGB1) in inflammation: Updates on receptor signalling. Eur. J. Pharmacol. 2019, 858, 172487. [Google Scholar] [CrossRef] [PubMed]
  75. Sun, S.; Zhang, W.; Cui, Z.; Chen, Q.; Xie, P.; Zhou, C.; Liu, B.; Peng, X.; Zhang, Y. High mobility group box-1 and its clinical value in breast cancer. OncoTargets Ther. 2015, 8, 413–419. [Google Scholar] [CrossRef] [Green Version]
  76. Zhang, J.; Shao, S.; Han, D.; Xu, Y.; Jiao, D.; Wu, J.; Yang, F.; Ge, Y.; Shi, S.; Li, Y.; et al. High mobility group box 1 promotes the epithelial-to-mesenchymal transition in prostate cancer PC3 cells via the RAGE/NF-kappaB signaling pathway. Int. J. Oncol. 2018, 53, 659–671. [Google Scholar] [CrossRef]
  77. Zhu, L.; Li, X.; Chen, Y.; Fang, J.; Ge, Z. High-mobility group box 1: A novel inducer of the epithelial-mesenchymal transition in colorectal carcinoma. Cancer Lett. 2015, 357, 527–534. [Google Scholar] [CrossRef]
  78. Moorre, K.; OíGarra, A.; de Waal Malefyt, R.; Viera, P.; Mosmann, T. Interleukin-10. Annu. Rev. Imunol. 1993, 11, 165–190. [Google Scholar] [CrossRef]
  79. Liu, C.Y.; Xu, J.Y.; Shi, X.Y.; Huang, W.; Ruan, T.Y.; Xie, P.; Ding, J.L. M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab. Invest. 2013, 93, 844–854. [Google Scholar] [CrossRef] [Green Version]
  80. Miyazono, K. Transforming growth factor-beta signaling in epithelial-mesenchymal transition and progression of cancer. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 314–323. [Google Scholar] [CrossRef] [Green Version]
  81. Ivanovic, V.; Todorovic-Rakovic, N.; Demajo, M.; Neskovic-Konstantinovic, Z.; Subota, V.; Ivanisevic-Milovanovic, O.; Nikolic-Vukosavljevic, D. Elevated plasma levels of transforming growth factor-beta 1 (TGF-beta 1) in patients with advanced breast cancer: Association with disease progression. Eur. J. Cancer 2003, 39, 454–461. [Google Scholar] [CrossRef]
  82. Ahmadi, A.; Najafi, M.; Farhood, B.; Mortezaee, K. Transforming growth factor-beta signaling: Tumorigenesis and targeting for cancer therapy. J. Cell Physiol. 2019, 234, 12173–12187. [Google Scholar] [CrossRef] [PubMed]
  83. Maeda, H.; Kuwahara, H.; Ichimura, Y.; Ohtsuki, M.; Kurakata, S.; Shiraishi, A. TGF-beta enhances macrophage ability to produce IL-10 in normal and tumor-bearing mice. J. Immunol. 1995, 155, 4926–4932. [Google Scholar] [PubMed]
  84. Li, Y.; Wang, P.; Zhao, J.; Li, H.; Liu, D.; Zhu, W. HMGB1 attenuates TGF-beta-induced epithelial-mesenchymal transition of FaDu hypopharyngeal carcinoma cells through regulation of RAGE expression. Mol. Cell Biochem. 2017, 431, 1–10. [Google Scholar] [CrossRef] [PubMed]
  85. Loh, J.K.; Hwang, S.L.; Lieu, A.S.; Huang, T.Y.; Howng, S.L. The alteration of prostaglandin E2 levels in patients with brain tumors before and after tumor removal. J. Neurooncol. 2002, 57, 147–150. [Google Scholar] [CrossRef] [PubMed]
  86. O’Callaghan, G.; Houston, A. Prostaglandin E2 and the EP receptors in malignancy: Possible therapeutic targets? Br. J. Pharmacol. 2015, 172, 5239–5250. [Google Scholar] [CrossRef] [Green Version]
  87. Dohadwala, M.; Yang, S.C.; Luo, J.; Sharma, S.; Batra, R.K.; Huang, M.; Lin, Y.; Goodglick, L.; Krysan, K.; Fishbein, M.C.; et al. Cyclooxygenase-2-dependent regulation of E-cadherin: Prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res. 2006, 66, 5338–5345. [Google Scholar] [CrossRef] [Green Version]
  88. Mann, J.R.; Backlund, M.G.; Buchanan, F.G.; Daikoku, T.; Holla, V.R.; Rosenberg, D.W.; Dey, S.K.; DuBois, R.N. Repression of prostaglandin dehydrogenase by epidermal growth factor and snail increases prostaglandin E2 and promotes cancer progression. Cancer Res. 2006, 66, 6649–6656. [Google Scholar] [CrossRef] [Green Version]
  89. Ha, B.; Lee, E.B.; Cui, J.; Kim, Y.; Jang, H.H. YB-1 overexpression promotes a TGF-beta1-induced epithelial-mesenchymal transition via Akt activation. Biochem. Biophys. Res. Commun. 2015, 458, 347–351. [Google Scholar] [CrossRef]
  90. Zhang, A.; Wang, M.-H.; Dong, Z.; Yang, T. Prostaglandin E2 is a potent inhibitor of epithelial-to-mesenchymal transition: Interaction with hepatocyte growth factor. Am. J. Physiol. Renal. Physiol. 2006, 291, F1323–F1331. [Google Scholar] [CrossRef] [Green Version]
  91. Takai, E.; Tsukimoto, M.; Kojima, S. TGF-1 downregulates COX-2 expression leading to decrease of PGE2 production in human lung cancer A549 cells, which is involved in fibrotic response to TGF-1. PLoS ONE 2013, 8, e76346. [Google Scholar] [CrossRef] [Green Version]
  92. Carpagnano, G.E.; Palladino, G.P.; Lacedonia, D.; Koutelou, A.; Orlando, S.; Foschino-Barbaro, M.P. Neutrophilic airways inflammation in lung cancer: The role of exhaled LTB-4 and IL-8. BMC Cancer 2011, 11, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Park, M.K.; Park, Y.; Shim, J.; Lee, H.J.; Kim, S.; Lee, C.H. Novel involvement of leukotriene B4 receptor 2 through ERK activation by PP2A down-regulation in leukotriene B4-induced keratin phosphorylation and reorganization of pancreatic cancer cells. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2012, 1823, 2120–2129. [Google Scholar] [CrossRef] [Green Version]
  94. Kim, H.; Choi, J.A.; Kim, J.H. Ras promotes transforming growth factor-beta (TGF-beta)-induced epithelial-mesenchymal transition via a leukotriene B4 receptor-2-linked cascade in mammary epithelial cells. J. Biol. Chem. 2014, 289, 22151–22160. [Google Scholar] [CrossRef] [Green Version]
  95. Kim, Y.R.; Park, M.K.; Kang, G.J.; Kim, H.J.; Kim, E.J.; Byun, H.J.; Lee, M.Y.; Lee, C.H. Leukotriene B4 induces EMT and vimentin expression in PANC-1 pancreatic cancer cells: Involvement of BLT2 via ERK2 activation. Prostaglandins Leukot. Essent. Fatty Acids 2016, 115, 67–76. [Google Scholar] [CrossRef] [PubMed]
  96. Burke, L.; Butler, C.T.; Murphy, A.; Moran, B.; Gallagher, W.M.; O’Sullivan, J.; Kennedy, B.N. Evaluation of Cysteinyl Leukotriene Signaling as a Therapeutic Target for Colorectal Cancer. Front. Cell Dev. Biol. 2016, 4, 103. [Google Scholar] [CrossRef] [Green Version]
  97. Zhou, Y.; Guo, D.; Li, H.; Jie, S. Circulating LTD4 in patients with hepatocellular carcinoma. Tumor Biol. 2011, 32, 139–144. [Google Scholar] [CrossRef] [PubMed]
  98. Kanaoka, Y.; Boyce, J.A. Cysteinyl leukotrienes and their receptors; emerging concepts. Allergy Asthma Immunol. Res. 2014, 6, 288–295. [Google Scholar] [CrossRef]
  99. Hosoki, K.; Kainuma, K.; Toda, M.; Harada, E.; Chelakkot-Govindalayathila, A.L.; Roeen, Z.; Nagao, M.; D’Alessandro-Gabazza, C.N.; Fujisawa, T.; Gabazza, E.C. Montelukast suppresses epithelial to mesenchymal transition of bronchial epithelial cells induced by eosinophils. Biochem. Biophys. Res. Commun. 2014, 449, 351–356. [Google Scholar] [CrossRef]
  100. Lukic, A.; Wahlund, C.J.E.; Gomez, C.; Brodin, D.; Samuelsson, B.; Wheelock, C.E.; Gabrielsson, S.; Radmark, O. Exosomes and cells from lung cancer pleural exudates transform LTC4 to LTD4, promoting cell migration and survival via CysLT1. Cancer Lett. 2019, 444, 1–8. [Google Scholar] [CrossRef]
  101. Kang, H.S.; Lee, J.Y.; Kim, C.J. Anti-inflammatory activity of arctigenin from Forsythiae Fructus. J. Ethnopharmacol. 2008, 116, 305–312. [Google Scholar] [CrossRef] [PubMed]
  102. Xu, Y.; Lou, Z.; Lee, S.H. Arctigenin represses TGF-beta-induced epithelial mesenchymal transition in human lung cancer cells. Biochem. Biophys. Res. Commun. 2017, 493, 934–939. [Google Scholar] [CrossRef] [PubMed]
  103. Li, A.; Wang, J.; Zhu, D.; Zhang, X.; Pan, R.; Wang, R. Arctigenin suppresses transforming growth factor-beta1-induced expression of monocyte chemoattractant protein-1 and the subsequent epithelial-mesenchymal transition through reactive oxygen species-dependent ERK/NF-kappaB signaling pathway in renal tubular epithelial cells. Free Radic. Res. 2015, 49, 1095–1113. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, X.; Yang, J.; Zou, H. Baicalin inhibits IL-17-mediated joint inflammation in murine adjuvant-induced arthritis. Clin. Dev. Immunol. 2013, 2013, 268065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Chung, H.; Choi, H.S.; Seo, E.K.; Kang, D.H.; Oh, E.S. Baicalin and baicalein inhibit transforming growth factor-beta1-mediated epithelial-mesenchymal transition in human breast epithelial cells. Biochem. Biophys. Res. Commun. 2015, 458, 707–713. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, Y.; Wang, H.; Zhou, R.; Zhong, W.; Lu, S.; Ma, Z.; Chai, Y. Baicalin inhibits human osteosarcoma cells invasion, metastasis, and anoikis resistance by suppressing the transforming growth factor-beta1-induced epithelial-to-mesenchymal transition. Anticancer Drugs 2017, 28, 581–587. [Google Scholar] [CrossRef]
  107. Zhou, T.; Zhang, A.; Kuang, G.; Gong, X.; Jiang, R.; Lin, D.; Li, J.; Li, H.; Zhang, X.; Wan, J.; et al. Baicalin inhibits the metastasis of highly aggressive breast cancer cells by reversing epithelial-to-mesenchymal transition by targeting beta-catenin signaling. Oncol. Rep. 2017, 38, 3599–3607. [Google Scholar] [CrossRef] [Green Version]
  108. Jin, Y.; Khadka, D.B.; Cho, W.J. Pharmacological effects of berberine and its derivatives: A patent update. Expert Opin. Ther. Pat. 2016, 26, 229–243. [Google Scholar] [CrossRef]
  109. Zhang, Y.; Li, X.; Zhang, Q.; Li, J.; Ju, J.; Du, N.; Liu, X.; Chen, X.; Cheng, F.; Yang, L.; et al. Berberine hydrochloride prevents postsurgery intestinal adhesion and inflammation in rats. J. Pharmacol. Exp. Ther. 2014, 349, 417–426. [Google Scholar] [CrossRef] [Green Version]
  110. Chu, S.C.; Yu, C.C.; Hsu, L.S.; Chen, K.S.; Su, M.Y.; Chen, P.N. Berberine reverses epithelial-to-mesenchymal transition and inhibits metastasis and tumor-induced angiogenesis in human cervical cancer cells. Mol. Pharmacol. 2014, 86, 609–623. [Google Scholar] [CrossRef] [Green Version]
