Next Article in Journal
Identification of T Cell Receptors Targeting a Neoantigen Derived from Recurrently Mutated FGFR3
Next Article in Special Issue
Methods to Evaluate Changes in Mitochondrial Structure and Function in Cancer
Previous Article in Journal
Combined Inhibition of IAPs and WEE1 Enhances TNFα- and Radiation-Induced Cell Death in Head and Neck Squamous Carcinoma
Previous Article in Special Issue
Bone Metabolism Effects of Medical Therapy in Advanced Renal Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cannabinoids Transmogrify Cancer Metabolic Phenotype via Epigenetic Reprogramming and a Novel CBD Biased G Protein-Coupled Receptor Signaling Platform

Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L3N6, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(4), 1030; https://doi.org/10.3390/cancers15041030
Submission received: 7 January 2023 / Revised: 29 January 2023 / Accepted: 3 February 2023 / Published: 6 February 2023
(This article belongs to the Special Issue At the Interface of Genetics, Epigenetics and Metabolism in Cancer)

Abstract

:

Simple Summary

The epigenome is dynamic and flexible and can influence its phenotype by turning the genes on and off based on the environment to which it is exposed. Significant epigenetic changes detected are DNA methylation, histone modifications, and RNA-associated alterations. These modifications are potentially heritable across the genome, leading to transgenerational inheritance, and they can even be reversible based on lifestyle modifications. This review focuses on the environmental impact of cannabinoid factors and the subsequent combined effects on epigenetic modifications, such as cancer metabolism, through a biased G protein-coupled receptor signaling paradigm regulating several hallmarks of cancer.

Abstract

The concept of epigenetic reprogramming predicts long-term functional health effects. This reprogramming can be activated by exogenous or endogenous insults, leading to altered healthy and different disease states. The exogenous or endogenous changes that involve developing a roadmap of epigenetic networking, such as drug components on epigenetic imprinting and restoring epigenome patterns laid down during embryonic development, are paramount to establishing youthful cell type and health. This epigenetic landscape is considered one of the hallmarks of cancer. The initiation and progression of cancer are considered to involve epigenetic abnormalities and genetic alterations. Cancer epigenetics have shown extensive reprogramming of every component of the epigenetic machinery in cancer development, including DNA methylation, histone modifications, nucleosome positioning, non-coding RNAs, and microRNA expression. Endocannabinoids are natural lipid molecules whose levels are regulated by specific biosynthetic and degradative enzymes. They bind to and activate two primary cannabinoid receptors, type 1 (CB1) and type 2 (CB2), and together with their metabolizing enzymes, form the endocannabinoid system. This review focuses on the role of cannabinoid receptors CB1 and CB2 signaling in activating numerous receptor tyrosine kinases and Toll-like receptors in the induction of epigenetic landscape alterations in cancer cells, which might transmogrify cancer metabolism and epigenetic reprogramming to a metastatic phenotype. Strategies applied from conception could represent an innovative epigenetic target for preventing and treating human cancer. Here, we describe novel cannabinoid-biased G protein-coupled receptor signaling platforms (GPCR), highlighting putative future perspectives in this field.

1. Introduction

‘Epigenetic’ refers to “on top of or in addition to genetics.” A genome is the set of genetic information in the cell’s DNA. Every body cell has the same genome, though their respective phenotypes are different due to the control of gene expression [1]. An epigenome consists of a series of chemical compounds and complex external modifications that can cause changes in the genomic DNA without causing any changes to the DNA sequences of a gene, giving each cell a unique cellular and developmental identity [2]. The epigenome of an organism is dynamic and flexible and can influence its phenotype by turning the genes on and off based on the environment to which it is exposed [1]. Some significant epigenetic changes detected are DNA methylation, histone modifications, and RNA-associated alterations. These modifications are potentially heritable across the genome, leading to transgenerational inheritance. They can even be reversible based on lifestyle modifications [1]. This review focuses on the impact of cannabinoid factors and the subsequent combined effects on epigenetic modifications, such as cancer metabolism, through a biased G protein-coupled receptor signaling paradigm regulating several hallmarks of cancer.

2. Cannabis and the Endocannabinoid System

The cannabis sativa plant comprises over 550 chemical compounds [3]. Of these compounds, more than 100 have been identified as phytocannabinoids—plant-derived natural products that act on the endocannabinoid system (ECS) [3]. The most well-studied phytocannabinoids include delta-9 tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabigerol (CBG) [4]. THC is the primary cannabis psychoactive component and is part of the chemical class that constitutes most of the phytocannabinoid content in C. sativa. [3,5]. In comparison, CBD is a principal non-psychoactive constituent of cannabis with various pharmacological actions. At the same time, CBG is a minor constituent that serves as the direct precursor to CBD and THC [4]. Endocannabinoids and synthetic cannabinoids represent two other groups of chemical compounds that share similar structures and characteristics to the phytocannabinoids isolated from C. sativa. [6]. Endocannabinoids are chemical compounds naturally produced within the human body, while synthetic cannabinoids are produced in the laboratory [6]. The two major endocannabinoids are anandamide (AEA) and 2-arachidonic glycerol (2-AG) [7]. Synthetic cannabinoids represent a large group of chemical compounds, and much research has focused on developing synthetic cannabinoids for medical research purposes and promising therapeutic tools [8,9].
Cannabinoid receptors, belonging to the G-protein coupled receptor (GPCR) family, mediate the biological activity of both endogenous and exogenous cannabinoids [10]. The primary cannabinoid receptors within the body are the CB1 and CB2 receptors [11]. The activation of CB1 and CB2 receptors stimulates cellular signaling via the G alpha subunit (Gi/o), leading to the inhibition of adenylyl cyclase and the subsequent stimulation of protein kinases that play a crucial role in multiple signaling pathways, including mitogen-activated protein kinase (MAPK) pathways, phosphoinositide 3-kinase (PI3K) pathways, and cyclogeneses (COX) 2 pathways [12]. CB1 is the most abundant GPCR expressed in the mammalian brain [13]. Specifically, the CB1 receptor has high expression in brain regions involved in learning and memory (e.g., hippocampus), motor coordination (e.g., basal ganglia, cerebellum), and cognitive and emotional processes (e.g., amygdala, prefrontal cortex), allowing it to mediate most of the neurobehavioral effects of THC [11,14]. Although a long-standing belief existed that CB2 receptor expression was restricted to peripheral tissues and predominantly in immune cells, low expression of CB2 receptors in the brain has also been reported [15]. CB1 and CB2 receptor expression occurs in tissues outside the central nervous system, including skeletal muscle, liver, immune cells, and reproductive organs [16]. Furthermore, recent studies suggest that other GPCRs besides CB1 and CB2 could also mediate the intracellular effects of cannabinoids, such as GPR18 and GPR55 [17].
Cannabis is widely recognized for its recreational use due to its psychiatric effects, including anxiolysis and euphoria [18]. Moreover, medicinal cannabis is being increasingly explored as a potential therapy to treat various conditions, including chronic pain, vomiting, and nausea due to chemotherapy, depression, anxiety disorder, sleep disorder, and even cancer therapy [19,20]. Despite growing interest in the therapeutic potential of cannabinoids, further research is needed to develop a more comprehensive understanding of the adverse effects of cannabis use [21,22]. Of particular significance are studies that assess the impact of cannabinoids on the tumor microenvironment and epigenetic reprogramming.

3. Cannabis and Cancer Metabolism

Cannabis and the Endocannabinoid System’s Potential as a Cancer Therapeutic

Given the growing interest in cannabis, there is more research on the effects CB1 and CB2 have on tumor growth and metastasis [23]. Several studies have identified a correlation between cannabinoid receptor upregulation, endocannabinoid metabolic enzymes, and ligands in cancerous tissue [24,25,26,27]. If ligand and receptor upregulation is correlated with cancerous tissue, it suggests that the ECS plays a role in cancer modulation. Chakravarti et al., [26] found that cannabinoids mediate cancer cell signaling. However, a study by Velasco et al., [28] demonstrated that C. sativa L.’s cannabinoid and non-cannabinoid secondary metabolites have apoptotic, anti-inflammatory, and anti-metastatic properties suggesting cannabis’ properties may be harnessed for anti-cancer therapeutics.
Several in vitro and in vivo studies found that THC and JWH-133, a CB2 agonist, exert anti-proliferative and anti-angiogenic effects on ErbB-2 breast cancer by reducing MMP-2 (Matrix metalloproteinase) and MMP-9 (Matrix Metalloproteinase-9) levels [29,30,31]. Additionally, CBD reduces A549 lung cancer cells through upregulating tissue inhibitors of MMP-1 (TIMP-1) and reducing plasminogen activator inhibitor-1 (PAI-1) [32,33]. JWH-133 inhibits MMP-2 secretion to induce anti-angiogenic properties in A549 cells [34]. Moreover, cannabinoids appear to have anti-proliferative effects on tumors through anti-invasion properties and inhibiting MMPs, impacting cancer metastasis through ECM (Extracellular matrix) degradation [31]. Additionally, the results of some studies suggest cannabinoids may affect EMT (Epithelial-mesenchymal transition) and chemoresistance. For example, the cannabinoid 2-methyl-2′-F-anandamide reduces β-catenin nuclear translocation and transcriptional activity, inhibiting the Wnt/β-catenin pathway [35]. As a result, the expression of β-catenin target genes such as MMP-2, c-Myc, and cyclin D are reduced [35]. Additionally, mesenchymal markers such as N-cadherin, Slug, and Snail are reduced [35]. One study by Caffarel et al., [31] found that a selective inhibitor of the monoacylglycerol lipase (MAGL), a key enzyme in the human endocannabinoid system, that degrades 2-arachidonoylglycerol metabolism, reducing EMT markers and upregulates epithelial markers, including E-cadherin. In breast cancer, the GPCR receptor, GPR55, heterodimerizes with CB receptors, and its targeting impacts tumor growth [36]. Additionally, CB2 heterodimerization with human epidermal growth factor receptor 2 (HER2) and C-X-C chemokine receptor type 4 activates CB2 and inhibits signaling of the aforementioned (word for HER2 and C-X-C), suggesting its potential as an anti-tumor therapeutic [37]. Several studies report that dysregulation of the ECS during carcinogenesis may result in increased cancer aggression [38,39,40,41,42] as the endocannabinoid system is involved in various aspects of tumorigenesis [28,43,44,45]. For example, studies have linked cannabinoids to promoting apoptosis, inducing cell cycle arrest, and having a role in the invasion and self-renewal of tumor cells [28,44,45,46]. In pancreatic cancer, cannabinoids cause apoptosis by inhibiting pancreatic beta cell insulin receptor signaling, which interacts with the CB1 receptor from the MAPK and extracellular signal-regulated kinase (ERK) pathways [47,48]. In breast cancer, the inhibition of the epidermal growth factor (EGF), nuclear factor kappa B (NF-kB), ERK/Akt, and MMP 2 and 9 signaling pathways due to ECS activation results in anti-proliferative effects [49]. The ECS’s ability to inhibit tumorigenesis and other aspects of cancer growth makes it a novel target when treating various cancers. However, more research is needed to define the ECS’ role in tumor heterogeneity and how its signaling impacts the activation of other signaling pathways involved in cancer progression.