  111. Liu, C.H.; Tang, W.C.; Sia, P.; Huang, C.C.; Yang, P.M.; Wu, M.H.; Lai, I.L.; Lee, K.H. Berberine inhibits the metastatic ability of prostate cancer cells by suppressing epithelial-to-mesenchymal transition (EMT)-associated genes with predictive and prognostic relevance. Int. J. Med. Sci. 2015, 12, 63–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Wang, J.; Kang, M.; Wen, Q.; Qin, Y.T.; Wei, Z.X.; Xiao, J.J.; Wang, R.S. Berberine sensitizes nasopharyngeal carcinoma cells to radiation through inhibition of Sp1 and EMT. Oncol. Rep. 2017, 37, 2425–2432. [Google Scholar] [CrossRef] [PubMed]
  113. Harmer, R. Occurrence, chemistry and application of betanin. Food Chem. 1980, 5, 81–90. [Google Scholar] [CrossRef]
  114. Martinez, R.M.; Longhi-Balbinot, D.T.; Zarpelon, A.C.; Staurengo-Ferrari, L.; Baracat, M.M.; Georgetti, S.R.; Sassonia, R.C.; Verri, W.A., Jr.; Casagrande, R. Anti-inflammatory activity of betalain-rich dye of Beta vulgaris: Effect on edema, leukocyte recruitment, superoxide anion and cytokine production. Arch. Pharm. Res. 2015, 38, 494–504. [Google Scholar] [CrossRef] [PubMed]
  115. Sutariya, B.; Taneja, N.; Badgujar, L.; Saraf, M. Modulatory effect of betanin on high glucose induced epithelial to mesenchymal transition in renal proximal tubular cells. Biomed. Pharmacother. 2017, 89, 18–28. [Google Scholar] [CrossRef] [PubMed]
  116. Turpaev, K.; Welsh, N. Brusatol inhibits the response of cultured beta-cells to pro-inflammatory cytokines in vitro. Biochem. Biophys. Res. Commun. 2015, 460, 868–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Lu, Z.; Lai, Z.Q.; Leung, A.W.N.; Leung, P.S.; Li, Z.S.; Lin, Z.X. Exploring brusatol as a new anti-pancreatic cancer adjuvant: Biological evaluation and mechanistic studies. Oncotarget 2017, 8, 84974–84985. [Google Scholar] [CrossRef]
  118. Park, M.K.; Lee, H.J.; Choi, J.K.; Kim, H.J.; Kang, J.H.; Lee, E.J.; Kim, Y.R.; Kang, J.H.; Yoo, J.K.; Cho, H.Y.; et al. Novel anti-nociceptive effects of cardamonin via blocking expression of cyclooxygenase-2 and transglutaminase-2. Pharmacol. Biochem. Behav. 2014, 118, 10–15. [Google Scholar] [CrossRef]
  119. Park, M.K.; Choi, J.K.; Kim, H.J.; Nakahata, N.; Lim, K.M.; Kim, S.Y.; Lee, C.H. Novel inhibitory effects of cardamonin on thromboxane A2-induced scratching response: Blocking of Gh/transglutaminase-2 binding to thromboxane A2 receptor. Pharmacol. Biochem. Behav. 2014, 126, 131–135. [Google Scholar] [CrossRef]
  120. Kim, E.J.; Kim, H.J.; Park, M.K.; Kang, G.J.; Byun, H.J.; Lee, H.; Lee, C.H. Cardamonin Suppresses TGF-beta1-Induced Epithelial Mesenchymal Transition via Restoring Protein Phosphatase 2A Expression. Biomol. Ther. (Seoul) 2015, 23, 141–148. [Google Scholar] [CrossRef] [Green Version]
  121. Shrivastava, S.; Jeengar, M.K.; Thummuri, D.; Koval, A.; Katanaev, V.L.; Marepally, S.; Naidu, V.G.M. Cardamonin, a chalcone, inhibits human triple negative breast cancer cell invasiveness by downregulation of Wnt/beta-catenin signaling cascades and reversal of epithelial-mesenchymal transition. Biofactors 2017, 43, 152–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Park, M.K.; Jo, S.H.; Lee, H.J.; Kang, J.H.; Kim, Y.R.; Kim, H.J.; Lee, E.J.; Koh, J.Y.; Ahn, K.O.; Jung, K.C.; et al. Novel suppressive effects of cardamonin on the activity and expression of transglutaminase-2 lead to blocking the migration and invasion of cancer cells. Life Sci. 2013, 92, 154–160. [Google Scholar] [CrossRef] [PubMed]
  123. Lo, A.H.; Liang, Y.C.; Lin-Shiau, S.Y.; Ho, C.T.; Lin, J.K. Carnosol, an antioxidant in rosemary, suppresses inducible nitric oxide synthase through down-regulating nuclear factor-kappaB in mouse macrophages. Carcinogenesis 2002, 23, 983–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Guerrero, I.C.; Andres, L.S.; Leon, L.G.; Machin, R.P.; Padron, J.M.; Luis, J.G.; Delgadillo, J. Abietane diterpenoids from Salvia pachyphylla and S. clevelandii with cytotoxic activity against human cancer cell lines. J. Nat. Prod. 2006, 69, 1803–1805. [Google Scholar] [CrossRef] [PubMed]
  125. Yeo, I.J.; Park, J.H.; Jang, J.S.; Lee, D.Y.; Park, J.E.; Choi, Y.E.; Joo, J.H.; Song, J.K.; Jeon, H.O.; Hong, J.T. Inhibitory effect of Carnosol on UVB-induced inflammation via inhibition of STAT3. Arch. Pharm Res. 2019, 42, 274–283. [Google Scholar] [CrossRef] [Green Version]
  126. Kashyap, D.; Kumar, G.; Sharma, A.; Sak, K.; Tuli, H.S.; Mukherjee, T.K. Mechanistic insight into carnosol-mediated pharmacological effects: Recent trends and advancements. Life Sci. 2017, 169, 27–36. [Google Scholar] [CrossRef]
  127. Giacomelli, C.; Daniele, S.; Natali, L.; Iofrida, C.; Flamini, G.; Braca, A.; Trincavelli, M.L.; Martini, C. Carnosol controls the human glioblastoma stemness features through the epithelial-mesenchymal transition modulation and the induction of cancer stem cell apoptosis. Sci. Rep. 2017, 7, 15174. [Google Scholar] [CrossRef]
  128. Zhao, H.; Wang, Z.; Tang, F.; Zhao, Y.; Feng, D.; Li, Y.; Hu, Y.; Wang, C.; Zhou, J.; Tian, X.; et al. Carnosol-mediated Sirtuin 1 activation inhibits Enhancer of Zeste Homolog 2 to attenuate liver fibrosis. Pharmacol. Res. 2018, 128, 327–337. [Google Scholar] [CrossRef]
  129. Venkatesha, S.H.; Moudgil, K.D. Celastrol suppresses experimental autoimmune encephalomyelitis via MAPK/SGK1-regulated mediators of autoimmune pathology. Inflamm. Res. 2019, 68, 285–296. [Google Scholar] [CrossRef]
  130. Kang, H.; Lee, M.; Jang, S.W. Celastrol inhibits TGF-beta1-induced epithelial-mesenchymal transition by inhibiting Snail and regulating E-cadherin expression. Biochem. Biophys. Res. Commun. 2013, 437, 550–556. [Google Scholar] [CrossRef]
  131. Kashyap, D.; Sharma, A.; Tuli, H.S.; Sak, K.; Mukherjee, T.; Bishayee, A. Molecular targets of celastrol in cancer: Recent trends and advancements. Crit. Rev. Oncol. Hematol. 2018, 128, 70–81. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, S.; Cai, R.; Ma, J.; Liu, T.; Ke, X.; Lu, H.; Fu, J. The natural compound codonolactone impairs tumor induced angiogenesis by downregulating BMP signaling in endothelial cells. Phytomedicine 2015, 22, 1017–1026. [Google Scholar] [CrossRef] [PubMed]
  133. Liu, C.; Zhao, H.; Ji, Z.H.; Yu, X.Y. Neuroprotection of atractylenolide III from Atractylodis macrocephalae against glutamate-induced neuronal apoptosis via inhibiting caspase signaling pathway. Neurochem. Res. 2014, 39, 1753–1758. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, W.; Chen, B.; Zou, R.; Tu, X.; Tan, S.; Lu, H.; Liu, Z.; Fu, J. Codonolactone, a sesquiterpene lactone isolated from Chloranthus henryi Hemsl, inhibits breast cancer cell invasion, migration and metastasis by downregulating the transcriptional activity of Runx2. Int. J. Oncol. 2014, 45, 1891–1900. [Google Scholar] [CrossRef] [PubMed]
  135. Cunningham, K.G.; Manson, W.; Spring, F.S.; Hutchinson, S.A. Cordycepin, a metabolic product isolated from cultures of Cordyceps militaris (Linn.) Link. Nature 1950, 166, 949. [Google Scholar] [CrossRef]
  136. Qing, R.; Huang, Z.; Tang, Y.; Xiang, Q.; Yang, F. Cordycepin negatively modulates lipopolysaccharide-induced cytokine production by up-regulation of heme oxygenase-1. Int. Immunopharmacol. 2017, 47, 20–27. [Google Scholar] [CrossRef]
  137. Wang, C.W.; Lee, B.H.; Tai, C.J. The inhibition of cordycepin on cancer stemness in TGF-beta induced chemo-resistant ovarian cancer cell. Oncotarget 2017, 8, 111912–111921. [Google Scholar] [CrossRef] [Green Version]
  138. Su, N.W.; Wu, S.H.; Chi, C.W.; Liu, C.J.; Tsai, T.H.; Chen, Y.J. Metronomic Cordycepin Therapy Prolongs Survival of Oral Cancer-Bearing Mice and Inhibits Epithelial-Mesenchymal Transition. Molecules 2017, 22, 629. [Google Scholar] [CrossRef] [Green Version]
  139. Yao, W.L.; Ko, B.S.; Liu, T.A.; Liang, S.M.; Liu, C.C.; Lu, Y.J.; Tzean, S.S.; Shen, T.L.; Liou, J.Y. Cordycepin suppresses integrin/FAK signaling and epithelial-mesenchymal transition in hepatocellular carcinoma. Anticancer Agents Med. Chem. 2014, 14, 29–34. [Google Scholar] [CrossRef]
  140. Feng, Z.; Zheng, W.; Li, X.; Lin, J.; Xie, C.; Li, H.; Cheng, L.; Wu, A.; Ni, W. Cryptotanshinone protects against IL-1beta-induced inflammation in human osteoarthritis chondrocytes and ameliorates the progression of osteoarthritis in mice. Int. Immunopharmacol. 2017, 50, 161–167. [Google Scholar] [CrossRef]
  141. Zhang, Y.; Cabarcas, S.M.; Zheng, J.I.; Sun, L.; Mathews, L.A.; Zhang, X.; Lin, H.; Farrar, W.L. Cryptotanshinone targets tumor-initiating cells through down-regulation of stemness genes expression. Oncol. Lett. 2016, 11, 3803–3812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Zhang, L.; Cheng, X.; Gao, Y.; Zhang, C.; Bao, J.; Guan, H.; Yu, H.; Lu, R.; Xu, Q.; Sun, Y. Curcumin inhibits metastasis in human papillary thyroid carcinoma BCPAP cells via down-regulation of the TGF-beta/Smad2/3 signaling pathway. Exp. Cell Res. 2016, 341, 157–165. [Google Scholar] [CrossRef]
  143. Bahrami, A.; Majeed, M.; Sahebkar, A. Curcumin: A potent agent to reverse epithelial-to-mesenchymal transition. Cell Oncol. (Dordr) 2019, 42, 405–421. [Google Scholar] [CrossRef] [PubMed]
  144. Jiang, G.-M.; Xie, W.-Y.; Wang, H.-S.; Du, J.; Wu, B.-P.; Xu, W.; Liu, H.-F.; Xiao, P.; Liu, Z.-G.; Li, H.-Y. Curcumin combined with FAPαc vaccine elicits effective antitumor response by targeting indolamine-2, 3-dioxygenase and inhibiting EMT induced by TNF-α in melanoma. Oncotarget 2015, 6, 25932–25942. [Google Scholar]
  145. Aedo-Aguilera, V.; Carrillo-Beltran, D.; Calaf, G.M.; Munoz, J.P.; Guerrero, N.; Osorio, J.C.; Tapia, J.C.; Leon, O.; Contreras, H.R.; Aguayo, F. Curcumin decreases epithelialmesenchymal transition by a Pirindependent mechanism in cervical cancer cells. Oncol. Rep. 2019. [Google Scholar] [CrossRef]
  146. Huber, M.A.; Beug, H.; Wirth, T. Epithelial-mesenchymal transition: NF-kappaB takes center stage. Cell Cycle 2004, 3, 1477–1480. [Google Scholar] [CrossRef] [Green Version]
  147. Tyagi, N.; Singh, D.K.; Dash, D.; Singh, R. Curcumin Modulates Paraquat-Induced Epithelial to Mesenchymal Transition by Regulating Transforming Growth Factor-beta (TGF-beta) in A549 Cells. Inflammation 2019, 42, 1441–1455. [Google Scholar] [CrossRef]
  148. Yin, J.; Wang, L.; Wang, Y.; Shen, H.; Wang, X.; Wu, L. Curcumin reverses oxaliplatin resistance in human colorectal cancer via regulation of TGF-beta/Smad2/3 signaling pathway. OncoTargets Ther. 2019, 12, 3893–3903. [Google Scholar] [CrossRef] [Green Version]
  149. Lim, W.C.; Kim, H.; Kim, Y.J.; Choi, K.C.; Lee, I.H.; Lee, K.H.; Kim, M.K.; Ko, H. Dioscin suppresses TGF-beta1-induced epithelial-mesenchymal transition and suppresses A549 lung cancer migration and invasion. Bioorg. Med. Chem. Lett. 2017, 27, 3342–3348. [Google Scholar] [CrossRef]
  150. Wu, S.; Xu, H.; Peng, J.; Wang, C.; Jin, Y.; Liu, K.; Sun, H.; Qin, J. Potent anti-inflammatory effect of dioscin mediated by suppression of TNF-α-induced VCAM-1, ICAM-1and EL expression via the NF-κB pathway. Biochimie 2015, 110, 62–72. [Google Scholar] [CrossRef]
  151. Chen, B.; Zhou, S.; Zhan, Y.; Ke, J.; Wang, K.; Liang, Q.; Hou, Y.; Zhu, P.; Ao, W.; Wei, X.; et al. Dioscin Inhibits the Invasion and Migration of Hepatocellular Carcinoma HepG2 Cells by Reversing TGF-beta1-Induced Epithelial-Mesenchymal Transition. Molecules 2019, 24, 2222. [Google Scholar] [CrossRef] [Green Version]
  152. Chang, H.Y.; Kao, M.C.; Way, T.D.; Ho, C.T.; Fu, E. Diosgenin suppresses hepatocyte growth factor (HGF)-induced epithelial-mesenchymal transition by down-regulation of Mdm2 and vimentin. J. Agric. Food Chem. 2011, 59, 5357–5363. [Google Scholar] [CrossRef] [PubMed]
  153. Bhuvanalakshmi, G.; Rangappa, K.S.; Dharmarajan, A.; Sethi, G.; Kumar, A.P.; Warrier, S. Breast Cancer Stem-Like Cells Are Inhibited by Diosgenin, a Steroidal Saponin, by the Attenuation of the Wnt beta-Catenin Signaling via the Wnt Antagonist Secreted Frizzled Related Protein-4. Front. Pharmacol. 2017, 8, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Sogo, T.; Terahara, N.; Hisanaga, A.; Kumamoto, T.; Yamashiro, T.; Wu, S.; Sakao, K.; Hou, D.X. Anti-inflammatory activity and molecular mechanism of delphinidin 3-sambubioside, a Hibiscus anthocyanin. BioFactors 2015, 41, 58–65. [Google Scholar] [CrossRef] [PubMed]
  155. Ouanouki, A.; Lamy, S.; Annabi, B. Anthocyanidins inhibit epithelial–mesenchymal transition through a TGF/Smad2 signaling pathway in glioblastoma cells. Mol. Carcinog. 2017, 56, 1088–1099. [Google Scholar] [CrossRef] [PubMed]
  156. Lim, W.C.; Kim, H.; Ko, H. Delphinidin inhibits epidermal growth factor-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma cells. J. Cell Biochem. 2019, 120, 9887–9899. [Google Scholar] [CrossRef]
  157. Byun, E.B.; Kim, W.S.; Sung, N.Y.; Byun, E.H. Epigallocatechin-3-Gallate Regulates Anti-Inflammatory Action Through 67-kDa Laminin Receptor-Mediated Tollip Signaling Induction in Lipopolysaccharide-Stimulated Human Intestinal Epithelial Cells. Cell Physiol. Biochem. 2018, 46, 2072–2081. [Google Scholar] [CrossRef]
  158. Shi, J.; Liu, F.; Zhang, W.; Liu, X.; Lin, B.; Tang, X. Epigallocatechin-3-gallate inhibits nicotine-induced migration and invasion by the suppression of angiogenesis and epithelial-mesenchymal transition in non-small cell lung cancer cells. Oncol. Rep. 2015, 33, 2972–2980. [Google Scholar] [CrossRef]
  159. Li, T.; Zhao, N.; Lu, J.; Zhu, Q.; Liu, X.; Hao, F.; Jiao, X. Epigallocatechin gallate (EGCG) suppresses epithelial-Mesenchymal transition (EMT) and invasion in anaplastic thyroid carcinoma cells through blocking of TGF-beta1/Smad signaling pathways. Bioengineered 2019, 10, 282–291. [Google Scholar] [CrossRef] [Green Version]
  160. Boldbaatar, A.; Lee, S.; Han, S.; Jeong, A.L.; Ka, H.I.; Buyanravjikh, S.; Lee, J.H.; Lim, J.S.; Lee, M.S.; Yang, Y. Eupatolide inhibits the TGF-beta1-induced migration of breast cancer cells via downregulation of SMAD3 phosphorylation and transcriptional repression of ALK5. Oncol. Lett. 2017, 14, 6031–6039. [Google Scholar] [CrossRef] [Green Version]
  161. Lee, J.; Tae, N.; Lee, J.J.; Kim, T.; Lee, J.H. Eupatolide inhibits lipopolysaccharide-induced COX-2 and iNOS expression in RAW264.7 cells by inducing proteasomal degradation of TRAF6. Eur. J. Pharmacol. 2010, 636, 173–180. [Google Scholar] [CrossRef] [PubMed]
  162. Fan, Y.; Piao, C.H.; Hyeon, E.; Jung, S.Y.; Eom, J.E.; Shin, H.S.; Song, C.H.; Chai, O.H. Gallic acid alleviates nasal inflammation via activation of Th1 and inhibition of Th2 and Th17 in a mouse model of allergic rhinitis. Int. Immunopharmacol. 2019, 70, 512–519. [Google Scholar] [CrossRef] [PubMed]
  163. BenSaad, L.A.; Kim, K.H.; Quah, C.C.; Kim, W.R.; Shahimi, M. Anti-inflammatory potential of ellagic acid, gallic acid and punicalagin A&B isolated from Punica granatum. BMC Complement. Altern. Med. 2017, 17, 47. [Google Scholar] [CrossRef] [Green Version]
  164. Sunil Gowda, S.N.; Rajasowmiya, S.; Vadivel, V.; Banu Devi, S.; Celestin Jerald, A.; Marimuthu, S.; Devipriya, N. Gallic acid-coated sliver nanoparticle alters the expression of radiation-induced epithelial-mesenchymal transition in non-small lung cancer cells. Toxicol. In Vitro 2018, 52, 170–177. [Google Scholar] [CrossRef]
  165. Chang, Y.C.; Chen, P.N.; Chu, S.C.; Lin, C.Y.; Kuo, W.H.; Hsieh, Y.S. Black tea polyphenols reverse epithelial-to-mesenchymal transition and suppress cancer invasion and proteases in human oral cancer cells. J. Agric. Food Chem. 2012, 60, 8395–8403. [Google Scholar] [CrossRef]
  166. Ren, J.; Li, L.; Wang, Y.; Zhai, J.; Chen, G.; Hu, K. Gambogic acid induces heme oxygenase-1 through Nrf2 signaling pathway and inhibits NF-kappaB and MAPK activation to reduce inflammation in LPS-activated RAW264.7 cells. Biomed. Pharmacother. 2019, 109, 555–562. [Google Scholar] [CrossRef]
  167. Zhao, K.; Zhang, S.; Song, X.; Yao, Y.; Zhou, Y.; You, Q.; Guo, Q.; Lu, N. Gambogic acid suppresses cancer invasion and migration by inhibiting TGFbeta1-induced epithelial-to-mesenchymal transition. Oncotarget 2017, 8, 27120–27136. [Google Scholar] [CrossRef]
  168. Yue, Q.; Feng, L.; Cao, B.; Liu, M.; Zhang, D.; Wu, W.; Jiang, B.; Yang, M.; Liu, X.; Guo, D. Proteomic Analysis Revealed the Important Role of Vimentin in Human Cervical Carcinoma HeLa Cells Treated With Gambogic Acid. Mol. Cell. Proteom. 2016, 15, 26–44. [Google Scholar] [CrossRef] [Green Version]
  169. Tharmarajah, L.; Samarakoon, S.R.; Ediriweera, M.K.; Piyathilaka, P.; Tennekoon, K.H.; Senathilake, K.S.; Rajagopalan, U.; Galhena, P.B.; Thabrew, I. In Vitro Anticancer Effect of Gedunin on Human Teratocarcinomal (NTERA-2) Cancer Stem-Like Cells. Biomed. Res. Int. 2017, 2017, 2413197. [Google Scholar] [CrossRef] [Green Version]
  170. Borges, P.V.; Moret, K.H.; Raghavendra, N.M.; Maramaldo Costa, T.E.; Monteiro, A.P.; Carneiro, A.B.; Pacheco, P.; Temerozo, J.R.; Bou-Habib, D.C.; das Gracas Henriques, M.; et al. Protective effect of gedunin on TLR-mediated inflammation by modulation of inflammasome activation and cytokine production: Evidence of a multitarget compound. Pharmacol. Res. 2017, 115, 65–77. [Google Scholar] [CrossRef]
  171. Subramani, R.; Gonzalez, E.; Nandy, S.B.; Arumugam, A.; Camacho, F.; Medel, J.; Alabi, D.; Lakshmanaswamy, R. Gedunin inhibits pancreatic cancer by altering sonic hedgehog signaling pathway. Oncotarget 2017, 8, 10891–10904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Bhattarai, G.; Poudel, S.B.; Kook, S.H.; Lee, J.C. Anti-inflammatory, anti-osteoclastic, and antioxidant activities of genistein protect against alveolar bone loss and periodontal tissue degradation in a mouse model of periodontitis. J. Biomed. Mater. Res. A 2017, 105, 2510–2521. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, A.; Wei, J.; Lu, C.; Chen, H.; Zhong, X.; Lu, Y.; Li, L.; Huang, H.; Dai, Z.; Han, L. Genistein suppresses psoriasis-related inflammation through a STAT3-NF-kappaB-dependent mechanism in keratinocytes. Int. Immunopharmacol. 2019, 69, 270–278. [Google Scholar] [CrossRef] [PubMed]
  174. Spagnuolo, C.; Moccia, S.; Russo, G.L. Anti-inflammatory effects of flavonoids in neurodegenerative disorders. Eur. J. Med. Chem. 2018, 153, 105–115. [Google Scholar] [CrossRef]
  175. Lee, G.A.; Hwang, K.A.; Choi, K.C. Roles of Dietary Phytoestrogens on the Regulation of Epithelial-Mesenchymal Transition in Diverse Cancer Metastasis. Toxins (Basel) 2016, 8, 162. [Google Scholar] [CrossRef] [Green Version]
  176. Zhou, P.; Wang, C.; Hu, Z.; Chen, W.; Qi, W.; Li, A. Genistein induces apoptosis of colon cancer cells by reversal of epithelial-to-mesenchymal via a Notch1/NF-kappaB/slug/E-cadherin pathway. BMC Cancer 2017, 17, 813. [Google Scholar] [CrossRef]
  177. Ma, J.; Zeng, F.; Ma, C.; Pang, H.; Fang, B.; Lian, C.; Yin, B.; Zhang, X.; Wang, Z.; Xia, J. Synergistic reversal effect of epithelial-to-mesenchymal transition by miR-223 inhibitor and genistein in gemcitabine-resistant pancreatic cancer cells. Am. J. Cancer Res. 2016, 6, 1384–1395. [Google Scholar]
  178. Nam, H.H.; Nan, L.; Park, J.C.; Choo, B.K. Geraniin ameliorate experimental acute reflux esophagitis via NF-κB regulated anti-inflammatory activities in rats. App. Biol. Chem. 2019, 62, 13. [Google Scholar] [CrossRef]
  179. Lee, J.C.; Tsai, C.Y.; Kao, J.Y.; Kao, M.C.; Tsai, S.C.; Chang, C.S.; Huang, L.J.; Kuo, S.C.; Lin, J.K.; Way, T.D. Geraniin-mediated apoptosis by cleavage of focal adhesion kinase through up-regulation of Fas ligand expression in human melanoma cells. Mol. Nutr. Food Res. 2008, 52, 655–663. [Google Scholar] [CrossRef]
  180. Ko, H. Geraniin inhibits TGF-beta1-induced epithelial-mesenchymal transition and suppresses A549 lung cancer migration, invasion and anoikis resistance. Bioorg. Med. Chem. Lett. 2015, 25, 3529–3534. [Google Scholar] [CrossRef]
  181. Won, J.H.; Kim, J.Y.; Yun, K.J.; Lee, J.H.; Back, N.I.; Chung, H.G.; Chung, S.A.; Jeong, T.S.; Choi, M.S.; Lee, K.T. Gigantol isolated from the whole plants of Cymbidium goeringii inhibits the LPS-induced iNOS and COX-2 expression via NF-kappaB inactivation in RAW 264.7 macrophages cells. Planta Med. 2006, 72, 1181–1187. [Google Scholar] [CrossRef] [PubMed]