4. Cannabis and Epigenetics

4.1. Cannabis-Induced Epigenetic Effects

Although experimental data about the epigenetic modifications induced by cannabis remain sparse, studies have begun to reveal specific epigenetic modifications associated with cannabis exposure [20]. Modifications of the epigenetic marks such as DNA methylation, histone modifications, and changes in non-coding RNA (ncRNA) regulate gene expression by altering DNA accessibility and chromatin structure [50]. Recently, much research has focused on the association between developmental cannabinoid exposure and aberrant epigenetic markers that could modulate the risk of disease [51]. One study by Schrott et al. demonstrated an association between cannabis use in adult males with significant hypomethylation at 17 CpG sites within the autism candidate gene Discs-Large Associated Protein 2 (DLGAP2) in sperm [52]. In a follow-up animal study, the research group demonstrated intergenerational inheritance of this altered DNA methylation pattern, specifically identifying four CpG sites within the DLGAP2 gene with significant hypomethylation in cannabis-exposed offspring rats compared to controls [52]. Another study by Szutorisz et al. demonstrated that the offspring of parents exposed to THC during their adolescence had an increased propensity to self-administer heroin as adults despite not being exposed to THC themselves [53]. A subsequent investigation of the molecular mechanisms that enable parental THC exposure to influence addictive behavior in offspring revealed 1027 differentially methylated regions in the nucleus accumbens of rats with parental THC exposure [54].
Moreover, one study demonstrated that prenatal cannabis exposure decreased the expression of dopamine receptor D2 (D2R) mRNA—a consistent feature observed in adults with substance use disorders in the human ventral striatum [55,56]. Mechanistic studies in rats revealed that decreased D2R expression in THC-exposed offspring is mediated by epigenetic alterations, specifically by increased dimethylated lysine 9 (2meH3K9) and decreased trimethylated lysine-4 (3meH3K4) on histone H3 [56]. The authors propose that these epigenetic modifications could contribute to addiction vulnerability later in life [56]. Prenatal THC exposure can also lead to the differential expression of micro RNA (miRNAs) [57]. In one study, prenatal THC exposure was associated with the upregulation of miR-122-5p and the resultant downregulation of insulin-like growth factor 1 receptor (Igf1r) [57]. The downregulation of this receptor resulted in an increase in follicular apoptosis among THC-exposed offspring, leading the authors to speculate whether prenatal THC exposure may interfere with critical pathways in the developing ovary, ultimately leading to subfertility [57]. Although studies have begun to reveal associations between cannabis exposure and epigenetic effects, there is a significant gap in knowledge concerning the molecular pathways that mediate the epigenetic modifications induced by cannabis [58].

4.2. Molecular Insights into Cannabis-Induced Epigenetic Effects

Current research has only begun to uncover the pathways involved in cannabis-induced changes in DNA methylation, histone modifications, and miRNA expression [59]. Of the existing research, evidence supports the hypothesis that cannabis can alter the activity of histone and DNA-modifying enzymes [58]. For example, Khare et al. demonstrated that THC exposure caused a 3.5-fold increase in histone deacetylase 3 (HDAC3) expression in the BeWo trophoblast cell line [60]. HDAC3 is a component of the nuclear receptor corepressor (N-Cor) repressor complex, responsible for the deacetylation of lysine residues on the N-terminal of core histones [61]. Moreover, multiple studies have examined the effect of phytocannabinoids on DNA methyltransferases (DNMT) expression [62,63,64,65,66]. One study compared epigenetic changes in THC-treated and untreated myeloid-derived suppressor cells (MDSCs) [63]. This study revealed an increase in methylation at the promoter regions of DNMT3a and DNMT3b in THC-exposed cells, resulting in a significantly reduced expression of these enzymes [63]. A different study revealed that CBD increased global DNA methylation levels in human keratinocytes by selectively enhancing DNMT1 expression without affecting DNMT3a or DNMT3b expression [62]. Previous studies indicate that phytocannabinoids can modulate the expression of histone and DNA-modifying enzymes; however, the interactions and factors involved in these changes have yet to be elucidated.
Nevertheless, a study by Paradisi et al. investigating the role of the endocannabinoid anandamide in inhibiting human keratinocyte differentiation began to characterize the molecular mechanisms that modulate DNMT activity [67]. The investigators determined that anandamide can decrease the expression of genes involved in keratinocyte differentiation by increasing de novo DNMT activity in a CB1-dependent manner [67]. Most importantly, this study revealed that the interaction between anandamide and CB-1 increases DNMT activity through a p38 and, to a lesser extent, p42/44 MAPK-dependent pathway [67]. Similarly, a more recent study revealed that CBD upregulates the expression of specific genes involved in melanogenesis in a CB1-dependent manner, mediated by the activation of p38 and p42/44 MAPK; however, this study did not investigate the role of DNMTs [68].
Although signaling cascades are often described as isolated entities, biological signaling involves complex interactions between these cascades [69]. For instance, p38 and related kinases can act as cofactors in NF-κB activation, and significant overlap exists between the stimuli that activate NF-κB and the stimuli that activate MAPKs [69]. Interestingly, NF-κB signaling has yet to be studied in cannabinoid-mediated epigenetic effects, even though its association with MAPKs and its ability to induce epigenetic modifications support its involvement [69,70]. The NF-κB family consists of five structurally related transcription factors that form homo- and heterodimers with one another [71]. Inactivated NF-κB dimers are located in the cytoplasm in association with inhibitory proteins called inhibitors of kB (IkB) [72]. A plethora of factors can induce the phosphorylation and subsequent ubiquitin-dependent degradation of IkB, enabling the translocation of its associated NF-κB subunits to the nucleus, where they can regulate the transcription of many genes [72].
Notably, many studies have demonstrated the role of NF-κB in epigenetic regulation [70]. In human acute myeloid leukemia (AML) cells, Tage et al. demonstrated that NF-κB complexes with MUC1-C, a transmembrane oncoprotein at the DNMT1 promoter, drive DNMT1 transcription [73]. A different study supported these results by demonstrating that the introduction of a proteasome inhibitor known to inhibit NF-κB translocation caused the transcriptional repression of the DNMT1 gene, leading to the reduced expression of the DNMT1 protein [74]. Ultimately, this induced global DNA hypomethylation, enabling the re-expression of epigenetically silenced genes in AML cells [74].
Furthermore, NF-κB can modulate histone acetylation and methylation [75,76]. Numerous studies have demonstrated that can recruit and position chromatin modifiers, such as acetyltransferases and deacetylases, to the regulatory regions of different genes [70,76,77,78]. Additionally, one study revealed that the direct binding of NF-κB to the promoter of Jumonji domain-containing protein-3 (Jmjd3) is necessary to induce the expression of this histone demethylase [75]. The Jmjd3 demethylase erased the trimethylation marker of lysine 27 on histone H3 (H3K27me3) and was found to induce the expression of specific genes controlling cell fate and differentiation [75]. Another relevant study investigated the involvement of NF-κB in the epigenetic changes induced by the Epstein-Barr virus (EBV) during the transformation of resting B-lymphocytes into permanently growing lymphoblastoid cell lines [79]. This study demonstrated that the NF-κB pathway is essential to EBV-mediated histone methylation changes, including H3K27me3 and H3K4me3 [79]. Additionally, NF-κB was found to bind directly to the transcription start site of upregulated and downregulated miRNAs [79]. In this way, NF-κB directly mediates the expression of miRNAs that control the translation of mRNA transcripts involved in the growth transformation of B-cells [79].
Existing literature confirms the involvement of NF-κB in the mediation of specific epigenetic modifications; thus, it is an exciting candidate that may contribute to the epigenetic changes induced by cannabis. What is intriguing about the effects of cannabis on the NF-κB pathway is that certain cannabinoids act antagonistically to inhibit this pathway while others induce its activation [80]. Generally, studies have found that the interaction of CBD with CB1 and CB2 receptors inhibits the translocation of NF-κB to the nucleus, preventing it from exerting its effects on gene expression [49,81,82,83,84]. In contrast, most studies demonstrate that THC enhances NF-κB translocation to the nucleus [85,86,87,88]. One study showed that treatment with THC induced the phosphorylation of IkB-alpha in dendritic cells in a CB1- and CB2-dependent manner, enabling the release of NF-κB subunits and their translocation to the nucleus [85]. The literature suggests that specific components of cannabis can activate the signaling pathway, which may lead to epigenetic modifications; however, it has yet to be elucidated how these CB1 and CB2 ligands can induce opposing effects on the NF-κB pathway.
Nevertheless, the discovery of biased GPCR agonism may provide unprecedented insights into the opposing effects mediated by cannabinoids [89]. Upon ligand binding, GPCRs undergo a conformational change that leads to the activation of the heterotrimeric G-protein and its dissociation into the Gα subunit and Gβ/y heterodimer [90]. These subunits interact with many effectors, leading to complex G-protein-mediated signaling pathways [90]. Biased GPCR agonism refers to the ability of GPCR ligands to activate distinct intracellular signaling pathways by preferentially stabilizing different active conformational states of the receptor [91]. Both orthosteric ligands, such as THC, and allosteric ligands, such as CBD, tend to induce biased signaling at GPCRs [91]. Importantly, biased agonism and allosteric modulation have been observed at the CB1 receptor; however, to our knowledge, biased modulation of NF-κB signaling has yet to be studied in the context of CB receptors [91]. Cannabinoids may induce epigenetic modifications through the activation of these glycosylated receptors and the subsequent initiation of NF-κB; however, cannabinoids do not activate these receptors directly [92]. Rather, the binding of cannabinoids to CB receptors, as depicted in Figure 1, can stimulate the activity of RTKs, TNFRs, and TLRs through a process known as transactivation [93,94,95].
Matrix metalloproteinase-9 plays a crucial role in the remodeling of the extracellular matrix. With the novel discovery of receptor transactivation or GPCR agonist-bias signaling, MMP-9 has been shown to play a more prominent role in ECM remodeling. Through activation of associated GPCRs, such as the angiotensin AT1R and bradykinin receptors, MMP-9 is activated to induce activation of RTKs such as the TrkA receptor, EGFR, and IR as well as Toll-like receptors in the complex with NMBR, IR, and Neu1 in naïve (unstimulated) and stimulated RAW-blue macrophage cells. Here, a molecular link regulating the interaction and signaling mechanism(s) between these molecules on the cell surface uncovers a biased GPCR agonist-induced cell surface and intracellular Toll-like receptor (TLR) transactivation-signaling axis, mediated by Neu1 sialidase and the glycosylation modification of TLRs. The biased GPCR-signaling platform here potentiates Neu1 and MMP-9 cross-talk on the cell surface, which is essential for the transactivation of TLRs and subsequent cellular signaling.
The initiation of NF-κB signaling can begin with the activation of a variety of glycosylated transmembrane receptors, including receptor tyrosine kinases (RTKs), receptors for tumor necrosis factor (TNFR), and Toll-like receptors (TLRs) [101,102,103]. The signaling pathways induced by these receptors can all lead to the phosphorylation of IkBα, enabling the translocation of NF-κB to the nucleus [101]. Cannabinoids may induce epigenetic modifications by activating these glycosylated receptors and initiating NF-kB; however, cannabinoids do not activate these receptors directly [92]. Rather, the binding of cannabinoids to CB receptors can stimulate the activity of RTKs, TNFRs, and TLRs through a process known as transactivation, as depicted in Figure 1 [94,95,104].
The factors controlling the transactivation of glycosylated receptors by CB receptor agonists remain poorly understood. To this end, the biased NMBR-MMP9-Neu1 signaling axis highlighted throughout this review may be essential to transacting RTKs and TLRs by CB receptor ligands. A series of published studies demonstrated the involvement of this signaling axis in the transactivation of various receptors, including the insulin receptor [105], Trk receptors [106], and Toll-like receptors [107]. Notably, one study demonstrated that the GPCR agonist bombesin could induce TLR4 transactivation via this signaling paradigm, leading to the activation of NF-κB in BMC-2 and RAW-blue macrophage cells [107].

4.3. Future Directions of Cannabis-Induced Epigenetic Rewiring Research

The literature is conclusive that cannabis can evoke epigenetic modifications; however, the research on the pathways that mediate these epigenetic changes is still in its infancy [58]. A promising candidate for mediating the epigenetic effects induced by cannabis is NF-κB [108]. Existing literature demonstrates that certain cannabinoids, including THC and WIN 55, 212-2, enhance the activation of this pathway, while others, such as CBD, suppress its activation [80,87,109,110]. This finding supports the involvement of a ligand-biased signaling paradigm [91]. Specifically, the biased GPCR-Neu1-MMP9 signaling axis, as depicted in Figure 2, may control the transactivation of RTKs by the cannabinoid receptors, ultimately leading to the downstream activation of NF-κB and the induction of epigenetic reprogramming. Interestingly, Rozenfeld et al., [111] found heteromers of type I cannabinoid GPCR receptor (CB1R) with angiotensin II receptor type 1 (AT1R) mediating enhancement of angiotensin II-mediated signaling. Haxho et al., [105] found that angiotensin II GPCR receptor type I forms heteromers with NMBR in a multimeric receptor complex with Neu1, IRβ, and NMBR in naïve (unstimulated) and stimulated HTC-IR cells with insulin, bradykinin, angiotensin I and angiotensin II (Figure 2).
These innovative research findings have the potential to elucidate an entirely new signaling process for these receptors and cell responses. To gain a deeper understanding of how cannabinoids evoke epigenetic modifications, we must ascertain a more comprehensive understanding of the activity of the cannabinoid receptor itself and its interactions at the cell surface.

5. Neuraminidase-1

5.1. The Neuraminidase-1 Complex

Neuraminidase-1 (Neu-1) is a sialidase expressed in all mammalian tissues and is active on sialylated glycoproteins [114]. Recent research has found that Neu-1 is involved in a nerve growth factor (NGF)-induced TrkA receptor involving neuromedin B (NMBR) GPCR-signaling process in complex with MMP-9 to form a multimeric enzymatic axis with Neu-1 sialidase, which has a critical role in ligand-induced activation of RTKs and TLRs [115,116]. NMBR-MMP-9-Neu-1 cross-talk occurs when ligand-binding its receptor such as epidermal growth factor receptors (EGFR) [106], nerve growth factor TrkA [116], and insulin receptor (IR) [112]. Upon binding to an RTK or TLR, a ligand-dependent GPCR signaling occurs where NMBR GPCR signaling through a Gαi-protein subunit activates MMP-9 to induce Neu-1 activity [117]. The activation of Neu-1 is mediated by MMP-9, which removes the elastic binding protein (EBP) in complex with both Neu-1 and a protective protein cathepsin A. Neu-1 hydrolyzes α-2,3-sialic acid residues on glycosylated receptors to allow receptor activation and dimerization by removing steric hindrance [118]. Upon activation, downstream signaling occurs, resulting in conditions such as tumorigenesis and insulin resistance (Figure 1), which will be discussed in this review [112,115,116].

5.2. Neuraminidase-1′s Impact on Cancer and Diabetes Progression

There are many cellular mechanisms by which cancer and diabetes may occur. However, recently, glycosylation has been a critical factor in cancer metastasis. A review by Vajaria et al., [119] discussed that the glycosylation of proteins on a cell surface and the loss of E-cadherin due to MUC alterations has a significant role in tumor metastasis as it contributes to metastasis, angiogenesis, proliferation, and growth suppressor evasion. Moreover, since glycosylation plays an essential role in cancer, therapies targeting the mechanisms of these receptors’ glycosylation would likely have significant therapeutic potential.
The IR, a highly glycosylated receptor, has 18 glycosylation sites [120] and has implications for insulin resistance, diabetes, and cancer if the insulin-induced IR activation is not functioning correctly [121,122,123]. A study by Arabkhari et al. determined that when rat skeletal L6 myoblast cells were treated with Neu1 sialidase, desialylation of IR occurred, resulting in an increased proliferative response to lower insulin doses [124]. These findings indicate that glycosylation of the insulin receptor increases the receptor’s sensitivity when the ligand binds. Previous studies identify a novel cross-talk between Neu-1 and MMP-9 along with the neuromedin B GPCR, showing that Neu-1, MMP-9, and neuromedin B GPCR (NMBR) form a complex with IRβ subunits on a cell’s surface as neuromedin B is critical for insulin-induced IR activation [112]. The study’s results highlight that Neu-1 activation plays a role in the activation of the IR, supporting Fischoeder et al.’s findings that insulin mediates increases in MMP-9 via IR activation [125]. These findings highlight Neu-1′s importance in IR signaling and how overexpression of Neu-1 may result in overexpression of IR, therefore causing insulin resistance, diabetes, and cancer.
The EGFR is an ErbB RTK receptor that is a critical pathway involved in cell growth, regulation, proliferation, and differentiation of cells [126]. The EGFR is activated along with cytoplasmic tyrosine kinase activity when the EGF binds to the extracellular domain of the EGFR [127]. It is well known that EGFR is often mutated or overexpressed in various cancer types and is a target for anti-cancer therapies [128]. Moreover, the receptor exists as a single, highly glycosylated subunit, making it susceptible to overexpression and cancer. A study by Gilmour et al. found Neu-1 and MMP-9 cross-talk with EGF receptors on EGFR-expressing cells and human pancreatic MiaPaCa-2 and PANC-1 cancer cells, which is critical for EGF activation of EGFRs and eventual cellular signaling [116]. The cross-talk likely occurs from EGFR’s activation of NMBR as it activates the Gαi-protein, triggering the activation of MMP-9 and the removal of the EBP, which activates Neu-1 [129]. The activated EGFRs can then promote tumorigenesis by activating the JAK/STAT (Janus kinase/signal transducers and activators of transcription), PI3K/Akt, and MAPK signaling pathways [106,130,131].
Neu-1′s involvement in regulating glycosylated growth factors receptors such as IR, EGFR, and TrkA indicates it impacts a tumor’s characteristics, such as cell growth, proliferation, and differentiation [106]. Moreover, Neu-1, a regulator of these receptors, is a novel targeted approach for cancer therapeutics. Haxho et al., [132] reported a review on the role of mammalian Neu1 in complex with MMP-9 and G protein-coupled receptors tethered to RTKs and TLRs as a major target in multistage tumorigenesis. The therapeutic efficacy of targeting Neu1 may disrupt several molecular signaling pathways. Given the ability of oseltamivir phosphate (OP) to increase E-cadherin expression and decrease N-cadherin and VE-cadherin expression, as previously reported [133], tumors treated with this OP drug may become more adherent to the surrounding tissue and not metastasize. Haxho et al., [132] proposed a graphical illustration (Figure 3) depicting a Snail-MMP9 signaling axis [134], maintaining several important cancer growth factor receptor signaling platforms in promoting Neu1-MMP9 cross-talk in complex with glycosylated receptors. OP treatment strategies under dose dependence would have a horizontal targeted approach, in which different oncogenic signaling pathways involved in tumorigenesis are targeted with promising therapeutic intent.

5.3. Inhibition of the Neu-1 Complex as a Potential Therapeutic to Cancer

Cancer treatments aim to modulate cellular pathways essential for cancer survival and growth, such as RAS, EGFR, vascular endothelial growth factor VEGF, and MMPs. Moreover, identifying and inhibiting complexes influencing these pathways, such as Neu-1 and MMP-9, is a beneficial approach to decreasing tumorigenesis [135]. An essential aspect of utilizing therapies that target different molecular pathways is that the risk of drug resistance is mitigated [136,137,138]. Oseltamivir phosphate (OP), also known pro-drug of Tamiflu, is a licensed medication that targets the neuraminidase protein [139]. Many studies have highlighted the efficacy and therapeutic potential of OP on various types of cancers, such as breast and pancreatic cancer [116,133,140]. Studies performed by Amith et al., [141] and Abdulkhalek et al., [142] found that OP specifically targeted and inhibited Neu-1 activity. Following Neu-1 inhibition, OP exerted its anti-cancer effects through increased E-cadherin expression and decreased VE-cadherin and N-cadherin in human pancreatic cells (PANC1) as EMT and E-cadherin loss [133]. Cell-to-cell adhesion promotes metastasis and tumor progression [133]. OP inhibition of Neu-1 inhibition was also found to downregulate EGFR pathways such as the JAK/STAT, PI3K/Akt, and MAPK [133], all of which are involved in tumorigenesis [106,130,131].
A significant issue regarding cancer therapies is the occurrence of chemo and drug resistance. A study performed by Zhang et al., [143] repurposed the drug aspirin (ASA) as it is an inhibitor of NF-κB signaling, which is involved in the upregulation of MMP-9 [144,145]. The study found that ASA increases the efficacy of gemcitabine, a chemotherapy medication, by inhibiting inflammatory proteins such as NF-κB and reducing the self-renewal potential of cancer stem-like cells (CSCs). They also found that ASA has a preventive effect on the development of pancreatic ductal adenocarcinoma (PDA) and targets highly aggressive PDA cells [143], indicating that ASA has strong therapeutic potential in treating cancer. A recent discovery found that ASA and celecoxib, a selective COX-2 inhibitor, dose-dependently inhibit EGF-induced sialidase activity in PANC1 and MiaPaCa-2 cells through the inhibition of Neu-1 cleavage of the α-2,3-sialic acid [146]. Using this information as a rationale, Sambi et al., [147] utilized the triple combination of ASA, metformin (Met), and OP treatment with tamoxifen on breast cancer cell lines. They found that the cocktail reduced cell proliferation, upended tamoxifen chemoresistance, and increased apoptotic activity in MDA-MB-231 triple-negative breast cancer cells and their tamoxifen-resistant variant. These results suggest a therapeutic approach to multistage tumorigenesis in triple-negative breast cancer.
Recently, Qorri et al., [148,149] investigated the therapeutic potential of OP, ASA, and the chemotherapeutic agent gemcitabine (GEM) in pancreatic ductal adenocarcinoma cancer (PDAC) cells. They found that ASA+OP+GEM upended MiaPaCa-2 and PANC-1 pancreatic cell survival mechanisms, including viability, promoting apoptosis, and expressing extra-cellular matrix proteins. Moreover, it is a novel approach to targeting the survival mechanisms of pancreatic cancer metastasis.
In preclinical studies, Qorri et al., [149] reported that a continuous therapeutic targeting of Neu-1 using parenteral perfusion of oseltamivir phosphate (OP) and aspirin (ASA) with gemcitabine (GEM) treatment significantly disrupted tumor progression, critical compensatory signaling mechanisms, EMT program, cancer stem cells (CSC), and metastases in a preclinical mouse model of human pancreatic cancer. In addition, they demonstrated that ASA- and OP-treated xenotumors significantly inhibited the metastatic potential when transferred into animals.

6. MMP-9 as a Therapeutic Target of Cancer

6.1. MMP-9 and Its Interaction with the Neu-1 Complex

As mentioned previously, MMP-9 has an essential role with Neu-1 as it forms a trimeric complex involved in ligand-induced activation of RTKs [115,116]. Snail’s transcription factor mediates ovarian tumor neovascularization and is closely associated with MMP-9 expressions in invasive tumors due to similarities in extra-cellular matrix remodeling during invasive processes [134,150]. Jorda et al., [151] found that Snail can up-regulate the expression of MMP-9 by activating the MAPK and PI3K signaling pathways, indicating a link between them. The connection suggests that the Snail-MMP-9 signaling axis is the missing component in promoting growth factor glycosylation modification that results in Neu-1-MMP-9 cross-talk at the ectodomain of RTKs [134]. From there, downstream signaling occurs, involving various pathological conditions, including tumorigenesis and insulin resistance, making MMP-9 a viable target for cancer and diabetes therapies [112,115,116].