  182. Unahabhokha, T.; Chanvorachote, P.; Pongrakhananon, V. The attenuation of epithelial to mesenchymal transition and induction of anoikis by gigantol in human lung cancer H460 cells. Tumour Biol. 2016, 37, 8633–8641. [Google Scholar] [CrossRef] [PubMed]
  183. Bhummaphan, N.; Chanvorachote, P. Gigantol Suppresses Cancer Stem Cell-Like Phenotypes in Lung Cancer Cells. Evid. Based Complement. Alternat. Med. 2015, 2015, 836564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Kotake, D.; Hirasawa, N. Activation of a retinoic acid receptor pathway by thiazolidinediones induces production of vascular endothelial growth factor/vascular permeability factor in OP9 adipocytes. Eur. J. Pharmacol. 2013, 707, 95–103. [Google Scholar] [CrossRef]
  185. Baek, S.H.; Ko, J.H.; Lee, J.H.; Kim, C.; Lee, H.; Nam, D.; Lee, J.; Lee, S.G.; Yang, W.M.; Um, J.Y.; et al. Ginkgolic Acid Inhibits Invasion and Migration and TGF-beta-Induced EMT of Lung Cancer Cells Through PI3K/Akt/mTOR Inactivation. J. Cell Physiol. 2017, 232, 346–354. [Google Scholar] [CrossRef]
  186. Yi, Y.S. Roles of ginsenosides in inflammasome activation. J. Ginseng Res. 2019, 43, 172–178. [Google Scholar] [CrossRef]
  187. Tian, L.; Shen, D.; Li, X.; Shan, X.; Wang, X.; Yan, Q.; Liu, J. Ginsenoside Rg3 inhibits epithelial-mesenchymal transition (EMT) and invasion of lung cancer by down-regulating FUT4. Oncotarget 2016, 7, 1619–1632. [Google Scholar] [CrossRef] [Green Version]
  188. Wang, J.; Tian, L.; Khan, M.N.; Zhang, L.; Chen, Q.; Zhao, Y.; Yan, Q.; Fu, L.; Liu, J. Ginsenoside Rg3 sensitizes hypoxic lung cancer cells to cisplatin via blocking of NF-kappaB mediated epithelial-mesenchymal transition and stemness. Cancer Lett. 2018, 415, 73–85. [Google Scholar] [CrossRef]
  189. Liu, D.; Liu, T.; Teng, Y.; Chen, W.; Zhao, L.; Li, X. Ginsenoside Rb1 inhibits hypoxia-induced epithelial-mesenchymal transition in ovarian cancer cells by regulating microRNA-25. Exp. Ther. Med. 2017, 14, 2895–2902. [Google Scholar] [CrossRef] [Green Version]
  190. Zhang, L.; Shan, X.; Chen, Q.; Xu, D.; Fan, X.; Yu, M.; Yan, Q.; Liu, J. Downregulation of HDAC3 by ginsenoside Rg3 inhibits epithelial-mesenchymal transition of cutaneous squamous cell carcinoma through c-Jun acetylation. J. Cell. Physiol. 2019, 234, 22207–22219. [Google Scholar] [CrossRef]
  191. Cheng, Z.; Xing, D. Ginsenoside Rg3 inhibits growth and epithelial-mesenchymal transition of human oral squamous carcinoma cells by down-regulating miR-221. Eur. J. Pharmacol. 2019, 853, 353–363. [Google Scholar] [CrossRef] [PubMed]
  192. Chang, H.Y.; Chen, S.Y.; Wu, C.H.; Lu, C.C.; Yen, G.C. Glycyrrhizin Attenuates the Process of Epithelial-to-Mesenchymal Transition by Modulating HMGB1 Initiated Novel Signaling Pathway in Prostate Cancer Cells. J. Agric. Food Chem. 2019, 67, 3323–3332. [Google Scholar] [CrossRef] [PubMed]
  193. Woodbury, A.; Yu, S.P.; Wei, L.; Garcia, P. Neuro-modulating effects of honokiol: A review. Front. Neurol. 2013, 4, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Lee, J.; Jung, E.; Park, J.; Jung, K.; Lee, S.; Hong, S.; Park, J.; Park, E.; Kim, J.; Park, S.; et al. Anti-inflammatory effects of magnolol and honokiol are mediated through inhibition of the downstream pathway of MEKK-1 in NF-kappaB activation signaling. Planta Med. 2005, 71, 338–343. [Google Scholar] [CrossRef]
  195. Lv, X.Q.; Qiao, X.R.; Su, L.; Chen, S.Z. Honokiol inhibits EMT-mediated motility and migration of human non-small cell lung cancer cells in vitro by targeting c-FLIP. Acta Pharmacol. Sin. 2016, 37, 1574–1586. [Google Scholar] [CrossRef]
  196. Avtanski, D.B.; Nagalingam, A.; Bonner, M.Y.; Arbiser, J.L.; Saxena, N.K.; Sharma, D. Honokiol inhibits epithelial-mesenchymal transition in breast cancer cells by targeting signal transducer and activator of transcription 3/Zeb1/E-cadherin axis. Mol. Oncol. 2014, 8, 565–580. [Google Scholar] [CrossRef]
  197. Li, W.; Wang, Q.; Su, Q.; Ma, D.; An, C.; Ma, L.; Liang, H. Honokiol suppresses renal cancer cells’ metastasis via dual-blocking epithelial-mesenchymal transition and cancer stem cell properties through modulating miR-141/ZEB2 signaling. Mol. Cells 2014, 37, 383–388. [Google Scholar] [CrossRef] [Green Version]
  198. Joo, Y.N.; Eun, S.Y.; Park, S.W.; Lee, J.H.; Chang, K.C.; Kim, H.J. Honokiol inhibits U87MG human glioblastoma cell invasion through endothelial cells by regulating membrane permeability and the epithelial-mesenchymal transition. Int. J. Oncol. 2014, 44, 187–194. [Google Scholar] [CrossRef]
  199. Nesterova, Y.V.; Povetieva, T.N.; Suslov, N.I.; Zyuz’kov, G.N.; Aksinenko, S.G.; Pushkarskii, S.V.; Krapivin, A.V. Anti-inflammatory activity of diterpene alkaloids from Aconitum baikalense. Bull. Exp. Biol. Med. 2014, 156, 665–668. [Google Scholar] [CrossRef]
  200. Feng, H.T.; Zhao, W.W.; Lu, J.J.; Wang, Y.T.; Chen, X.P. Hypaconitine inhibits TGF-beta1-induced epithelial-mesenchymal transition and suppresses adhesion, migration, and invasion of lung cancer A549 cells. Chin. J. Nat. Med. 2017, 15, 427–435. [Google Scholar] [CrossRef]
  201. Frohlich, J.K.; Stein, T.; da Silva, L.A.; Biavatti, M.W.; Tonussi, C.R.; Lemos-Senna, E. Antinociceptive and anti-inflammatory activities of the Jatropha isabellei dichloromethane fraction and isolation and quantitative determination of jatrophone by UFLC-DAD. Pharm. Biol. 2017, 55, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
  202. Fatima, I.; El-Ayachi, I.; Taotao, L.; Lillo, M.A.; Krutilina, R.I.; Seagroves, T.N.; Radaszkiewicz, T.W.; Hutnan, M.; Bryja, V.; Krum, S.A.; et al. The natural compound Jatrophone interferes with Wnt/beta-catenin signaling and inhibits proliferation and EMT in human triple-negative breast cancer. PLoS ONE 2017, 12, e0189864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Wei, Y.; Liu, J.; Zhang, H.; Du, X.; Luo, Q.; Sun, J.; Liu, F.; Li, M.; Xu, F.; Wei, K.; et al. Ligustrazine attenuates inflammation and the associated chemokines and receptors in ovalbumine-induced mouse asthma model. Environ. Toxicol. Pharmacol. 2016, 46, 55–61. [Google Scholar] [CrossRef] [PubMed]
  204. Hashim, P.; Selamat, J.; Syed Muhammad, S.K.; Ali, A. Effect of mass and turning time on free amino acid, peptide-N, sugar and pyrazine concentration during cocoa fermentation. J. Sci. Food Agric. 1998, 78, 543–550. [Google Scholar] [CrossRef]
  205. Donkor, P.O.; Chen, Y.; Ding, L.; Qiu, F. Locally and traditionally used Ligusticum species—A review of their phytochemistry, pharmacology and pharmacokinetics. J. Ethnopharmacol. 2016, 194, 530–548. [Google Scholar] [CrossRef]
  206. Luan, Y.; Liu, J.; Liu, X.; Xue, X.; Kong, F.; Sun, C.; Wang, J.; Liu, L.; Jia, H. Tetramethypyrazine inhibits renal cell carcinoma cells through inhibition of NKG2D signaling pathways. Int. J. Oncol. 2016, 49, 1704–1712. [Google Scholar] [CrossRef]
  207. Chen, K.C.; Chen, C.Y.; Lin, C.R.; Yang, T.Y.; Chen, T.H.; Wu, L.C.; Wu, C.C. Luteolin attenuates TGF-beta1-induced epithelial-mesenchymal transition of lung cancer cells by interfering in the PI3K/Akt-NF-kappaB-Snail pathway. Life Sci. 2013, 93, 924–933. [Google Scholar] [CrossRef]
  208. Gao, G.; Ge, R.; Li, Y.; Liu, S. Luteolin exhibits anti-breast cancer property through up-regulating miR-203. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3265–3271. [Google Scholar] [CrossRef] [Green Version]
  209. Lin, D.; Kuang, G.; Wan, J.; Zhang, X.; Li, H.; Gong, X.; Li, H. Luteolin suppresses the metastasis of triple-negative breast cancer by reversing epithelial-to-mesenchymal transition via downregulation of beta-catenin expression. Oncol. Rep. 2017, 37, 895–902. [Google Scholar] [CrossRef]
  210. Li, C.; Wang, Q.; Shen, S.; Wei, X.; Li, G. HIF-1alpha/VEGF signaling-mediated epithelial-mesenchymal transition and angiogenesis is critically involved in anti-metastasis effect of luteolin in melanoma cells. Phytother. Res. 2019, 33, 798–807. [Google Scholar] [CrossRef]
  211. Liu, Y.; Lang, T.; Jin, B.; Chen, F.; Zhang, Y.; Beuerman, R.W.; Zhou, L.; Zhang, Z. Luteolin inhibits colorectal cancer cell epithelial-to-mesenchymal transition by suppressing CREB1 expression revealed by comparative proteomics study. J. Proteom. 2017, 161, 1–10. [Google Scholar] [CrossRef] [PubMed]
  212. Lin, T.H.; Hsu, W.H.; Tsai, P.H.; Huang, Y.T.; Lin, C.W.; Chen, K.C.; Tsai, I.H.; Kandaswami, C.C.; Huang, C.J.; Chang, G.D.; et al. Dietary flavonoids, luteolin and quercetin, inhibit invasion of cervical cancer by reduction of UBE2S through epithelial-mesenchymal transition signaling. Food Funct. 2017, 8, 1558–1568. [Google Scholar] [CrossRef] [PubMed]
  213. Zang, M.D.; Hu, L.; Fan, Z.Y.; Wang, H.X.; Zhu, Z.L.; Cao, S.; Wu, X.Y.; Li, J.F.; Su, L.P.; Li, C.; et al. Luteolin suppresses gastric cancer progression by reversing epithelial-mesenchymal transition via suppression of the Notch signaling pathway. J. Transl. Med. 2017, 15, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Dia, V.P.; Pangloli, P. Epithelial-to-Mesenchymal Transition in Paclitaxel-Resistant Ovarian Cancer Cells Is Downregulated by Luteolin. J. Cell. Physiol. 2017, 232, 391–401. [Google Scholar] [CrossRef]
  215. Huang, X.; Dai, S.; Dai, J.; Xiao, Y.; Bai, Y.; Chen, B.; Zhou, M. Luteolin decreases invasiveness, deactivates STAT3 signaling, and reverses interleukin-6 induced epithelial-mesenchymal transition and matrix metalloproteinase secretion of pancreatic cancer cells. OncoTargets Ther. 2015, 8, 2989–3001. [Google Scholar] [CrossRef] [Green Version]
  216. Seo, J.Y.; Lee, C.; Hwang, S.W.; Chun, J.; Im, J.P.; Kim, J.S. Nimbolide Inhibits Nuclear Factor-capital KA, CyrillicB Pathway in Intestinal Epithelial Cells and Macrophages and Alleviates Experimental Colitis in Mice. Phytother. Res. 2016, 30, 1605–1614. [Google Scholar] [CrossRef] [Green Version]
  217. Subramani, R.; Gonzalez, E.; Arumugam, A.; Nandy, S.; Gonzalez, V.; Medel, J.; Camacho, F.; Ortega, A.; Bonkoungou, S.; Narayan, M.; et al. Nimbolide inhibits pancreatic cancer growth and metastasis through ROS-mediated apoptosis and inhibition of epithelial-to-mesenchymal transition. Sci. Rep. 2016, 6, 19819. [Google Scholar] [CrossRef]
  218. Lin, H.; Qiu, S.; Xie, L.; Liu, C.; Sun, S. Nimbolide suppresses non-small cell lung cancer cell invasion and migration via manipulation of DUSP4 expression and ERK1/2 signaling. Biomed. Pharmacother. 2017, 92, 340–346. [Google Scholar] [CrossRef]
  219. Wang, Z.; Jiang, W.; Zhang, Z.; Qian, M.; Du, B. Nitidine chloride inhibits LPS-induced inflammatory cytokines production via MAPK and NF-kappaB pathway in RAW 264.7 cells. J. Ethnopharmacol. 2012, 144, 145–150. [Google Scholar] [CrossRef]
  220. Sun, M.; Zhang, N.; Wang, X.; Li, Y.; Qi, W.; Zhang, H.; Li, Z.; Yang, Q. Hedgehog pathway is involved in nitidine chloride induced inhibition of epithelial-mesenchymal transition and cancer stem cells-like properties in breast cancer cells. Cell Biosci. 2016, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  221. Cheng, Z.; Guo, Y.; Yang, Y.; Kan, J.; Dai, S.; Helian, M.; Li, B.; Xu, J.; Liu, C. Nitidine chloride suppresses epithelial-to-mesenchymal transition in osteosarcoma cell migration and invasion through Akt/GSK-3beta/Snail signaling pathway. Oncol. Rep. 2016, 36, 1023–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Fan, H.; Gao, Z.; Ji, K.; Li, X.; Wu, J.; Liu, Y.; Wang, X.; Liang, H.; Liu, Y.; Li, X.; et al. The in vitro and in vivo anti-inflammatory effect of osthole, the major natural coumarin from Cnidium monnieri (L.) Cuss, via the blocking of the activation of the NF-kappaB and MAPK/p38 pathways. Phytomedicine 2019, 58, 152864. [Google Scholar] [CrossRef] [PubMed]
  223. Hung, C.M.; Kuo, D.H.; Chou, C.H.; Su, Y.C.; Ho, C.T.; Way, T.D. Osthole suppresses hepatocyte growth factor (HGF)-induced epithelial-mesenchymal transition via repression of the c-Met/Akt/mTOR pathway in human breast cancer cells. J. Agric. Food Chem. 2011, 59, 9683–9690. [Google Scholar] [CrossRef] [PubMed]
  224. Lin, Y.C.; Lin, J.C.; Hung, C.M.; Chen, Y.; Liu, L.C.; Chang, T.C.; Kao, J.Y.; Ho, C.T.; Way, T.D. Osthole inhibits insulin-like growth factor-1-induced epithelial to mesenchymal transition via the inhibition of PI3K/Akt signaling pathway in human brain cancer cells. J. Agric. Food Chem. 2014, 62, 5061–5071. [Google Scholar] [CrossRef]
  225. Wen, Y.C.; Lee, W.J.; Tan, P.; Yang, S.F.; Hsiao, M.; Lee, L.M.; Chien, M.H. By inhibiting snail signaling and miR-23a-3p, osthole suppresses the EMT-mediated metastatic ability in prostate cancer. Oncotarget 2015, 6, 21120–21136. [Google Scholar] [CrossRef]
  226. Feng, H.; Lu, J.J.; Wang, Y.; Pei, L.; Chen, X. Osthole inhibited TGF beta-induced epithelial-mesenchymal transition (EMT) by suppressing NF-kappaB mediated Snail activation in lung cancer A549 cells. Cell Adh. Migr. 2017, 11, 464–475. [Google Scholar] [CrossRef] [Green Version]
  227. Chen, Y.; Qi, Z.; Qiao, B.; Lv, Z.; Hao, Y.; Li, H. Oxymatrine can attenuate pathological deficits of Alzheimer’s disease mice through regulation of neuroinflammation. J. Neuroimmunol. 2019, 334, 576978. [Google Scholar] [CrossRef]
  228. Chen, Y.; Chen, L.; Zhang, J.Y.; Chen, Z.Y.; Liu, T.T.; Zhang, Y.Y.; Fu, L.Y.; Fan, S.Q.; Zhang, M.Q.; Gan, S.Q.; et al. Oxymatrine reverses epithelial-mesenchymal transition in breast cancer cells by depressing alphabeta3 integrin/FAK/PI3K/Akt signaling activation. OncoTargets Ther. 2019, 12, 6253–6265. [Google Scholar] [CrossRef] [Green Version]
  229. Liang, L.; Huang, J. Oxymatrine inhibits epithelial-mesenchymal transition through regulation of NF-kappaB signaling in colorectal cancer cells. Oncol. Rep. 2016, 36, 1333–1338. [Google Scholar] [CrossRef]
  230. Xiong, Y.; Wang, J.; Zhu, H.; Liu, L.; Jiang, Y. Chronic oxymatrine treatment induces resistance and epithelialmesenchymal transition through targeting the long non-coding RNA MALAT1 in colorectal cancer cells. Oncol. Rep. 2018, 39, 967–976. [Google Scholar] [CrossRef]
  231. Yan, D.; Saito, K.; Ohmi, Y.; Fujie, N.; Ohtsuka, K. Paeoniflorin, a novel heat shock protein-inducing compound. Cell Stress Chaperones 2004, 9, 378–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Tu, J.; Guo, Y.; Hong, W.; Fang, Y.; Han, D.; Zhang, P.; Wang, X.; Korner, H.; Wei, W. The Regulatory Effects of Paeoniflorin and Its Derivative Paeoniflorin-6′-O-Benzene Sulfonate CP-25 on Inflammation and Immune Diseases. Front. Pharmacol. 2019, 10, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Zhang, J.W.; Li, L.X.; Wu, W.Z.; Pan, T.J.; Yang, Z.S.; Yang, Y.K. Anti-Tumor Effects of Paeoniflorin on Epithelial-To-Mesenchymal Transition in Human Colorectal Cancer Cells. Med. Sci. Monit. 2018, 24, 6405–6413. [Google Scholar] [CrossRef] [PubMed]
  234. Wang, Z.; Liu, Z.; Yu, G.; Nie, X.; Jia, W.; Liu, R.E.; Xu, R. Paeoniflorin Inhibits Migration and Invasion of Human Glioblastoma Cells via Suppression Transforming Growth Factor beta-Induced Epithelial-Mesenchymal Transition. Neurochem. Res. 2018, 43, 760–774. [Google Scholar] [CrossRef] [Green Version]
  235. Fukuhara, Y.; Yoshida, D. Paeonol: A bio-antimutagen isolated from a crude drug, Moutan cortex. Agric. Biol. Chem. 1987, 51, 1441–1442. [Google Scholar]
  236. Lei, H.; Wen, Q.; Li, H.; Du, S.; Wu, J.J.; Chen, J.; Huang, H.; Chen, D.; Li, Y.; Zhang, S.; et al. Paeonol Inhibits Lipopolysaccharide-Induced HMGB1 Translocation from the Nucleus to the Cytoplasm in RAW264.7 Cells. Inflammation 2016, 39, 1177–1187. [Google Scholar] [CrossRef]
  237. Yang, L.; Xing, S.; Wang, K.; Yi, H.; Du, B. Paeonol attenuates aging MRC-5 cells and inhibits epithelial-mesenchymal transition of premalignant HaCaT cells induced by aging MRC-5 cell-conditioned medium. Mol. Cell Biochem. 2018, 439, 117–129. [Google Scholar] [CrossRef]
  238. Wang, M.; Li, Q. Parthenolide could become a promising and stable drug with anti-inflammatory effects. Nat. Prod. Res. 2015, 29, 1092–1101. [Google Scholar] [CrossRef]
  239. Zhu, S.M.; Park, Y.R.; Seo, S.Y.; Kim, I.H.; Lee, S.T.; Kim, S.W. Parthenolide inhibits transforming growth factor beta1-induced epithelial-mesenchymal transition in colorectal cancer cells. Intest. Res. 2019. [Google Scholar] [CrossRef]
  240. Kim, S.L.; Park, Y.R.; Lee, S.T.; Kim, S.W. Parthenolide suppresses hypoxia-inducible factor-1alpha signaling and hypoxia induced epithelial-mesenchymal transition in colorectal cancer. Int. J. Oncol. 2017, 51, 1809–1820. [Google Scholar] [CrossRef] [Green Version]
  241. Ai, X.Y.; Zhang, H.; Gao, S.Y.; Qin, Y.; Zhong, W.L.; Gu, J.; Li, M.; Qiao, K.L.; Tian, Q.; Cui, Z.H.; et al. Sesquiterpene binding Gly-Leu-Ser/Lys-”co-adaptation pocket” to inhibit lung cancer cell epithelial-mesenchymal transition. Oncotarget 2017, 8, 70192–70203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Akhtar, N.; Jafri, L.; Green, B.D.; Kalsoom, S.; Mirza, B. A Multi-Mode Bioactive Agent Isolated From Ficus microcarpa L. Fill. With Therapeutic Potential for Type 2 Diabetes Mellitus. Front. Pharmacol. 2018, 9, 1376. [Google Scholar] [CrossRef] [PubMed]
  243. Akhtar, N.; Syed, D.N.; Khan, M.I.; Adhami, V.M.; Mirza, B.; Mukhtar, H. The pentacyclic triterpenoid, plectranthoic acid, a novel activator of AMPK induces apoptotic death in prostate cancer cells. Oncotarget 2016, 7, 3819–3831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Akhtar, N.; Syed, D.N.; Lall, R.K.; Mirza, B.; Mukhtar, H. Targeting epithelial to mesenchymal transition in prostate cancer by a novel compound, plectranthoic acid, isolated from Ficus microcarpa. Mol. Carcinog. 2018, 57, 653–663. [Google Scholar] [CrossRef]
  245. Chatterjee, A.; Dutta, C. Alkaloids of Piper longum Linn—I: Structure and synthesis of piperlongumine and piperlonguminine. Tetrahedron 1967, 23, 1769–1781. [Google Scholar] [CrossRef]
  246. Kim, N.; Do, J.; Bae, J.S.; Jin, H.K.; Kim, J.H.; Inn, K.S.; Oh, M.S.; Lee, J.K. Piperlongumine inhibits neuroinflammation via regulating NF-kappaB signaling pathways in lipopolysaccharide-stimulated BV2 microglia cells. J. Pharmacol. Sci. 2018, 137, 195–201. [Google Scholar] [CrossRef]
  247. Park, M.J.; Lee, D.E.; Shim, M.K.; Jang, E.H.; Lee, J.K.; Jeong, S.Y.; Kim, J.H. Piperlongumine inhibits TGF-beta-induced epithelial-to-mesenchymal transition by modulating the expression of E-cadherin, Snail1, and Twist1. Eur. J. Pharmacol. 2017, 812, 243–249. [Google Scholar] [CrossRef]
  248. Messeha, S.S.; Zarmouh, N.O.; Mendonca, P.; Kolta, M.G.; Soliman, K.F.A. The attenuating effects of plumbagin on pro-inflammatory cytokine expression in LPS-activated BV-2 microglial cells. J. Neuroimmunol. 2017, 313, 129–137. [Google Scholar] [CrossRef] [Green Version]
  249. Sakunrangsit, N.; Kalpongnukul, N.; Pisitkun, T.; Ketchart, W. Plumbagin Enhances Tamoxifen Sensitivity and Inhibits Tumor Invasion in Endocrine Resistant Breast Cancer through EMT Regulation. Phytother. Res. 2016, 30, 1968–1977. [Google Scholar] [CrossRef]
  250. Pan, S.T.; Qin, Y.; Zhou, Z.W.; He, Z.X.; Zhang, X.; Yang, T.; Yang, Y.X.; Wang, D.; Zhou, S.F.; Qiu, J.X. Plumbagin suppresses epithelial to mesenchymal transition and stemness via inhibiting Nrf2-mediated signaling pathway in human tongue squamous cell carcinoma cells. Drug Des. Devel. Ther. 2015, 9, 5511–5551. [Google Scholar] [CrossRef] [Green Version]