6.2. Inhibition of the NF-κB Pathway Results in Downregulation of MMP-9

NF-κB is a transcription factor that regulates innate and adaptive immune functions. Moreover, its activation induces pro-inflammatory gene expression, such as cytokines and chemokines [71]. NF-κB activates in response to infectious agents and pro-inflammatory cytokines from the IκB kinase complex. Studies have found that the deletion of the inhibitor kappa-B kinaseβ (IKKβ) in epithelial cells decreases tumor incidence, and the deletion of IKKβ in myeloid cells decreases tumor size, indicating that inactivating the IKK/NF-κB alters the formation of inflammation-associated tumors [152]. Moreover, the NF-κB pathway has a strong association with cancer. Various studies have shown that the promoter region of the MMP-9 gene includes binding sites for NF-κB and activator protein-1. MMP-9 expression requires both transcription factors to be present [153,154,155,156,157]. A study inhibiting the expression of NF-κB in CT26 colon cancer cells through the overexpression of the IκB-α super-repressor found a decrease in MMP-9 expression, highlighting a link between the two [158].
Cancer, a metabolic disease, often has metabolic alterations in cancerous cells, such as an increase in glycolytic metabolism rate known as the “Warburg effect” [159]. Interestingly, therapies such as the ketogenic diet (KD) have been developed to create a metabolic shift towards fatty acid utilization and develop antitumor activity [160], as recent research has shown that a KD may inhibit tumor progression by altering the activity of inflammatory responses [161]. A study by Zhang et al., [162] examined the impact of the limited carbohydrate KD on growth and apoptosis in CT26+ colon cancer cells and the role of protein expression, such as the NF-κB pathway. They found that KD treatment slows colon cancer progression by significantly reducing the protein expression of NF-κB 65, phosphor NF-κB p65, and MMP-9. The results indicate KD downregulates phosphor NF-κB expression, inhibiting the protein levels of their downstream targets such as MMP-9 [162].
Nitric oxide (NO) is also a regulator of MMP-9 activation [163,164,165,166,167]. In-vitro studies show murine macrophages secreting increased levels of MMP-9 during exposure to low NO concentrations due to suppressing the tissue inhibitor of metalloproteinase 1. However, increasing NO concentrations in guanylyl-cyclase-dependent and independent signaling pathways decreased MMP-9 activity [166]. Several studies have observed that the differences in MMP-9 expression from varying NO treatments also involve the regulation of NF-κB as its activation increases or decreases depending on low or high concentrations of NO [167,168], suggesting NO affects NF-κB function. Reports indicate that opioid agonists can induce apoptosis in cancer cells and inhibit angiogenesis [168,169,170,171]. Morphine, an opiate, has been shown to impact tumor progression from its effects on MMP-9 expression and increase NO production [169,170,172,173,174]. A study by Roy et al., [175] found that morphine modulates NF-κB transcription factor and has opioid functions in various cells, indicating morphine regulates MMP-9 production by interfering with NF-κB binding to the promoter region of MMP-9 via opioid receptor-mediated and NO-dependent mechanisms [175,176]. Moreover, morphine as an anti-cancer drug through MMP-9 inhibition shows therapeutic potential. However, despite determining that morphine’s anti-cancer properties, its activation and inhibition vary with cancer and cell types, the dose of morphine, and other experimental factors, suggesting more information is needed on the mechanisms of these receptors and pathways [173,177,178,179,180,181,182].
Epigenetics is the interaction between the genome and the environment as changes to one’s environment result in modifications such as DNA methylation, histone modification, and altering chromatin structure, resulting in changes in gene expression [50]. These changes have been thought to have the potential to silence genes promoting cancer, making epigenetics a promising area of research for novel cancer therapeutics [183]. A study by Sung et al., [184] found that anacardic acid inhibits the acetylation of p65, a subunit of NF-κB, by suppressing histone acetyltransferase (HAT) activity, thereby impeding TNF induced NF-κB activation. The suppression of HAT activity prevents the addition of acetyl groups which typically promote gene expression. They also found that anacardic acid decreases MMP-9 activation due to NF-κB being unable to bind to the promoter of MMP-9. These results are supported by other studies [185] and prove that inhibition of the NF-κB pathway decreases the expression of MMP-9 and suggests epigenetic changes may be used in cancer therapeutics. MicroRNAs, another epigenetic modulator, are single-stranded, non-coding sequences of RNA that are involved in gene expression regulation at the post-transcriptional and translational levels [186,187]. NF-κB regulates four miRNAs, miR-10b, miR-17, miR-21, and miR-9, which are involved in tumor growth and metastasis [188,189,190,191]. Using the luciferase reporter assay, Li et al., [192] found that p65 NF-κB directly regulates miR-10b, miR-17, miR-21, and miR-9 and that the NSAID, sulindac (SS) inhibits the nuclear translocation of NF-κB by decreasing the phosphorylation of IKKβ and IκB. As a result, SS inhibits tumor cell invasion by hindering the NF-κB pathway and miRNAs involved in tumorigenesis. SS inhibiting NF-κB increases the likelihood that MMP-9 is decreased as reduced NF-κB expression is associated with a reduction in MMP-9. However, research needs to be conducted to determine this hypothesis.

6.3. Inhibition of the COX-2 Decreases MMP-9 Activity

Cyclooxygenase (COX) -1 and -2 are the most well-known oxygenases involved in catalyzing molecular oxygen into organic compounds. COX-1 is typically expressed in most tissues, whereas COX-2 responds to cytokines and growth factors. They catalyze the reduction of arachidonic acid to prostaglandin G2 (PG) and PGE2 [193]. PGH2 often undergoes metabolism by enzymes to form PGs, which exert their effects through GPCR mechanisms [194]. Research has found that several cancers, including breast, colon, and pancreatic cancers, have increased levels of PG [195,196,197]. Additionally, a 2001 study by Surh et al., [197] found that the NF-κB pathway modulates the expression of COX-2, resulting in metastasis and tumorigenesis, highlighting a link between the NF-κB, COX-2, and cancer. Several studies have found that MMP-9, COX-2, and VEGF expressions exist in many tumor tissues [198,199]. One study found that VEGF expression in colon cancer increases following transfection with COX-2 and that NS398, a COX-2 inhibitor, reduces its effect, indicating COX-2 stimulates tumor angiogenesis through the VEGF pathway [200]. Studies by Larkins et al., [201] and others [202] support the previous findings as they found that selective inhibitors of COX-2 down-regulate MMP-9, -2, and VEGF expression, thereby reducing head and neck squamous cell viability. Moreover, the inhibition of COX-2 is a promising therapeutic in inhibiting MMP-9 and the Neu-1 complex.
The non-steroidal anti-inflammatory drug (NSAID) aspirin inhibits COX-1 and -2 isoforms, exerting anti-inflammatory, antipyretic, and analgesic effects. However, the exact mechanism needs to be studied further, but COX-dependent and independent mechanisms have been proposed [203]. One COX-dependent mechanism proposed is that ASA inhibits COX-2, resulting in the downregulation of phosphatidylinositol 3-kinase (PI3K) signaling [204]. The PI3K pathway is involved in MMP-9 expression [205], which explains why regular ASA use following a colorectal diagnosis increases survival among patients with PIK3CA tumors [204]. ASA also acts through COX-independent pathways to inhibit IKKβ, preventing NF-κB activation and contributing to ASA’s anti-cancer effects [205]. A study by Xue et al., [206] found that aspirin decreases COX-2 mRNA expression, thereby reducing MMP-9 content in macrophages derived from THP-1 cells due to a decrease in mPGES-1 mRNA. They verified that MMP-9 formation was through the COX-2/mPGES-1 pathway when MMP-9 expression increased following increases in mPGES-1 production due to PGE2 stimulation [205].
Curcumin, a naturally occurring polyphenol derived from turmeric, demonstrates anti-cancer properties through modulating various molecular targets such as STAT3, EGFR, PI3KAkt/mTOR, E-cadherin, MMPs, and COX-2 [207]. Curcumin downregulates COX-2 and EGFR expressions since PGE2 transactivates the EGFR pathway to promote cancer cell motility, indicating a cross-talk between the two pathways. The association between the two pathways is a reduction in extracellular signal-regulated kinase (ERK) activity, thereby increasing apoptosis and decreasing cell survival [208]. Again, COX-2 downregulation through curcumin is associated with inhibiting the NF-κB pathway. Here curcumin suppresses IKK, inhibiting phosphorylation and degradation of IKB-kinase alpha and nuclear translocation of p65 [209]. MMP-9 protein levels decreased following curcumin treatment through downregulating ERK signaling pathways that have been established to involve COX-2 [210]. However, not every cancer cell contains COX-2, as studies have found that MiaPa-Ca-2 pancreatic cancer cells have absent levels of COX-1 and -2, and PANC-1 cells have high COX-1 expression and little COX-2 expression [148]. The reduction of COX in pancreatic cancer cells does create problems regarding treatment as the use of NSAIDs becomes cell and dose-dependent to ensure proper treatment.
Additionally, more research can be conducted on the mechanisms of COX on cancers to help determine the exact mechanisms of COX inhibitors such as ASA and celecoxib [146]. However, these findings support COX-2′s role in the Neu-1 complex through its ability to inhibit MMP-9 through the NF-κB pathway. Moreover, COX-2′s role in MMP-9 inhibition indicates its potential as an anti-cancer therapeutic.

7. Effects of Biased GPCR Agonists on the Insulin Receptor (IR)

Bias GPCR Agonism and the Insulin Receptor

The IR is a tyrosine kinase transmembrane signaling protein that regulates cell differentiation, growth, and metabolism. The IR is a ligand-activated receptor whose primary function is metabolic regulation [211]. The receptor comprises two α and two β subunits linked through disulfide bridges [212,213,214]. Insulin receptor binding induces a conformational change, activating IR-β kinase activity [215]. As a result, the downstream effects of IR signaling occur, such as glucose homeostasis for various cell types [216,217]. However, insulin resistance may occur when normal or increased insulin levels produce a reduced biological response, usually when there is reduced insulin sensitivity [218,219]. Although it is suggested that insulin resistance occurs due to the downregulation of the IR, or insulin receptor substrate (IRS) proteins, the precise mechanism of insulin resistance is unknown [123,220]. Insulin resistance has clinical implications in many pathological conditions, including cancer, type 2 diabetes (metabolic syndrome), cardiovascular disease, hypertension, and polycystic ovary syndrome (PCOS) [220]. Blaise et al., [221] found that κ-elastin (kE) exposure decreases sialic activity on the β-chain of the IR, suggesting that Neu-1 desialylates the IR. This study’s results confirm the findings from others [222] that Neu-1 has a significant role in IR regulation, as mice with a Neu-1 deficiency develop hyperglycemia and insulin resistance twice as fast as the wild-type when exposed to a high-fat diet.
Moreover, a neuromedin B GPCR-MMP-9-Neu-1 complex is tethered to IRβ subunits, indicating that MMP-9-Neu-1 cross-talk occurs after insulin binding to its receptor initiates GPCR signaling/MMP-9 activation to induce Neu-1 [112]. Evidence of this was found in a study by Schlien et al., [223], where insulin binding to its receptor induces a conformational change at the C-terminus domain. Given that GPCR agonists positively regulate the insulin receptor, and MMP-9 and Neu-1 are associated with the IR, biased GPCR agonism of these GPCRs likely affects the receptor. A study by Haxho et al., [105] found that bradykinin, angiotensin I, and II dose-dependently induce sialidase activity in HTC-IR cells, indicating they exist in a multimeric receptor complex with Neu-1, IRβ, and NMBR. These findings confer that Neu-1-mediated desialylation of the IRβ subunits transactivates insulin receptors following GPCR agonism and IR glycosylation. Moreover, the results confirm findings from previous literature [112] of NMBR’s regulatory role in inducing Neu-1 sialidase and MMP-9 cross-talk for IRβ desialylation and receptor activation. Studies show that IR and GPCR cross-talk regulate various cellular processes, and the dysfunction between the two receptors results in diseases such as metabolic syndrome, type 2 diabetes, and cancer [224]. The dysfunction between the receptors can be caused by GPCR agonists, which can positively regulate IR signaling despite the absence of insulin [105]. A study by Isami et al., [225] confirms Haxho et al.’s, [105] findings that GPCR agonism mediated by Neu-1 transactivates the IR without insulin, possibly enabling various pathological diseases.