  251. Wang, F.; Wang, Q.; Zhou, Z.W.; Yu, S.N.; Pan, S.T.; He, Z.X.; Zhang, X.; Wang, D.; Yang, Y.X.; Yang, T.; et al. Plumbagin induces cell cycle arrest and autophagy and suppresses epithelial to mesenchymal transition involving PI3K/Akt/mTOR-mediated pathway in human pancreatic cancer cells. Drug Des. Devel. Ther. 2015, 9, 537–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Qiu, J.X.; Zhou, Z.W.; He, Z.X.; Zhao, R.J.; Zhang, X.; Yang, L.; Zhou, S.F.; Mao, Z.F. Plumbagin elicits differential proteomic responses mainly involving cell cycle, apoptosis, autophagy, and epithelial-to-mesenchymal transition pathways in human prostate cancer PC-3 and DU145 cells. Drug Des. Devel. Ther. 2015, 9, 349–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Wang, Q.; Zhou, X.; Zhao, Y.; Xiao, J.; Lu, Y.; Shi, Q.; Wang, Y.; Wang, H.; Liang, Q. Polyphyllin I Ameliorates Collagen-Induced Arthritis by Suppressing the Inflammation Response in Macrophages Through the NF-kappaB Pathway. Front. Immunol. 2018, 9, 2091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Lou, W.; Chen, Y.; Zhu, K.Y.; Deng, H.; Wu, T.; Wang, J. Polyphyllin I Overcomes EMT-Associated Resistance to Erlotinib in Lung Cancer Cells via IL-6/STAT3 Pathway Inhibition. Biol. Pharm. Bull. 2017, 40, 1306–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Mak, K.K.; Wu, A.T.; Lee, W.H.; Chang, T.C.; Chiou, J.F.; Wang, L.S.; Wu, C.H.; Huang, C.Y.F.; Shieh, Y.S.; Chao, T.Y. Pterostilbene, a bioactive component of blueberries, suppresses the generation of breast cancer stem cells within tumor microenvironment and metastasis via modulating NF-κB/microRNA 448 circuit. Mol. Nutr. Food Res. 2013, 57, 1123–1134. [Google Scholar] [CrossRef] [PubMed]
  256. Su, C.-M.; Lee, W.-H.; Wu, A.T.; Lin, Y.-K.; Wang, L.-S.; Wu, C.-H.; Yeh, C.-T. Pterostilbene inhibits triple-negative breast cancer metastasis via inducing microRNA-205 expression and negatively modulates epithelial-to-mesenchymal transition. J. Nutr. Biochem. 2015, 26, 675–685. [Google Scholar] [CrossRef]
  257. Huang, Y.; Du, J.; Mi, Y.; Li, T.; Gong, Y.; Ouyang, H.; Hou, Y. Long Non-coding RNAs Contribute to the Inhibition of Proliferation and EMT by Pterostilbene in Human Breast Cancer. Front. Oncol. 2018, 8, 629. [Google Scholar] [CrossRef]
  258. Inoue, H.; Nakata, R. Resveratrol Targets in Inflammation. Endocr. Metab. Immune Disord. Drug Targets 2015, 15, 186–195. [Google Scholar] [CrossRef]
  259. Ji, Q.; Liu, X.; Han, Z.; Zhou, L.; Sui, H.; Yan, L.; Jiang, H.; Ren, J.; Cai, J.; Li, Q. Resveratrol suppresses epithelial-to-mesenchymal transition in colorectal cancer through TGF-1/Smads signaling pathway mediated Snail/E-cadherin expression. BMC Cancer 2015, 15, 97. [Google Scholar] [CrossRef] [Green Version]
  260. Wang, H.; Zhang, H.; Tang, L.; Chen, H.; Wu, C.; Zhao, M.; Yang, Y.; Chen, X.; Liu, G. Resveratrol inhibits TGF-1-induced epithelial-to-mesenchymal transition and suppresses lung cancer invasion and metastasis. Toxicology 2013, 303, 139–146. [Google Scholar] [CrossRef]
  261. Li, W.; Ma, J.; Ma, Q.; Li, B.; Han, L.; Liu, J.; Xu, Q.; Duan, W.; Yu, S.; Wang, F.; et al. Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-kappaB pathway. Curr. Med. Chem. 2013, 20, 4185–4194. [Google Scholar] [CrossRef] [PubMed]
  262. Simic, P.; Williams, E.O.; Bell, E.L.; Gong, J.J.; Bonkowski, M.; Guarente, L. SIRT1 suppresses the epithelial-to-mesenchymal transition in cancer metastasis and organ fibrosis. Cell Rep. 2013, 3, 1175–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Byles, V.; Zhu, L.; Lovaas, J.D.; Chmilewski, L.K.; Wang, J.; Faller, D.V.; Dai, Y. SIRT1 induces EMT by cooperating with EMT transcription factors and enhances prostate cancer cell migration and metastasis. Oncogene 2012, 31, 4619–4629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Zhang, X.C.; Chen, J.Q.; Li, B. Salvianolic acid A suppresses CCL-20 expression in TNF-alpha-treated macrophages and ApoE-deficient mice. J. Cardiovasc. Pharmacol. 2014, 64, 318–325. [Google Scholar] [CrossRef] [PubMed]
  265. Tang, Q.; Zhong, H.; Xie, F.; Xie, J.; Chen, H.; Yao, G. Expression of miR-106b-25 induced by salvianolic acid B inhibits epithelial-to-mesenchymal transition in HK-2 cells. Eur. J. Pharmacol. 2014, 741, 97–103. [Google Scholar] [CrossRef]
  266. Wang, Q.L.; Tao, Y.Y.; Yuan, J.L.; Shen, L.; Liu, C.H. Salvianolic acid B prevents epithelial-to-mesenchymal transition through the TGF-beta1 signal transduction pathway in vivo and in vitro. BMC Cell Biol. 2010, 11, 31. [Google Scholar] [CrossRef] [Green Version]
  267. Drobnik, J.; Drobnik, E. Timeline and bibliography of early isolations of plant metabolites (1770–1820) and their impact to pharmacy: A critical study. Fitoterapia 2016, 115, 155–164. [Google Scholar] [CrossRef]
  268. Kang, H.; Jeong, H.D.; Choi, H.Y. The chloroform fraction of Solanum nigrum suppresses nitric oxide and tumor necrosis factor-alpha in LPS-stimulated mouse peritoneal macrophages through inhibition of p38, JNK and ERK1/2. Am. J. Chin. Med. 2011, 39, 1261–1273. [Google Scholar] [CrossRef]
  269. Shen, K.H.; Liao, A.C.; Hung, J.H.; Lee, W.J.; Hu, K.C.; Lin, P.T.; Liao, R.F.; Chen, P.S. alpha-Solanine inhibits invasion of human prostate cancer cell by suppressing epithelial-mesenchymal transition and MMPs expression. Molecules 2014, 19, 11896–11914. [Google Scholar] [CrossRef]
  270. Li, Y.; Liu, Y.; Yan, X.; Liu, Q.; Zhao, Y.H.; Wang, D.W. Pharmacological Effects and Mechanisms of Chinese Medicines Modulating NLRP3 Inflammasomes in Ischemic Cardio/Cerebral Vascular Disease. Am. J. Chin. Med. 2018, 46, 1727–1741. [Google Scholar] [CrossRef]
  271. Wang, D.X.; Zou, Y.J.; Zhuang, X.B.; Chen, S.X.; Lin, Y.; Li, W.L.; Lin, J.J.; Lin, Z.Q. Sulforaphane suppresses EMT and metastasis in human lung cancer through miR-616-5p-mediated GSK3beta/beta-catenin signaling pathways. Acta Pharmacol. Sin. 2017, 38, 241–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. Wu, J.; Han, J.; Hou, B.; Deng, C.; Wu, H.; Shen, L. Sulforaphane inhibits TGF-beta-induced epithelial-mesenchymal transition of hepatocellular carcinoma cells via the reactive oxygen species-dependent pathway. Oncol. Rep. 2016, 35, 2977–2983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Shan, Y.; Zhang, L.; Bao, Y.; Li, B.; He, C.; Gao, M.; Feng, X.; Xu, W.; Zhang, X.; Wang, S. Epithelial-mesenchymal transition, a novel target of sulforaphane via COX-2/MMP2, 9/Snail, ZEB1 and miR-200c/ZEB1 pathways in human bladder cancer cells. J. Nutr. Biochem. 2013, 24, 1062–1069. [Google Scholar] [CrossRef] [PubMed]
  274. Song, D.; Zhao, J.; Deng, W.; Liao, Y.; Hong, X.; Hou, J. Tannic acid inhibits NLRP3 inflammasome-mediated IL-1beta production via blocking NF-kappaB signaling in macrophages. Biochem. Biophys. Res. Commun. 2018, 503, 3078–3085. [Google Scholar] [CrossRef] [PubMed]
  275. Pattarayan, D.; Sivanantham, A.; Krishnaswami, V.; Loganathan, L.; Palanichamy, R.; Natesan, S.; Muthusamy, K.; Rajasekaran, S. Tannic acid attenuates TGF-beta1-induced epithelial-to-mesenchymal transition by effectively intervening TGF-beta signaling in lung epithelial cells. J. Cell. Physiol. 2018, 233, 2513–2525. [Google Scholar] [CrossRef] [PubMed]
  276. Bale, S.; Pulivendala, G.; Godugu, C. Withaferin A attenuates bleomycin-induced scleroderma by targeting FoxO3a and NF-kappabeta signaling: Connecting fibrosis and inflammation. Biofactors 2018, 44, 507–517. [Google Scholar] [CrossRef]
  277. Kyakulaga, A.H.; Aqil, F.; Munagala, R.; Gupta, R.C. Withaferin A inhibits Epithelial to Mesenchymal Transition in Non-Small Cell Lung Cancer Cells. Sci. Rep. 2018, 8, 15737. [Google Scholar] [CrossRef]
  278. Lee, J.; Hahm, E.R.; Marcus, A.I.; Singh, S.V. Withaferin A inhibits experimental epithelial-mesenchymal transition in MCF-10A cells and suppresses vimentin protein level in vivo in breast tumors. Mol. Carcinog. 2015, 54, 417–429. [Google Scholar] [CrossRef] [Green Version]
  279. Yang, Z.; Garcia, A.; Xu, S.; Powell, D.R.; Vertino, P.M.; Singh, S.; Marcus, A.I. Withania somnifera root extract inhibits mammary cancer metastasis and epithelial to mesenchymal transition. PLoS ONE 2013, 8, e75069. [Google Scholar] [CrossRef] [Green Version]
  280. Zhang, Q.; Zhu, B.; Li, Y. Resolution of Cancer-Promoting Inflammation: A New Approach for Anticancer Therapy. Front. Immunol. 2017, 8, 71. [Google Scholar] [CrossRef] [Green Version]
  281. Pirault, J.; Back, M. Lipoxin and Resolvin Receptors Transducing the Resolution of Inflammation in Cardiovascular Disease. Front. Pharmacol. 2018, 9, 1273. [Google Scholar] [CrossRef] [PubMed]
  282. Ohira, T.; Arita, M.; Omori, K.; Recchiuti, A.; Van Dyke, T.E.; Serhan, C.N. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J. Biol. Chem. 2010, 285, 3451–3461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Arita, M.; Bianchini, F.; Aliberti, J.; Sher, A.; Chiang, N.; Hong, S.; Yang, R.; Petasis, N.A.; Serhan, C.N. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J. Exp. Med. 2005, 201, 713–722. [Google Scholar] [CrossRef] [PubMed]
  284. Arita, M.; Ohira, T.; Sun, Y.P.; Elangovan, S.; Chiang, N.; Serhan, C.N. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. J. Immunol. 2007, 178, 3912–3917. [Google Scholar] [CrossRef] [Green Version]
  285. Wang, X.; Guo, J.; Wu, Q.; Niu, C.; Cheng, G.; Liu, D.; Liu, Z.; Zhao, Z.; Xiao, J. Chemerin/chemR23 association with endothelial-mesenchymal transition in diabetic nephropathy. Int. J. Clin. Exp. Pathol. 2017, 10, 7408–7416. [Google Scholar]
  286. Hayhoe, R.P.; Kamal, A.M.; Solito, E.; Flower, R.J.; Cooper, D.; Perretti, M. Annexin 1 and its bioactive peptide inhibit neutrophil-endothelium interactions under flow: Indication of distinct receptor involvement. Blood 2006, 107, 2123–2130. [Google Scholar] [CrossRef] [Green Version]
  287. Fiore, S.; Maddox, J.F.; Perez, H.D.; Serhan, C.N. Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J. Exp. Med. 1994, 180, 253–260. [Google Scholar] [CrossRef] [Green Version]
  288. Krishnamoorthy, S.; Recchiuti, A.; Chiang, N.; Fredman, G.; Serhan, C.N. Resolvin D1 receptor stereoselectivity and regulation of inflammation and proresolving microRNAs. Am. J. Pathol. 2012, 180, 2018–2027. [Google Scholar] [CrossRef] [Green Version]
  289. Arnardottir, H.H.; Dalli, J.; Norling, L.V.; Colas, R.A.; Perretti, M.; Serhan, C.N. Resolvin D3 Is Dysregulated in Arthritis and Reduces Arthritic Inflammation. J. Immunol. 2016, 197, 2362–2368. [Google Scholar] [CrossRef] [Green Version]
  290. Zugazagoitia, J.; Guedes, C.; Ponce, S.; Ferrer, I.; Molina-Pinelo, S.; Paz-Ares, L. Current challenges in cancer treatment. Clin. Ther. 2016, 38, 1551–1566. [Google Scholar] [CrossRef] [Green Version]