8. Bias GPCR Agonism on the IR and Cancer

Many studies have implicated that the insulin-like growth factor (IGF) and insulin signaling pathways influence carcinogenesis and tumor progression [226,227]. IGF-1 binds to both the IGF-1 receptor (IGF1R) and IR and, as a result, promotes mitogenic signaling events, inhibits apoptosis, and increases cell proliferation [227]. For example, analyses of specific IR ELISAs found that 80% of breast cancers have significantly higher IR expression than normal breast tissue. The study also found that 20% of the cancer tissue had IR expression ten times the mean value of normal breast tissue [226]. IR overexpression in cancers makes the IGF1R, IR, and the pathways they activate promising therapeutic targets [228,229,230]. For example, when IGF-1 binds to IGF1R and IR, the PI3K-AKT-TOR and RAF-MAPK pathways activate, promoting cell survival and proliferation [227,231]. These pathways are also involved in MMP-9/Neu-1 cross-talk, highlighting that transactivation of the IR is likely mediated by Neu-1 to promote signaling pathways involved in tumorigenesis, and its downregulation will likely lead to a decrease in cancer viability [231,232,233]. Further evidence of Neu-1′s involvement in IR signaling is that in-vitro and in-vivo studies have concluded that insulin affects tumor progression by acting on the IR and not solely by IGF-1R cross-talk [234,235]. GPCR agonists support these conclusions by indirectly transactivating insulin receptors via Neu-1 mediated desialylation of IRβ subunits without insulin, highlighting that the IR plays a critical role in maintaining the vitality of cancer cells and creates a therapeutic target for the treatment of these diseases [224].
Experimental studies have shown that OP and NMBR inhibitor BIM-23127 inhibits independent autophosphorylation of IRβ and IRS-1 in HTC-IR cells, blocking Neu-1-mediated transactivation of the IR, which may be used as a therapeutic target for cancer [112]. A study by Rozengurt et al., [235] found that the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) is critical for IR/IGF-1R and GPCR signaling. Metformin, an antidiabetic drug, induces AMP kinase activation, negatively regulating mTORC1 and suppressing the growth of PANC1 cells [236,237], showing that IGF-1R disruption decreases tumorigenesis as IR/IGF-1R cross-talk is disabled. Linsitinib, a small-molecule tyrosine kinase inhibitor (TKI), blocks autophosphorylation of IGF1R/IR and exerts promising anti-tumor activity [238]. However, clinical trials evaluating its efficacy in metastatic breast cancer found that toxicities such as hyperglycemia would occur as IR disruption would impact proper glucose homeostasis [239]. Similarly, BMS-754807, an ATP-competitive TKI of IGF1R and IR, inhibits pancreatic cancer cell lines. However, like Linsitinib, clinical trials were halted following patients reporting side effects such as impaired glucose tolerance, fatigue, and nausea likely due to excessive IR inhibition [240]. Although discouraging, these results promise that proper targeting of the IR and IGF-1R will inhibit cancer cell vitality and growth. Bromo- and extra-terminal domain (BET) proteins, such as BRD2, BRD3, BRD4, and BRDT, are epigenetic regulators that connect chromatin modifications to the transcriptional activation of genes. They have a critical role in homeostasis, and cell survival, as their dysfunction plays a role in cancer. BET proteins regulate EMT (Snail), which has a significant role in MMP-9 expression and Neu-1 activation [240]. BET protein action and the IGF system are connected as the IGF1R gene directly targets BRD4 in various cancer cell lines. Treatment of RKO-1, PC-3, and MCCL-357 cells with BET inhibitors JQ1 or MS417 reduces BRD4 binding to IGF1R, reducing mRNA expression, and inhibiting the PI3K/Akt pathway [241]. PI3K and BET inhibition hinders the PI3K pathway, thereby reducing cell growth and vitality in cell models [241]. However, more research must be conducted on BET inhibition and MMP-9/Neu-1 activation.
The insulin receptor substrate (IRS) proteins play a pivotal role in metabolism and cancer. Under normal physiological conditions, these proteins are involved in IR/IGF-1R-mediated metabolic regulation by amplifying the PI3K signaling pathway to activate the serine-threonine kinase AKT [242,243]. Evidence of this is that IRS proteins have multiple PI3K binding motifs to recruit and activate PI3K via SH2-domains [244]. Additionally, IRS proteins are signaling intermediates in insulin-regulated glucose homeostasis by promoting glucose uptake [245]. However, dysregulation and overexpression of these proteins lead to pathological conditions such as cancer and diabetes. Many studies have found that IRS-1 negatively regulates tumor progression, whereas IRS-2 promotes tumor behavior through metabolism regulation [246]. As a result, targeting these proteins may have strong potential in novel cancer therapeutics. A study by Zhao et al., [247] found that miR-766, a miRNA, directly targets IRS2 and inhibits activation of the PI3K/Akt pathway to reduce papillary thyroid cancer (PTC) malignancy. However, a limitation of the study is that the association between miR-766 and the prognosis of PTC patients was not analyzed, making its clinical implications unclear. Another study found that long noncoding adipogenesis regulatory factor-antisense RNA 1 (ADIRF-AS1) is overexpressed in osteosarcoma (OS).
Moreover, ADIRD-AS1 impedes tumor growth and is a competitive endogenous RNA for miR-761 as it is removed from IRS1, leading to increased IRS1 overexpression. The study’s results suggest that reducing miR-761 in OS cells neutralizes the effect of ADIRF-AS1 as IRS1 is restored and no longer overexpressed [248]. These two studies highlight the therapeutic potential of miRNAs and how their use may be efficacious in reducing IR expression in cancer [159,160]. Another miRNA, miR-30e, is downregulated in many cancer cell lines [249,250]. A study by Liu et al., [251] found that miR-30e recognizes the 3′-UTR of IRS1 transcripts, indicating that miR-30e expression impacts IRS1 expression. Evidence of this is that cells expressing miR-30e abolish IRS1 in breast cancer cells, thereby suppressing breast cancer cell growth [251]. A preclinical study by Garofalo et al., [252] supports the evidence of inhibiting IRS-1 and -2 in treating cancer. They found that NT157, a selective inhibitor of IRS-1 and -2, decreases the expression of these proteins and downregulates IGF-1R-mediated AKT activation. As a result, NT157 treatment leads to cell cycle arrest, apoptosis, and reduced cell motility in OS cells, thereby reducing OS metastasis [252].

9. Conclusions

Here, we reviewed reports demonstrating that GPCR agonists bombesin, bradykinin, angiotensin I, and angiotensin II, each significantly and dose-dependently induced sialidase activity in live IR [105,112], EGFR [116], Trk [106], and TLR [107,115,141,142] expressing cells, in vitro. If Neu1 sialidase activity is required for these glycosylated receptor activations in a mechanism dependent on GPCR Gαi signaling and subsequent MMP9 activation, this led us to hypothesize that GPCR agonists may indirectly transactivate receptors via Neu1-mediated desialylation process and downstream signaling. The reported data support this hypothesis. Furthermore, GPCR agonist-induced sialidase activity in these cells resulted in receptor ligand-independent signaling in these cells. This process is blocked by Neu1 inhibitor oseltamivir phosphate and the neuromedin B GPCR (NMBR) inhibitor BIM-23127. These findings are consistent with our previous report describing the regulatory role of NMBR in inducing Neu1 sialidase and MMP9 cross-talk required for IRβ desialylation and receptor activation [112]. The capability of GPCR agonists to positively regulate glycosylated receptor signaling in the absence of their ligands presents a role of cannabinoid receptors CB1 and CB2 signaling in activating numerous receptor tyrosine kinases and Toll-like receptors in the induction of altered epigenetic landscape(s) in cancer cells which might transmogrify cancer metabolism and epigenetic reprogramming to a metastatic phenotype.

10. Future Research Directions and Limitations

The gaps in our knowledge regarding how CB cannabinoids are implicated in host metabolism via GPCR receptors reinforce the importance of research needed to understand the mechanistic action of these drugs on the body as promising metabolic candidates. The specific mechanisms and details of the effects of CB cannabinoids on host metabolism and energy homeostasis still remain to be elucidated. It is likely that future research in the area of cannabinoid pharmacology will be directed at:
  • Exploring the structure-activity relationships of ligands that target the CB1 allosteric site or that behave as neutral CB1 and/or CB2 receptor antagonists.
  • Assessing the therapeutic potential of CB1 and/or CB2 receptor allosteric modulators and neutral antagonists.
  • CB1 and/or CB2 receptor allosteric modulators and neutral antagonists modulate for or against the presence of endocannabinoid mechanisms in mammalian cells.
  • Validating and characterizing non-CB1 and non-CB2 targets such as RTK and TLR receptors for particular cannabinoids and developing compounds that can selectively activate or block such targets with reasonable potency for metabolic health and related diseases.
  • Validating and characterizing that cannabinoid receptors may exist as homodimers or form heterodimers or oligomers with one or more classes of the non-cannabinoid receptor.
  • Validating and characterizing the role played by the endocannabinoid system in ameliorating the symptoms and/or the underlying pathology of certain disorders.

Author Contributions

D.A.B., J.M. and M.R.S. analyzed the literature and wrote and read the manuscript. D.A.B. is the recipient of the 2018 Queen’s Entrance Award for Non-Ontario Students, the 2018 Queen’s Scholarship of Excellence, the 2020–2022 Dean’s Honour List, the 2022 Queen’s Graduate Award (QGA) and the 2022 FHS Graduate Fund (FHSGF). J.M. is the recipient of the 2019–2022 Dean’s Honors List with Distinction, the 2022 Nixon Academic Leadership Award, the 2019 Principal’s Entrance Scholarship, the 2019–2022 Academic All-Canadian Award and the 2019–2022 Athletic Financial Award. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants to M.R.S. from the Natural Sciences and Engineering Research Council of Canada (NSERC) Grant # RGPIN-2020-03869 and NSERC Alliance COVID-19 Grant # ALLRP-550110-20.

Acknowledgments

All authors acknowledge the educational and scholarly alliance of the Graduate Program in Experimental Medicine and the Health Sciences, HSCI BHSc Research program.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CB1: Cannabinoid receptors type 1, CB2: Cannabinoid receptors type 2, GPCR: G-Protein coupled receptor, ECS: Endocannabinoid system, THC: Delta-9 tetrahydrocannabinol, CBD: Cannabidiol, CBG: Cannabigerol, AEA: Anandamide, 2-AG: 2-arachidonic glycerol, G alpha subunit: Gi/o, MAPK: Mitogen activated protein kinase, PI3K: Phosphoinositol 3-kinase, COX: Cyclooxygenase, PAI-1: Plasminogen activator inhibitor-1, MMP: Matrix metalloproteinase-2, MMP-9: Matrix metalloproteinase-9, ECM: Extracellular matrix, EMT: Epithelial-mesenchymal transition, MAGL: Monoacylglycerol lipase, GPR55: G-protein coupled receptor 55, HER2: Human epidermal growth factor receptor, ERK: Extracellular signal-regulated kinase, NF-kB: Nuclear factor kappa B, ncRNA: Non-coding RNA, DLGAP2: Discs-Large Associated Protein 2, D2R: Dopamine receptor D2, 2meH3K9: Demethylated lysine 9, 3meH3K4: Trimethylated lysine-4, Igf1r: Insulin-like growth factor 1 receptor, miRNA: Micro RNA, HDAC: Histone deacetylase, Nuclear receptor corepressor (N-Cor), DNMT: DNA methyltransferases, MDSCs: Myeloid-derived suppressor cells, IkB: Inhibitors of kB, AML: Acute myeloid leukemia, JMJD3: Jumonji domain-containing protein-3, H3K27me3: Trimethylation marker of lysine 27 on histone 3, Epstein-Barr virus (EBV), RTK: Receptor tyrosine kinase, TNFR: Tumor necrosis factor, TLR: Toll-like receptors, NGF: Nerve growth factor, IR: Insulin receptor, EBP: Elastic binding protein, NMBR: neuromedin B GPCR, JAK/STAT: Janus kinase/signal transducers and activators of transcription, VEGF: Vascular endothelial growth factor, Oseltamivir Phosphate: OP, ASA: Aspirin, CSCs: Cancer stem like cells, PDA: Pancreatic ductal adenocarcinoma, Met: Metformin, GEM: Gemcitabine, IKKβ: Inhibitor kappa-B kinaseβ, KD: Ketogenic diet, NO: Nitric oxide, HAT: Histone acetyltransferase, Sulindac: SS, PG: Prostaglandin, NSAID: Non-steroidal anti-inflammatory drug, ERK: extracellular signal-regulated kinase, PCOS: Polycystic ovary syndrome, kE: κ-elastin, IGF: Insulin growth factor, IGF1R: IGF-1 receptor, mTOR: Mammalian target of rapamycin, mTORC1: mTOR complex 1, TKI: Tyrosine kinase inhibitor, BET: Bromo extra-terminal domain, IRS: Insulin substrate receptor, PTC: Papillary thyroid cancer, ADIRF-AS1: Adipogenesis regulatory factor-antisense RNA 1, OS: Osteosarcoma, T2DM: Type 2 diabetes, GPR40: G-protein coupled receptor-40.

References

  1. Kanherkar, R.R.; Bhatia-Dey, N.; Csoka, A.B. Epigenetics across the human lifespan. Front. Cell Dev. Biol. 2014, 2, 49. [Google Scholar] [CrossRef] [PubMed]
  2. Egger, G.; Liang, G.; Aparicio, A.; Jones, P.A. Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004, 429, 457–463. [Google Scholar] [CrossRef] [PubMed]
  3. Rock, E.M.; Parker, L.A. Constituents of Cannabis Sativa. Adv. Exp. Med. Biol. 2021, 1264, 1–13. [Google Scholar] [CrossRef] [PubMed]
  4. Filipiuc, L.E.; Ababei, D.C.; Alexa-Stratulat, T.; Pricope, C.V.; Bild, V.; Stefanescu, R.; Stanciu, G.D.; Tamba, B.-I. Major Phytocannabinoids and Their Related Compounds: Should We Only Search for Drugs That Act on Cannabinoid Receptors? Pharmaceutics 2021, 13, 1823. [Google Scholar] [CrossRef] [PubMed]
  5. Turner, S.; Williams, C.; Iversen, L.; Whalley, B. Molecular Pharmacology of Phytocannabinoids. Phytocannabinoids 2017, 103, 61–101. [Google Scholar]
  6. Wiley, J.L.; Owens, R.A.; Lichtman, A.H. Discriminative Stimulus Properties of Phytocannabinoids, Endocannabinoids, and Synthetic Cannabinoids. In The Behavioral Neuroscience of Drug Discrimination; Porter, J.H., Prus, A.J., Porter, J.H., Prus, A.J., Eds.; Springer: Cham, Switzerland, 2018; pp. 153–173. [Google Scholar]
  7. Stella, N.; Schweitzer, P.; Piomelli, D. A second endogenous cannabinoid that modulates long-term potentiation. Nature 1997, 388, 773–778. [Google Scholar] [CrossRef]
  8. Marzo, V.D.; Petrocellis, L.D. Plant, synthetic, and endogenous cannabinoids in medicine. Annu. Rev. Med. 2006, 57, 553–574. [Google Scholar] [CrossRef]
  9. Cohen, K.; Weinstein, A. The effects of cannabinoids on executive functions: Evidence from cannabis and synthetic cannabinoids—A systematic review. Brain Sci. 2018, 8, 40. [Google Scholar] [CrossRef]
  10. Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef]
  11. Pertwee, R.G. Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol. Ther. 1997, 74, 129–180. [Google Scholar] [CrossRef]
  12. Javid, F.A.; Phillips, R.M.; Afshinjavid, S.; Verde, R.; Ligresti, A. Cannabinoid pharmacology in cancer research: A new hope for cancer patients? Eur. J. Pharmacol. 2016, 775, 1–14. [Google Scholar] [CrossRef]
  13. Mielnik, C.A.; Lam, V.M.; Ross, R.A. CB1 allosteric modulators and their therapeutic potential in CNS disorders. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 106, 110163. [Google Scholar] [CrossRef]
  14. Mackie, K. Distribution of Cannabinoid Receptors in the Central and Peripheral Nervous System. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2005; Volume 168, pp. 299–325. [Google Scholar] [CrossRef]
  15. Onaivi, E.S.; Ishiguro, H.; Gong, J.-P.; Patel, S.; Perchuk, A.; Meozzi, P.A.; Myers, L.; Mora, Z.; Tagliaferro, P.; Gardner, E.; et al. Discovery of the Presence and Functional Expression of Cannabinoid CB2 Receptors in Brain. Ann. N. Y. Acad. Sci. 2006, 1074, 514–536. [Google Scholar] [CrossRef]
  16. Maccarrone, M.; Bab, I.; Bíró, T.; Cabral, G.A.; Dey, S.K.; Di Marzo, V.; Konje, J.C.; Kunos, G.; Mechoulam, R.; Pacher, P.; et al. Endocannabinoid signaling at the periphery: 50 years after THC. Trends Pharmacol. Sci. 2015, 36, 277–296. [Google Scholar] [CrossRef]
  17. Morales, P.; Reggio, P.H. An update on non-CB1, non-CB2 cannabinoid related G-protein-coupled receptors. Cannabis Cannabinoid Res. 2017, 2, 265–273. [Google Scholar] [CrossRef]
  18. Jett, J.; Stone, E.; Warren, G.; Cummings, K.M. Cannabis Use, Lung Cancer, and Related Issues. J. Thorac. Oncol. 2018, 13, 480–487. [Google Scholar] [CrossRef]
  19. Bogdanović, V.; Mrdjanović, J.; Borišev, I. A Review of the Therapeutic Antitumor Potential of Cannabinoids. J. Altern. Complement. Med. 2017, 23, 831–836. [Google Scholar] [CrossRef]
  20. Szutorisz, H.; Hurd, Y.L. High times for cannabis: Epigenetic imprint and its legacy on brain and behavior. Neurosci. Biobehav. Rev. 2018, 85, 93–101. [Google Scholar] [CrossRef]
  21. Montero-Oleas, N.; Arevalo-Rodriguez, I.; Nuñez-González, S.; Viteri-García, A.; Simancas-Racines, D. Therapeutic use of cannabis and cannabinoids: An evidence mapping and appraisal of systematic reviews. BMC Complement. Med. Ther. 2020, 20, 12. [Google Scholar] [CrossRef]
  22. Mouhamed, Y.; Vishnyakov, A.; Qorri, B.; Sambi, M.; Frank, S.S.; Nowierski, C.; Lamba, A.; Bhatti, U.; Szewczuk, M.R. Therapeutic potential of medicinal marijuana: An educational primer for health care professionals. Drug Healthc Patient Saf. 2018, 10, 45–66. [Google Scholar] [CrossRef]
  23. Peeri, H.; Koltai, H. Cannabis Biomolecule Effects on Cancer Cells and Cancer Stem Cells: Cytotoxic, Anti-Proliferative, and Anti-Migratory Activities. Biomolecules 2022, 12, 491. [Google Scholar] [CrossRef] [PubMed]
  24. Michalski, C.W.; Oti, F.E.; Erkan, M.; Sauliunaite, D.; Bergmann, F.; Pacher, P.; Batkai, S.; Müller, M.W.; Giese, N.A.; Friess, H.; et al. Cannabinoids in pancreatic cancer: Correlation with survival and pain. Int. J. Cancer 2008, 122, 742–750. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, Z.; Yang, J.; Zhao, H.; Fang, X.; Li, H. Cannabinoid receptor 2 is upregulated in melanoma. J. Cancer Res. Ther. 2012, 8, 549–554. [Google Scholar] [CrossRef] [PubMed]
  26. Chakravarti, B.; Ravi, J.; Ganju, R.K. Cannabinoids as therapeutic agents in cancer: Current status and future implications. Oncotarget 2014, 5, 5852–5872. [Google Scholar] [CrossRef]
  27. Pérez-Gómez, E.; Andradas, C.; Blasco-Benito, S.; Caffarel, M.M.; García-Taboada, E.; Villa-Morales, M.; Moreno, E.; Hamann, S.; Martín-Villar, E.; Flores, J.M.; et al. Role of cannabinoid receptor CB2 in HER2 pro-oncogenic signaling in breast cancer. J. Natl. Cancer Inst. 2015, 107, djv077. [Google Scholar] [CrossRef]
  28. Velasco, G.; Sánchez, C.; Guzmán, M. Anticancer mechanisms of cannabinoids. Curr. Oncol. 2016, 23, S23–S32. [Google Scholar] [CrossRef]
  29. Zhang, H.; Berezov, A.; Wang, Q.; Zhang, G.; Drebin, J.; Murali, R.; Greene, M.I. ErbB receptors: From oncogenes to targeted cancer therapies. J. Clin. Investig. 2007, 117, 2051–2058. [Google Scholar] [CrossRef]
  30. Ursini-Siegel, J.; Schade, B.; Cardiff, R.D.; Muller, W.J. Insights from transgenic mouse models of ERBB2-induced breast cancer. Nat. Rev. Cancer 2007, 7, 389–397. [Google Scholar] [CrossRef]
  31. Caffarel, M.M.; Andradas, C.; Mira, E.; Pérez-Gómez, E.; Cerutti, C.; Moreno-Bueno, G.; Flores, J.M.; García-Real, I.; Palacios, J.; Mañes, S.; et al. Cannabinoids reduce ErbB2-driven breast cancer progression through Akt inhibition. Mol. Cancer 2010, 9, 196. [Google Scholar] [CrossRef]
  32. Ramer, R.; Hinz, B. Inhibition of cancer cell invasion by cannabinoids via increased expression of tissue inhibitor of matrix metalloproteinases-1. J. Natl. Cancer Inst. 2008, 100, 59–69. [Google Scholar] [CrossRef]
  33. Massi, P.; Solinas, M.; Cinquina, V.; Parolaro, D. Cannabidiol as potential anticancer drug. Br. J. Clin. Pharmacol. 2013, 75, 303–312. [Google Scholar] [CrossRef] [PubMed]
  34. Vidinský, B.; Gál, P.; Pilátová, M.; Vidová, Z.; Solár, P.; Varinská, L.; Ivanová, L.; Mojžíš, J. Anti-proliferative and anti-angiogenic effects of CB2R agonist (JWH-133) in non-small lung cancer cells (A549) and human umbilical vein endothelial cells: An in vitro investigation. Folia Biol. 2012, 58, 75–80. [Google Scholar]
  35. Laezza, C.; D’Alessandro, A.; Paladino, S.; Maria Malfitano, A.; Chiara Proto, M.; Gazzerro, P.; Pisanti, S.; Santoro, A.; Ciaglia, E.; Bifulco, M.; et al. Anandamide inhibits the Wnt/β-catenin signalling pathway in human breast cancer MDA MB 231 cells. Eur. J. Cancer 2012, 48, 3112–3122. [Google Scholar] [CrossRef]
  36. Braile, M.; Marcella, S.; Marone, G.; Galdiero, M.R.; Varricchi, G.; Loffredo, S. The Interplay between the Immune and the Endocannabinoid Systems in Cancer. Cells 2021, 10, 1282. [Google Scholar] [CrossRef]
  37. Moreno, E.; Cavic, M.; Krivokuca, A.; Casadó, V.; Canela, E. The Endocannabinoid System as a Target in Cancer Diseases: Are We There Yet? Front. Pharmacol. 2019, 10, 339. [Google Scholar] [CrossRef]
  38. Zhu, L.X.; Sharma, S.; Stolina, M.; Gardner, B.; Roth, M.D.; Tashkin, D.P.; Dubinett, S.M. Δ-9-Tetrahydrocannabinol Inhibits Antitumor Immunity by a CB2 Receptor-Mediated, Cytokine-Dependent Pathway1. J. Immunol. 2000, 165, 373–380. [Google Scholar] [CrossRef]
  39. Hart, S.; Fischer, O.M.; Ullrich, A. Cannabinoids Induce Cancer Cell Proliferation via Tumor Necrosis Factor α-Converting Enzyme (TACE/ADAM17)-Mediated Transactivation of the Epidermal Growth Factor Receptor. Cancer Res. 2004, 64, 1943–1950. [Google Scholar] [CrossRef]
  40. McKallip, R.J.; Nagarkatti, M.; Nagarkatti, P.S. Δ-9-Tetrahydrocannabinol Enhances Breast Cancer Growth and Metastasis by Suppression of the Antitumor Immune Response1. J. Immunol. 2005, 174, 3281–3289. [Google Scholar] [CrossRef] [PubMed]
  41. Malfitano, A.M.; Ciaglia, E.; Gangemi, G.; Gazzerro, P.; Laezza, C.; Bifulco, M. Update on the endocannabinoid system as an anticancer target. Expert Opin. Ther. Targets 2011, 15, 297–308. [Google Scholar] [CrossRef]
  42. Sailler, S.; Schmitz, K.; Jäger, E.; Ferreiros, N.; Wicker, S.; Zschiebsch, K.; Pickert, G.; Geisslinger, G.; Walter, C.; Tegeder, I.; et al. Regulation of circulating endocannabinoids associated with cancer and metastases in mice and humans. Oncoscience 2014, 1, 272–282. [Google Scholar] [CrossRef] [PubMed]