  291. Lee, H.J.; Park, M.K.; Lee, E.J.; Lee, C.H. Resolvin D1 inhibits TGF-beta1-induced epithelial mesenchymal transition of A549 lung cancer cells via lipoxin A4 receptor/formyl peptide receptor 2 and GPR32. Int. J. Biochem. Cell Biol. 2013, 45, 2801–2807. [Google Scholar] [CrossRef] [PubMed]
  292. Chiang, N.; Dalli, J.; Colas, R.A.; Serhan, C.N. Identification of resolvin D2 receptor mediating resolution of infections and organ protection. J. Exp. Med. 2015, 212, 1203–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Krishnamoorthy, S.; Recchiuti, A.; Chiang, N.; Yacoubian, S.; Lee, C.H.; Yang, R.; Petasis, N.A.; Serhan, C.N. Resolvin D1 binds human phagocytes with evidence for proresolving receptors. Proc. Natl. Acad. Sci. USA 2010, 107, 1660–1665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Chiang, N.; Fredman, G.; Backhed, F.; Oh, S.F.; Vickery, T.; Schmidt, B.A.; Serhan, C.N. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature 2012, 484, 524–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Dalli, J.; Winkler, J.W.; Colas, R.A.; Arnardottir, H.; Cheng, C.Y.; Chiang, N.; Petasis, N.A.; Serhan, C.N. Resolvin D3 and aspirin-triggered resolvin D3 are potent immunoresolvents. Chem. Biol. 2013, 20, 188–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Zheng, S.; Wang, Q.; D’Souza, V.; Bartis, D.; Dancer, R.; Parekh, D.; Gao, F.; Lian, Q.; Jin, S.; Thickett, D.R. ResolvinD1 stimulates epithelial wound repair and inhibits TGF-beta-induced EMT whilst reducing fibroproliferation and collagen production. Lab. Invest. 2018, 98, 130–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. Zong, L.; Chen, K.; Jiang, Z.; Chen, X.; Sun, L.; Ma, J.; Zhou, C.; Xu, Q.; Duan, W.; Han, L.; et al. Lipoxin A4 reverses mesenchymal phenotypes to attenuate invasion and metastasis via the inhibition of autocrine TGF-beta1 signaling in pancreatic cancer. J. Exp. Clin. Cancer Res. 2017, 36, 181. [Google Scholar] [CrossRef] [Green Version]
  298. Wu, R.F.; Huang, Z.X.; Ran, J.; Dai, S.J.; Lin, D.C.; Ng, T.W.; Chen, Q.X.; Chen, Q.H. Lipoxin A4 Suppresses Estrogen-Induced Epithelial-Mesenchymal Transition via ALXR-Dependent Manner in Endometriosis. Reprod. Sci. 2018, 25, 566–578. [Google Scholar] [CrossRef]
  299. Xu, F.; Zhou, X.; Hao, J.; Dai, H.; Zhang, J.; He, Y.; Hao, H. Lipoxin A4 and its analog suppress hepatocarcinoma cell epithelial-mesenchymal transition, migration and metastasis via regulating integrin-linked kinase axis. Prostaglandins Other Lipid Mediat. 2018, 137, 9–19. [Google Scholar] [CrossRef]
  300. Liu, Y.; Yuan, X.; Li, W.; Cao, Q.; Shu, Y. Aspirin-triggered resolvin D1 inhibits TGF-beta1-induced EMT through the inhibition of the mTOR pathway by reducing the expression of PKM2 and is closely linked to oxidative stress. Int. J. Mol. Med. 2016, 38, 1235–1242. [Google Scholar] [CrossRef] [Green Version]
  301. Sun, L.; Wang, Y.; Wang, L.; Yao, B.; Chen, T.; Li, Q.; Liu, Z.; Liu, R.; Niu, Y.; Song, T.; et al. Resolvin D1 prevents epithelial-mesenchymal transition and reduces the stemness features of hepatocellular carcinoma by inhibiting paracrine of cancer-associated fibroblast-derived COMP. J. Exp. Clin. Cancer Res. 2019, 38, 170. [Google Scholar] [CrossRef] [PubMed]
  302. Cezar, T.L.C.; Martinez, R.M.; Rocha, C.D.; Melo, C.P.B.; Vale, D.L.; Borghi, S.M.; Fattori, V.; Vignoli, J.A.; Camilios-Neto, D.; Baracat, M.M.; et al. Treatment with maresin 1, a docosahexaenoic acid-derived pro-resolution lipid, protects skin from inflammation and oxidative stress caused by UVB irradiation. Sci. Rep. 2019, 9, 3062. [Google Scholar] [CrossRef] [PubMed]
  303. Gdula-Argasinska, J.; Czepiel, J.; Wozniakiewicz, A.; Wojton, K.; Grzywacz, A.; Wozniakiewicz, M.; Jurczyszyn, A.; Perucki, W.; Librowski, T. n-3 Fatty acids as resolvents of inflammation in the A549 cells. Pharmacol. Rep. 2015, 67, 610–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Sun, Q.; Wu, Y.; Zhao, F.; Wang, J. Maresin 1 inhibits transforming growth factor-beta1-induced proliferation, migration and differentiation in human lung fibroblasts. Mol. Med. Rep. 2017, 16, 1523–1529. [Google Scholar] [CrossRef] [Green Version]
  305. Bazan, N.G. Neuroprotectin D1-mediated anti-inflammatory and survival signaling in stroke, retinal degenerations, and Alzheimer’s disease. J. Lipid Res. 2009, 50, S400–S405. [Google Scholar] [CrossRef] [Green Version]
  306. Li, H.; Hao, Y.; Zhang, H.; Ying, W.; Li, D.; Ge, Y.; Ying, B.; Cheng, B.; Lian, Q.; Jin, S. Posttreatment with Protectin DX ameliorates bleomycin-induced pulmonary fibrosis and lung dysfunction in mice. Sci. Rep. 2017, 7, 46754. [Google Scholar] [CrossRef] [Green Version]
  307. Balta, M.G.; Loos, B.G.; Nicu, E.A. Emerging Concepts in the Resolution of Periodontal Inflammation: A Role for Resolvin E1. Front. Immunol. 2017, 8, 1682. [Google Scholar] [CrossRef] [Green Version]
  308. Hong, S.; Tjonahen, E.; Morgan, E.L.; Lu, Y.; Serhan, C.N.; Rowley, A.F. Rainbow trout (Oncorhynchus mykiss) brain cells biosynthesize novel docosahexaenoic acid-derived resolvins and protectins-Mediator lipidomic analysis. Prostaglandins Other Lipid Mediat. 2005, 78, 107–116. [Google Scholar] [CrossRef]
  309. Raatz, S.K.; Golovko, M.Y.; Brose, S.A.; Rosenberger, T.A.; Burr, G.S.; Wolters, W.R.; Picklo, M.J., Sr. Baking reduces prostaglandin, resolvin, and hydroxy-fatty acid content of farm-raised Atlantic salmon (Salmo salar). J. Agric. Food Chem. 2011, 59, 11278–11286. [Google Scholar] [CrossRef] [Green Version]
  310. Bannenberg, G.L.; Aliberti, J.; Hong, S.; Sher, A.; Serhan, C. Exogenous pathogen and plant 15-lipoxygenase initiate endogenous lipoxin A4 biosynthesis. J. Exp. Med. 2004, 199, 515–523. [Google Scholar] [CrossRef] [Green Version]
  311. Haas-Stapleton, E.J.; Lu, Y.; Hong, S.; Arita, M.; Favoreto, S.; Nigam, S.; Serhan, C.N.; Agabian, N. Candida albicans modulates host defense by biosynthesizing the pro-resolving mediator resolvin E1. PLoS ONE 2007, 2, e1316. [Google Scholar] [CrossRef] [PubMed]
  312. Colas, R.A.; Ashton, A.W.; Mukherjee, S.; Dalli, J.; Akide-Ndunge, O.B.; Huang, H.; Desruisseaux, M.S.; Guan, F.; Jelicks, L.A.; Matos Dos Santos, F.; et al. Trypanosoma cruzi Produces the Specialized Proresolving Mediators Resolvin D1, Resolvin D5, and Resolvin E2. Infect. Immun. 2018, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Epithelial–mesenchymal transition (EMT).E-cadherin: Epithelial cadherin; EGF: Epidermal Growth Factor; FGF: Fibroblast Growth Factor; GATA: GATA Binding Protein; HGF: Hepatocyte Growth Factor; IGF: Insulin Like Growth Factor; ILK: Integrin Linked Kinase; JAK: Janus Kinase; LATS: Large Tumor Suppressor Kinase; miR34: microRNA 34; miR200: microRNA 200; N-cadherin: Neural cadherin; P-cadherin: Placental cadherin; PDGF: Platelet Derived Growth Factor; Runx2: RUNX Family Transcription Factor 2; SMAD: Sma- And Mad-Related Protein; SMO: Smoothened, Frizzled Class Receptor; SOX: SRY-Box Transcription Factor; TGF-β: Transforming Growth Factor Beta; Wnt: Wingless-Type MMTV Integration Site Family; ZEB1: Zinc Finger E-Box Binding Homeobox 1; ZO-1: Zona Occludens 1.
Figure 1. Epithelial–mesenchymal transition (EMT).E-cadherin: Epithelial cadherin; EGF: Epidermal Growth Factor; FGF: Fibroblast Growth Factor; GATA: GATA Binding Protein; HGF: Hepatocyte Growth Factor; IGF: Insulin Like Growth Factor; ILK: Integrin Linked Kinase; JAK: Janus Kinase; LATS: Large Tumor Suppressor Kinase; miR34: microRNA 34; miR200: microRNA 200; N-cadherin: Neural cadherin; P-cadherin: Placental cadherin; PDGF: Platelet Derived Growth Factor; Runx2: RUNX Family Transcription Factor 2; SMAD: Sma- And Mad-Related Protein; SMO: Smoothened, Frizzled Class Receptor; SOX: SRY-Box Transcription Factor; TGF-β: Transforming Growth Factor Beta; Wnt: Wingless-Type MMTV Integration Site Family; ZEB1: Zinc Finger E-Box Binding Homeobox 1; ZO-1: Zona Occludens 1.
Cancers 11 01841 g001
Figure 2. Inflammatory contribution to EMT. PGE2 and LTB4 produced from arachidonic acid can induce EMT in epithelial cancer cells. In contrast, LxA4 and AT-LxA4 from arachidonic acid can repress EMT in cancer cells. RvD1, RvD2, PDX, and MaR1 from DHA can suppress EMT in cancer cells or immortalized cells. AT-LxA4 (15(R)-lipoxin A4) is produced from 15(R)-HETE from arachidonic acid by aspirin trigger. Modified from Lee’s report [6]. 14-HDHA: 14-hydroxy Docosahexaenoic Acid; AA: arachidonic acid; AT-LxA4: Aspirin-triggered lipoxin A4; EPA: Eicosapentaenoic acid; DHA: Docosahexaenoic acid; DPA: Docosapentaenoic acid; MaR1: Maresin 1; MaR2: Maresin 2; PD1: Protectin 1; PDX: Protectin X; PGD2: Prostaglandin D2; PGF2a: ProstaglandinF2a; PGI2: Prostaglandin I2; PLA2:Phospholipase A2; LTB4: Leukotriene B4; LTC4: Leukotriene C4; LxA4: LIpoxin A4; RvD1-2: Resolvin D1-2; RvD3-6: Resolvin D3-6; RvE1-2: Resolvin E1-E2; RvE3: Resolvin E3; RvT1-4: 13-series resolvins; TXs: Thromboxane.
Figure 2. Inflammatory contribution to EMT. PGE2 and LTB4 produced from arachidonic acid can induce EMT in epithelial cancer cells. In contrast, LxA4 and AT-LxA4 from arachidonic acid can repress EMT in cancer cells. RvD1, RvD2, PDX, and MaR1 from DHA can suppress EMT in cancer cells or immortalized cells. AT-LxA4 (15(R)-lipoxin A4) is produced from 15(R)-HETE from arachidonic acid by aspirin trigger. Modified from Lee’s report [6]. 14-HDHA: 14-hydroxy Docosahexaenoic Acid; AA: arachidonic acid; AT-LxA4: Aspirin-triggered lipoxin A4; EPA: Eicosapentaenoic acid; DHA: Docosahexaenoic acid; DPA: Docosapentaenoic acid; MaR1: Maresin 1; MaR2: Maresin 2; PD1: Protectin 1; PDX: Protectin X; PGD2: Prostaglandin D2; PGF2a: ProstaglandinF2a; PGI2: Prostaglandin I2; PLA2:Phospholipase A2; LTB4: Leukotriene B4; LTC4: Leukotriene C4; LxA4: LIpoxin A4; RvD1-2: Resolvin D1-2; RvD3-6: Resolvin D3-6; RvE1-2: Resolvin E1-E2; RvE3: Resolvin E3; RvT1-4: 13-series resolvins; TXs: Thromboxane.