  43. Munson, A.E.; Harris, L.S.; Friedman, M.A.; Dewey, W.L.; Carchman, R.A. Antineoplastic Activity of Cannabinoids2. JNCI J. Natl. Cancer Inst. 1975, 55, 597–602. [Google Scholar] [CrossRef]
  44. Demuth, D.G.; Molleman, A. Cannabinoid signalling. Life Sci. 2006, 78, 549–563. [Google Scholar] [CrossRef]
  45. Schwarz, R.; Ramer, R.; Hinz, B. Targeting the endocannabinoid system as a potential anticancer approach. Drug Metab. Rev. 2018, 50, 26–53. [Google Scholar] [CrossRef]
  46. Hinz, B.; Ramer, R. Anti-tumour actions of cannabinoids. Br. J. Pharmacol. 2019, 176, 1384–1394. [Google Scholar] [CrossRef]
  47. Kim, W.; Doyle, M.E.; Liu, Z.; Lao, Q.; Shin, Y.-K.; Carlson, O.D.; Kim, H.S.; Thomas, S.; Napora, J.K.; Lee, E.K.; et al. Cannabinoids Inhibit Insulin Receptor Signaling in Pancreatic β-Cells. Diabetes 2011, 60, 1198–1209. [Google Scholar] [CrossRef]
  48. Liu, J.; Zhou, L.; Xiong, K.; Godlewski, G.; Mukhopadhyay, B.; Tam, J.; Yin, S.; Gao, P.; Shan, X.; Pickel, J.; et al. Hepatic Cannabinoid Receptor-1 Mediates Diet-Induced Insulin Resistance via Inhibition of Insulin Signaling and Clearance in Mice. Gastroenterology 2012, 142, 1218–1228.e1211. [Google Scholar] [CrossRef]
  49. Elbaz, M.; Nasser, M.W.; Ravi, J.; Wani, N.A.; Ahirwar, D.K.; Zhao, H.; Oghumu, S.; Satoskar, A.R.; Shilo, K.; Carson, W.E.; et al. Modulation of the tumor microenvironment and inhibition of EGF/EGFR pathway: Novel anti-tumor mechanisms of Cannabidiol in breast cancer. Mol. Oncol. 2015, 9, 906–919. [Google Scholar] [CrossRef]
  50. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic modifications: Basic mechanisms and role in cardiovascular disease. Circulation 2011, 123, 2145–2156. [Google Scholar] [CrossRef] [PubMed]
  51. Smith, A.; Kaufman, F.; Sandy, M.S.; Cardenas, A. Cannabis Exposure During Critical Windows of Development: Epigenetic and Molecular Pathways Implicated in Neuropsychiatric Disease. Curr. Environ. Health Rpt. 2020, 7, 325–342. [Google Scholar] [CrossRef]
  52. Schrott, R.; Acharya, K.; Itchon-Ramos, N.; Hawkey, A.B.; Pippen, E.; Mitchell, J.T.; Kollins, S.H.; Levin, E.D.; Murphy, S.K. Cannabis use is associated with potentially heritable widespread changes in autism candidate gene DLGAP2 DNA methylation in sperm. Epigenetics 2020, 15, 161–173. [Google Scholar] [CrossRef]
  53. Szutorisz, H.; DiNieri, J.A.; Sweet, E.; Egervari, G.; Michaelides, M.; Carter, J.M.; Ren, Y.; Miller, M.L.; Blitzer, R.D.; Hurd, Y.L. Parental THC Exposure Leads to Compulsive Heroin-Seeking and Altered Striatal Synaptic Plasticity in the Subsequent Generation. Neuropsychopharmacology 2014, 39, 1315–1323. [Google Scholar] [CrossRef] [PubMed]
  54. Watson, C.T.; Szutorisz, H.; Garg, P.; Martin, Q.; Landry, J.A.; Sharp, A.J.; Hurd, Y.L. Genome-Wide DNA Methylation Profiling Reveals Epigenetic Changes in the Rat Nucleus Accumbens Associated With Cross-Generational Effects of Adolescent THC Exposure. Neuropsychopharmacology 2015, 40, 2993–3005. [Google Scholar] [CrossRef] [PubMed]
  55. Volkow, N.D.; Fowler, J.S.; Wang, G.J.; Swanson, J.M. Dopamine in drug abuse and addiction: Results from imaging studies and treatment implications. Mol. Psychiatry 2004, 9, 557–569. [Google Scholar] [CrossRef] [PubMed]
  56. DiNieri, J.A.; Wang, X.; Szutorisz, H.; Spano, S.M.; Kaur, J.; Casaccia, P.; Dow-Edwards, D.; Hurd, Y.L. Maternal Cannabis Use Alters Ventral Striatal Dopamine D2 Gene Regulation in the Offspring. Biol. Psychiatry 2011, 70, 763–769. [Google Scholar] [CrossRef]
  57. Martínez-Peña, A.A.; Lee, K.; Pereira, M.; Ayyash, A.; Petrik, J.J.; Hardy, D.B.; Holloway, A.C. Prenatal Exposure to Delta-9-tetrahydrocannabinol (THC) Alters the Expression of miR-122-5p and Its Target Igf1r in the Adult Rat Ovary. Int. J. Mol. Sci. 2022, 23, 8000. [Google Scholar] [CrossRef]
  58. D’Addario, C.; Di Francesco, A.; Pucci, M.; Finazzi Agrò, A.; Maccarrone, M. Epigenetic mechanisms and endocannabinoid signalling. FEBS J. 2013, 280, 1905–1917. [Google Scholar] [CrossRef]
  59. Basavarajappa, B.S.; Subbanna, S. Molecular Insights into Epigenetics and Cannabinoid Receptors. Biomolecules 2022, 12, 1560. [Google Scholar] [CrossRef]
  60. Khare, M.; Taylor, A.H.; Konje, J.C.; Bell, S.C. Δ9-Tetrahydrocannabinol inhibits cytotrophoblast cell proliferation and modulates gene transcription. Mol. Hum. Reprod. 2006, 12, 321–333. [Google Scholar] [CrossRef]
  61. Guenther, M.G.; Lazar, M.A. Biochemical isolation and analysis of a nuclear receptor corepressor complex. Methods Enzymol. 2003, 364, 246–257. [Google Scholar]
  62. Pucci, M.; Rapino, C.; Di Francesco, A.; Dainese, E.; D’Addario, C.; Maccarrone, M. Epigenetic control of skin differentiation genes by phytocannabinoids. Br. J. Pharmacol. 2013, 170, 581–591. [Google Scholar] [CrossRef]
  63. Sido, J.M.; Yang, X.; Nagarkatti, P.S.; Nagarkatti, M. Δ9-Tetrahydrocannabinol-mediated epigenetic modifications elicit myeloid-derived suppressor cell activation via STAT3/S100A8. J. Leukoc. Biol. 2015, 97, 677–688. [Google Scholar] [CrossRef]
  64. Mondal, N.K.; Behera, J.; Kelly, K.E.; George, A.K.; Tyagi, P.K.; Tyagi, N. Tetrahydrocurcumin epigenetically mitigates mitochondrial dysfunction in brain vasculature during ischemic stroke. Neurochem. Int. 2019, 122, 120–138. [Google Scholar] [CrossRef]
  65. Sales, A.J.; Guimarães, F.S.; Joca, S.R.L. CBD modulates DNA methylation in the prefrontal cortex and hippocampus of mice exposed to forced swim. Behav. Brain Res. 2020, 388, 112627. [Google Scholar] [CrossRef]
  66. Fuchs Weizman, N.; Wyse, B.A.; Szaraz, P.; Defer, M.; Jahangiri, S.; Librach, C.L. Cannabis alters epigenetic integrity and endocannabinoid signalling in the human follicular niche. Hum. Reprod. 2021, 36, 1922–1931. [Google Scholar] [CrossRef]
  67. Paradisi, A.; Pasquariello, N.; Barcaroli, D.; Maccarrone, M. Anandamide Regulates Keratinocyte Differentiation by Inducing DNA Methylation in a CB1 Receptor-dependent Manner. J. Biol. Chem. 2008, 283, 6005–6012. [Google Scholar] [CrossRef]
  68. Hwang, Y.S.; Kim, Y.-J.; Kim, M.O.; Kang, M.; Oh, S.W.; Nho, Y.H.; Park, S.-H.; Lee, J. Cannabidiol upregulates melanogenesis through CB1 dependent pathway by activating p38 MAPK and p42/44 MAPK. Chem. Biol. Interact. 2017, 273, 107–114. [Google Scholar] [CrossRef]
  69. Hoesel, B.; Schmid, J.A. The complexity of NF-κB signaling in inflammation and cancer. Mol. Cancer 2013, 12, 86. [Google Scholar] [CrossRef]
  70. Zhang, L.; Xiao, X.; Arnold, P.R.; Li, X.C. Transcriptional and epigenetic regulation of immune tolerance: Roles of the NF-κB family members. Cell Mol. Immunol. 2019, 16, 315–323. [Google Scholar] [CrossRef]
  71. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  72. Verma, I.M. Nuclear factor (NF)-B proteins: Therapeutic targets. Ann. Rheum. Dis. 2004, 63, ii57–ii61. [Google Scholar] [CrossRef]
  73. Tagde, A.; Rajabi, H.; Stroopinsky, D.; Gali, R.; Alam, M.; Bouillez, A.; Kharbanda, S.; Stone, R.; Avigan, D.; Kufe, D. MUC1-C induces DNA methyltransferase 1 and represses tumor suppressor genes in acute myeloid leukemia. Oncotarget 2016, 7, 38974–38987. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, S.; Liu, Z.; Xie, Z.; Pang, J.; Yu, J.; Lehmann, E.; Huynh, L.; Vukosavljevic, T.; Takeki, M.; Klisovic, R.B.; et al. Bortezomib induces DNA hypomethylation and silenced gene transcription by interfering with Sp1/NF-κB–dependent DNA methyltransferase activity in acute myeloid leukemia. Blood 2008, 111, 2364–2373. [Google Scholar] [CrossRef] [PubMed]
  75. De Santa, F.; Totaro, M.G.; Prosperini, E.; Notarbartolo, S.; Testa, G.; Natoli, G. The Histone H3 Lysine-27 Demethylase Jmjd3 Links Inflammation to Inhibition of Polycomb-Mediated Gene Silencing. Cell 2007, 130, 1083–1094. [Google Scholar] [CrossRef]
  76. Federman, N.; de la Fuente, V.; Zalcman, G.; Corbi, N.; Onori, A.; Passananti, C.; Romano, A. Nuclear Factor B-Dependent Histone Acetylation is Specifically Involved in Persistent Forms of Memory. J. Neurosci. 2013, 33, 7603–7614. [Google Scholar] [CrossRef] [PubMed]
  77. Teferedegne, B.; Green, M.R.; Guo, Z.; Boss, J.M. Mechanism of Action of a Distal NF-κB-Dependent Enhancer. Mol. Cell Biol. 2006, 26, 5759–5770. [Google Scholar] [CrossRef]
  78. Hayden, M.S.; Ghosh, S. Shared Principles in NF-κB Signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [PubMed]
  79. Vento-Tormo, R.; Rodríguez-Ubreva, J.; Lisio, L.D.; Islam, A.B.; Urquiza, J.M.; Hernando, H.; Lopez-Bigas, N.; Shannon-Lowe, C.; Martínez, N.; Montes-Moreno, S. NF-κB directly mediates epigenetic deregulation of common microRNAs in Epstein-Barr virus-mediated transformation of B-cells and in lymphomas. Nucleic Acids Res. 2014, 42, 11025–11039. [Google Scholar] [CrossRef]
  80. Kozela, E.; Pietr, M.; Juknat, A.; Rimmerman, N.; Levy, R.; Vogel, Z. Cannabinoids Δ9-Tetrahydrocannabinol and Cannabidiol Differentially Inhibit the Lipopolysaccharide-activated NF-κB and Interferon-β/STAT Proinflammatory Pathways in BV-2 Microglial Cells*. J. Biol. Chem. 2010, 285, 1616–1626. [Google Scholar] [CrossRef]
  81. Hassan, S. Effect of Cannabinoid Receptor Ligands on Microglial Cell Functions. Ph.D. Thesis, The University of Nottingham, Nottingham, UK, 2013. [Google Scholar]
  82. Huang, Y.; Wan, T.; Pang, N.; Zhou, Y.; Jiang, X.; Li, B.; Gu, Y.; Huang, Y.; Ye, X.; Lian, H.; et al. Cannabidiol protects livers against nonalcoholic steatohepatitis induced by high-fat high cholesterol diet via regulating NF-κB and NLRP3 inflammasome pathway. J. Cell Physiol. 2019, 234, 21224–21234. [Google Scholar] [CrossRef]