Cancers 11 01841 g002
Figure 3. EMT inducers from the chronic tumor microenvironment and EMT repressors from natural anti-inflammatory compounds and pro-resolving lipids. EMT signaling process is simplified due to space limitation. Dot line (--) means indirect effects. Arrow (→) means promotion or induction. Bar line (--I) indicates suppression. Inbox demonstrates the Lats2-mediated snail1 degradation. Receptors here have several subtypes coupled to different signaling pathway. For instance, EP4 use the PI3K/AKT pathway. This figure only covers limited parts of EMT receptor signaling. Modified from Lee’s report [6]. 4EBP1: Eukaryotic Translation Initiation Factor 4E Binding Protein 1; Act1: NF-κB activator 1; Akt: AKR mouse thymoma; ERK: Extracellular Signal-Regulated Kinase; FPRL1: Formyl Peptide Receptor-Like 1; GSK3: Glycogen Synthase Kinase 3 Beta; GRB2: Growth Factor Receptor Bound Protein 2; hnRNPE2: Heterogeneous Nuclear Ribonucleoprotein E2; IKK: Inhibitor Of Nuclear Factor Kappa B Kinase; JNK: JUN N-Terminal Kinase; LATS2: Large Tumor Suppressor Kinase; LEF: Lymphoid Enhancer Binding Factor; MEK: MAPK/ERK Kinase; MKK: Mitogen-Activated Protein Kinase Kinase; mTORC: mammalian Target of Rapamycin Complex; NF-κB: Nuclear Factor Kappa B; PI3K: Phosphatidylinositol-4,5-Bisphosphate 3-Kinase; PP2A: Protein Phosphatase 2A; RAF: Rapidly Accelerated Fibrosarcoma; RAS: Rat Sarcoma Viral Oncogene homolog; Runx2: RUNX Family Transcription Factor 2; S6K1: Ribosomal protein S6 kinase beta-1; SHCA: Src Homology 2 Domain Containing Adaptor protein; SIRT: Sirtuin; SMAD: Sma- and Mad-Related Protein; SOS: Son of Sevenless; STAT3: Signal Transducer And Activator of Transcription 3; TAK1: TGF- Activated Kinase 1; TCF: Transcription Factor; TRAF6: TNF Receptor Associated Factor 6; TWIST: Twist Family BHLH Transcription Factor; ZEB1: Zinc Finger E-Box Binding Homeobox 1.
Figure 3. EMT inducers from the chronic tumor microenvironment and EMT repressors from natural anti-inflammatory compounds and pro-resolving lipids. EMT signaling process is simplified due to space limitation. Dot line (--) means indirect effects. Arrow (→) means promotion or induction. Bar line (--I) indicates suppression. Inbox demonstrates the Lats2-mediated snail1 degradation. Receptors here have several subtypes coupled to different signaling pathway. For instance, EP4 use the PI3K/AKT pathway. This figure only covers limited parts of EMT receptor signaling. Modified from Lee’s report [6]. 4EBP1: Eukaryotic Translation Initiation Factor 4E Binding Protein 1; Act1: NF-κB activator 1; Akt: AKR mouse thymoma; ERK: Extracellular Signal-Regulated Kinase; FPRL1: Formyl Peptide Receptor-Like 1; GSK3: Glycogen Synthase Kinase 3 Beta; GRB2: Growth Factor Receptor Bound Protein 2; hnRNPE2: Heterogeneous Nuclear Ribonucleoprotein E2; IKK: Inhibitor Of Nuclear Factor Kappa B Kinase; JNK: JUN N-Terminal Kinase; LATS2: Large Tumor Suppressor Kinase; LEF: Lymphoid Enhancer Binding Factor; MEK: MAPK/ERK Kinase; MKK: Mitogen-Activated Protein Kinase Kinase; mTORC: mammalian Target of Rapamycin Complex; NF-κB: Nuclear Factor Kappa B; PI3K: Phosphatidylinositol-4,5-Bisphosphate 3-Kinase; PP2A: Protein Phosphatase 2A; RAF: Rapidly Accelerated Fibrosarcoma; RAS: Rat Sarcoma Viral Oncogene homolog; Runx2: RUNX Family Transcription Factor 2; S6K1: Ribosomal protein S6 kinase beta-1; SHCA: Src Homology 2 Domain Containing Adaptor protein; SIRT: Sirtuin; SMAD: Sma- and Mad-Related Protein; SOS: Son of Sevenless; STAT3: Signal Transducer And Activator of Transcription 3; TAK1: TGF- Activated Kinase 1; TCF: Transcription Factor; TRAF6: TNF Receptor Associated Factor 6; TWIST: Twist Family BHLH Transcription Factor; ZEB1: Zinc Finger E-Box Binding Homeobox 1.
Cancers 11 01841 g003
Figure 4. Structure of EMT inhibitors from natural sources.
Figure 4. Structure of EMT inhibitors from natural sources.
Cancers 11 01841 g004
Table 1. Lists of EMT inhibitors from natural sources.
Table 1. Lists of EMT inhibitors from natural sources.
NameSourceNameSource
ArctigeninAstraceaeGlycyrrhizinGlycyrrhiza glabra
BaicalinScutellaria baicalensis, Scutellaria laterifloraHonokiolMagnolia
BerberineBerberisHypaconitineAconitum
BetaninbeetsJatrophoneJatropha isabellei
BrusatolBrucea sumatranaLigustrazineNattō, fermented cocoa beans
CardamoninAlpinia katsumadaiLuteolinReseda luteola
CarnosolRosmarinus officinalis, Salvia pachyphyllaNimbolideAzadirachta indica
CelastrolTripterygium wilfordii, Celastrus regeliiNitidine chlorideZanthoxylum nitidium
CodonolactoneAtractylodes lanceaOstholeCnidium monnieri
Cordycepin Cordyceps militarisOxymatrineSophorae flavescentis
CryptotanshinoneSalvia miltiorrhizaPaeoniflorin Paeonia lactiflora
CurcuminZingiberaceaePaeonol Paeonia suffruticosa
DioscinDioscorea villosaParthenolideTanacetum parthenium
DelphinidinViola, DelphiniumPlectranthoic acidFicus microcarpa
Epigallocatechin-3-gallateGreen teaPiperlonguminePiper longum
EupatolideInula britannicaPlumbarginPlumbago
Galic acidgallnuts, sumac, witch hazel, tea leavesPolyphyllin IRhizoma of Paris
Gambogic acidGarcinia hanburyiPterostilbeneblueberries
GeduninAzadirachta indicaResveratrolgrapes
GenisteinGenista tinctoriaSalvianolic acidSalvia miltiorrhiza
GeraniinGeraniumsα−SolanineSolanum
GigantolCymbidium goeringiiSulforaphanecruciferous vegetables
Ginkgolic acidGinkgo bilobaTannic acidCaesalpinia spinosa
GinsenosidesGinsengWithaferin ASolanaceae
Table 2. Pro-resolving lipids. * Antagonist.
Table 2. Pro-resolving lipids. * Antagonist.
NameFormulaReceptorAnti-EMTSourceYields (pg/mg)
AA-derived lipoxins
LxA45S,6R,15S-trihydroxy-7E,9E,11Z,13E-ETEFPR2, GPR32+Toxoplasma gondii~0.43 ng
LxB45S,14R,15S-trihydroxy-6E,8Z,10E,12E-ETE
AT-LxA45S,6R,15R-trihydroxy-7E,9E,11Z,13E-eicosatetraenoic acidFPR2
AT-LxB45S,14R,15R-trihydroxy-6E,8Z,10E,12E-eicosatrienoic acid
EPA-derived resolvins
RvE15S,12R,18R-trihydroxy-6Z,8E,10E,14Z,16E-EPACMKLR1, BLT *, TRPV1 *,+ (?) Candida albicans
18S-RvE15S,12R,18S-trihydroxy-6Z,8E,10E,14Z,16E-EPACMKLR1,
BLT *
RvE25S,18R-dihydroxy-6E,8Z,11Z,14Z,16E-EPACMKLR1,
BLT *
Trypanosoma cruzi9.5–23.6
RvE317R,18R/S-dihydroxy-5Z,8Z,11Z,13E,15E-EPA
DHA-derived resolvins
RvD17S,8R,17S-trihydroxy-4Z,9E,11E,13Z,15E,19Z-DHAGPR32, FPR2, TRPV3 *,+Trypanosoma cruzi, Oncorhynchus mykiss1.8–7.0
RvD27S,16R,17S-trihydroxy-4Z,8E,10Z,12E,14E,19Z-DHAGPR32,
GPR18, FPR2,
+Oncorhynchus mykiss
RvD34S,11R,17S-trihydroxy-5Z,7E,9E,13Z,15E,19Z-DHAGPR32
RvD44S,5R,17S-trihydroxy-6E,8E,10Z,13Z,15E,19Z-DHA
RvD57S,17S-dihydroxy-4Z,8E,10Z,13Z,15E,19Z-DHAGPR32 Trypanosoma cruzi;
Oncorhynchus mykiss
RvD64S,17S-dihydroxy-5E,7Z,10Z,13Z,15E,19Z-DHA
AT-RvD17S,8R,17R-trihydroxy-4Z,9E,11E,13Z,15E,19Z-DHAFPR2, GPR32,
TRPV3 *,
+
AT-RvD27S,16R,17R-trihydroxy-4Z,8E,10Z,12E,14E,19Z-DHA
AT-RvD34S,11R,17R-trihydroxy-5Z,7E,9E,13Z,15E,19Z-DHAGPR32
AT-RvD44S,5R,17R-trihydroxy-6E,8E,10Z,13Z,15E,19ZDHA
AT-RvD57S,17R-dihydroxy-4Z,8E,10Z,13Z,15E,19Z-DHA
AT-RvD64S,17R-dihydroxy-5E,7Z,10Z,13Z,15E,19Z-DHA
n-3 DPA-derived resolvins
RvT17,13R,20-trihydroxy-8E,10Z,14E,16Z,18E-DPA
RvT27,8,13R-trihydroxy-9E,11E,14E,16Z,19Z-DPA
RvT37,12,13R-trihydroxy-8Z,10E,14E,16Z,19Z-DPA
RvT47,13R-dihydroxy-8E,10Z,14E,16Z,19Z-DPA
RvD1n-37,8,17-trihydroxy-8,10,13,15,19-DPA
RvD2n-37,16,17-trihydroxy-8,10,12,14,19-DPA
RvD5n-37,17-dihydroxy-8,10,13,15,19-DPA
DHA-derived protectins/neuroprotectins
PD1(NPD1)10R,17S-dihydroxy-4Z,7Z,11E,13E,15Z,19Z-DHA Oncorhynchus mykiss
PDX10S,17S-dihydroxy-4Z,7Z,11E,13Z,15E,19Z-DHA + (?)
22-hydroxy-PD110R,17S,22-trihydroxy-4Z,7Z,11E,13E,15Z,19Z-DHA
AT-PD110R,17R-dihydroxy-4Z,7Z,11E,13E,15Z,19Z-DHA
Ent-AT-NPD110S,17S-Dihydroxy-4Z,7Z,11E,13E,15Z,19Z-DHA
n-3 DPA-derived protectins/neuroprotectins
PD1n-310,17-dihydroxy-7,11,13,15,19-DPA
PD2n-316,17-dihydroxy-7,10,12,14,19-DPA
DHA-derived maresins
MaR17R,14S-dihydroxy-4Z,8E,10E,12Z,16Z,19Z-DHATRPV1 *, TRPA1 *+ (?)
MaR213R,14S-dihydroxy-4Z,7Z,9E,11E,16Z,19Z-DHA
7-epi-MaR17S,14S-dihydroxy-4Z,8E,10Z,12E,16Z,19Z-DHA
MaR-L114S,22-dihydroxy-4Z,7Z,10Z,12E,16Z,19Z-DHA
MaR-L214R,22-dihydroxy-4Z,7Z,10Z,12E,16Z,19Z-DHA
n-3 DPA-derived maresins
MaR1n-37S,14S-dihydroxy-8E,10E,12Z,16Z,19Z-DPA
MaR2n-313,14-dihydroxy-7Z,9,11,16Z,19Z-DPA
MaR3n-37,14-dihydroxy-8,10,12,16Z,19Z-DPA
* antagonist; +(?) confirmed in non-cancer cells.

Share and Cite

MDPI and ACS Style

Lee, C.H. Reversal of Epithelial–Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids. Cancers 2019, 11, 1841. https://doi.org/10.3390/cancers11121841

AMA Style

Lee CH. Reversal of Epithelial–Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids. Cancers. 2019; 11(12):1841. https://doi.org/10.3390/cancers11121841

Chicago/Turabian Style

Lee, Chang Hoon. 2019. "Reversal of Epithelial–Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids" Cancers 11, no. 12: 1841. https://doi.org/10.3390/cancers11121841

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

Article Metrics

Back to TopTop