  83. González-Mariscal, I.; Carmona-Hidalgo, B.; Winkler, M.; Unciti-Broceta, J.D.; Escamilla, A.; Gómez-Cañas, M.; Fernández-Ruiz, J.; Fiebich, B.L.; Romero-Zerbo, S.-Y.; Bermúdez-Silva, F.J.; et al. (+)-trans-Cannabidiol-2-hydroxy pentyl is a dual CB1R antagonist/CB2R agonist that prevents diabetic nephropathy in mice. Pharmacol. Res. 2021, 169, 105492. [Google Scholar] [CrossRef]
  84. Krzyżewska, A.; Baranowska-Kuczko, M.; Jastrząb, A.; Kasacka, I.; Koz\lowska, H. Cannabidiol Improves Antioxidant Capacity and Reduces Inflammation in the Lungs of Rats with Monocrotaline-Induced Pulmonary Hypertension. Molecules 2022, 27, 3327. [Google Scholar] [CrossRef]
  85. Do, Y.; McKallip, R.J.; Nagarkatti, M.; Nagarkatti, P.S. Activation through cannabinoid receptors 1 and 2 on dendritic cells triggers NF-κB-dependent apoptosis: Novel role for endogenous and exogenous cannabinoids in immunoregulation. J. Immunol. 2004, 173, 2373–2382. [Google Scholar] [CrossRef]
  86. Chen, R.; Zhang, J.; Fan, N.; Teng, Z.-q.; Wu, Y.; Yang, H.; Tang, Y.-p.; Sun, H.; Song, Y.; Chen, C. Δ9-THC-Caused Synaptic and Memory Impairments Are Mediated through COX-2 Signaling. Cell 2013, 155, 1154–1165. [Google Scholar] [CrossRef]
  87. Jouroukhin, Y.; Zhu, X.; Shevelkin, A.V.; Hasegawa, Y.; Abazyan, B.; Saito, A.; Pevsner, J.; Kamiya, A.; Pletnikov, M.V. Adolescent Δ9-THC exposure and astrocyte-specific genetic vulnerability converge on NF-κB-COX-2 signaling to impair memory in adulthood. Biol. Psychiatry 2019, 85, 891–903. [Google Scholar] [CrossRef]
  88. Wei, T.-T.; Chandy, M.; Nishiga, M.; Zhang, A.; Kumar, K.K.; Thomas, D.; Manhas, A.; Rhee, S.; Justesen, J.M.; Chen, I.Y.; et al. Cannabinoid receptor 1 antagonist genistein attenuates marijuana-induced vascular inflammation. Cell 2022, 185, 1676–1693.e1623. [Google Scholar] [CrossRef]
  89. Wootten, D.; Christopoulos, A.; Marti-Solano, M.; Babu, M.M.; Sexton, P.M. Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat. Rev. Mol. Cell Biol. 2018, 19, 638–653. [Google Scholar] [CrossRef]
  90. Baltoumas, F.A.; Theodoropoulou, M.C.; Hamodrakas, S.J. Interactions of the α-subunits of heterotrimeric G-proteins with GPCRs, effectors and RGS proteins: A critical review and analysis of interacting surfaces, conformational shifts, structural diversity and electrostatic potentials. J. Struct. Biol. 2013, 182, 209–218. [Google Scholar] [CrossRef]
  91. Khajehali, E.; Malone, D.T.; Glass, M.; Sexton, P.M.; Christopoulos, A.; Leach, K. Biased agonism and biased allosteric modulation at the CB1 cannabinoid receptor. Mol. Pharmacol. 2015, 88, 368–379. [Google Scholar] [CrossRef]
  92. Olianas, M.C.; Dedoni, S.; Onali, P. Cannabinoid CB1 and CB2 receptors differentially regulate TNF-α-induced apoptosis and LPA1-mediated pro-survival signaling in HT22 hippocampal cells. Life Sci. 2021, 276, 119407. [Google Scholar] [CrossRef]
  93. Yang, H.; Wang, Z.; Capó-Aponte, J.E.; Zhang, F.; Pan, Z.; Reinach, P.S. Epidermal growth factor receptor transactivation by the cannabinoid receptor (CB1) and transient receptor potential vanilloid 1 (TRPV1) induces differential responses in corneal epithelial cells. Exp. Eye Res. 2010, 91, 462–471. [Google Scholar] [CrossRef]
  94. Fitzpatrick, J.-M.; Hackett, B.; Costelloe, L.; Hind, W.; Downer, E.J. Botanically-Derived Δ9-Tetrahydrocannabinol and Cannabidiol, and Their 1: 1 Combination, Modulate Toll-like Receptor 3 and 4 Signalling in Immune Cells from People with Multiple Sclerosis. Molecules 2022, 27, 1763. [Google Scholar] [CrossRef] [PubMed]
  95. Keresztes, A.; Streicher, J.M. Synergistic interaction of the cannabinoid and death receptor systems—A potential target for future cancer therapies? FEBS Lett. 2017, 591, 3235–3251. [Google Scholar] [CrossRef] [PubMed]
  96. Laprairie, R.B.; Bagher, A.M.; Kelly, M.E.M.; Denovan-Wright, E.M. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br. J. Pharmacol. 2015, 172, 4790–4805. [Google Scholar] [CrossRef] [PubMed]
  97. Sabatucci, A.; Tortolani, D.; Dainese, E.; Maccarrone, M. In silico mapping of allosteric ligand binding sites in type-1 cannabinoid receptor. Biotechnol. Appl. Biochem. 2018, 65, 21–28. [Google Scholar] [CrossRef]
  98. Qorri, B.; Kalaydina, R.V.; Velickovic, A.; Kaplya, Y.; Decarlo, A.; Szewczuk, M.R. Agonist-Biased Signaling via Matrix Metalloproteinase-9 Promotes Extracellular Matrix Remodeling. Cells 2018, 7, 117. [Google Scholar] [CrossRef]
  99. Jakowiecki, J.; Abel, R.; Orzeł, U.; Pasznik, P.; Preissner, R.; Filipek, S. Allosteric Modulation of the CB1 Cannabinoid Receptor by Cannabidiol—A Molecular Modeling Study of the N-Terminal Domain and the Allosteric-Orthosteric Coupling. Molecules 2021, 26, 2456. [Google Scholar] [CrossRef]
  100. Reber, L.; Vermeulen, L.; Haegeman, G.; Frossard, N. Ser276 phosphorylation of NF-κBp65 by MSK1 controls SCF expression in inflammation. PLoS ONE 2009, 4, e4393. [Google Scholar] [CrossRef]
  101. Verstrepen, L.; Bekaert, T.; Chau, T.L.; Tavernier, J.; Chariot, A.; Beyaert, R. TLR-4, IL-1R and TNF-R signaling to NF-κB: Variations on a common theme. Cell. Mol. Life Sci. 2008, 65, 2964–2978. [Google Scholar] [CrossRef]
  102. Zhang, H.; Sun, S.-C. NF-κB in inflammation and renal diseases. Cell Biosci. 2015, 5, 63. [Google Scholar] [CrossRef]
  103. Hua, F.; Hao, W.; Wang, L.; Li, S. Linear Ubiquitination Mediates EGFR-Induced NF-κB Pathway and Tumor Development. Int. J. Mol. Sci. 2021, 22, 11875. [Google Scholar] [CrossRef]
  104. Asimaki, O.; Sakellaridis, N.; Mangoura, D. Transactivation of epidermal growth factor receptor EGFR by CB1 cannabinoid receptor agonists. Ep. Klin. Farmakol. Kai Farmakokinet. 2007, 25, 19–21. [Google Scholar]
  105. Haxho, F.; Haq, S.; Szewczuk, M.R. Biased G protein-coupled receptor agonism mediates Neu1 sialidase and matrix metalloproteinase-9 crosstalk to induce transactivation of insulin receptor signaling. Cell. Signal. 2018, 43, 71–84. [Google Scholar] [CrossRef]
  106. Jayanth, P.; Amith, S.R.; Gee, K.; Szewczuk, M.R. Neu1 sialidase and matrix metalloproteinase-9 cross-talk is essential for neurotrophin activation of Trk receptors and cellular signaling. Cell. Signal. 2010, 22, 1193–1205. [Google Scholar] [CrossRef]
  107. Abdulkhalek, S.; Guo, M.; Amith, S.R.; Jayanth, P.; Szewczuk, M.R. G-protein coupled receptor agonists mediate Neu1 sialidase and matrix metalloproteinase-9 cross-talk to induce transactivation of TOLL-like receptors and cellular signaling. Cell. Signal. 2012, 24, 2035–2042. [Google Scholar] [CrossRef]
  108. Erica, Z.; Cristina, M.; Marina, G.; Tiziana, R.; Daniela, P. Chapter 6—Correlates and consequences of cannabinoid exposure on adolescent brain remodeling: Focus on glial cells and epigenetics. In Cannabis and the Developing Brain; Melis, M., Manzoni, O.J.J., Melis, M., Manzoni, O.J.J., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 95–106. [Google Scholar]
  109. Costa, B.; Siniscalco, D.; Trovato, A.E.; Comelli, F.; Sotgiu, M.L.; Colleoni, M.; Maione, S.; Rossi, F.; Giagnoni, G. AM404, an inhibitor of anandamide uptake, prevents pain behaviour and modulates cytokine and apoptotic pathways in a rat model of neuropathic pain. Br. J. Pharmacol. 2006, 148, 1022–1032. [Google Scholar] [CrossRef]
  110. Paszcuk, A.F.; Dutra, R.C.; da Silva, K.A.B.S.; Quintão, N.L.M.; Campos, M.M.; Calixto, J.B. Cannabinoid Agonists Inhibit Neuropathic Pain Induced by Brachial Plexus Avulsion in Mice by Affecting Glial Cells and MAP Kinases. PLoS ONE 2011, 6, e24034. [Google Scholar] [CrossRef]
  111. Rozenfeld, R.; Gupta, A.; Gagnidze, K.; Lim, M.P.; Gomes, I.; Lee-Ramos, D.; Nieto, N.; Devi, L.A. AT1R-CB₁R heteromerization reveals a new mechanism for the pathogenic properties of angiotensin II. EMBO J. 2011, 30, 2350–2363. [Google Scholar] [CrossRef]
  112. Alghamdi, F.; Guo, M.; Abdulkhalek, S.; Crawford, N.; Amith, S.R.; Szewczuk, M.R. A novel insulin receptor-signaling platform and its link to insulin resistance and type 2 diabetes. Cell. Signal. 2014, 26, 1355–1368. [Google Scholar] [CrossRef]
  113. Bordoni, L.; Gabbianelli, R. Primers on nutrigenetics and nutri(epi)genomics: Origins and development of precision nutrition. Biochimie 2019, 160, 156–171. [Google Scholar] [CrossRef]
  114. Hinek, A.; Bodnaruk, T.D.; Bunda, S.; Wang, Y.; Liu, K. Neuraminidase-1, a subunit of the cell surface elastin receptor, desialylates and functionally inactivates adjacent receptors interacting with the mitogenic growth factors PDGF-BB and IGF-2. Am. J. Pathol. 2008, 173, 1042–1056. [Google Scholar] [CrossRef]
  115. Abdulkhalek, S.; Amith, S.R.; Franchuk, S.L.; Jayanth, P.; Guo, M.; Finlay, T.; Gilmour, A.; Guzzo, C.; Gee, K.; Beyaert, R. Neu1 sialidase and matrix metalloproteinase-9 cross-talk is essential for Toll-like receptor activation and cellular signaling. J. Biol. Chem. 2011, 286, 36532–36549. [Google Scholar] [CrossRef] [PubMed]
  116. Gilmour, A.M.; Abdulkhalek, S.; Cheng, T.S.W.; Alghamdi, F.; Jayanth, P.; O’Shea, L.K.; Geen, O.; Arvizu, L.A.; Szewczuk, M.R. A novel epidermal growth factor receptor-signaling platform and its targeted translation in pancreatic cancer. Cell. Signal. 2013, 25, 2587–2603. [Google Scholar] [CrossRef] [PubMed]
  117. Liang, F.; Seyrantepe, V.; Landry, K.; Ahmad, R.; Ahmad, A.; Stamatos, N.M.; Pshezhetsky, A.V. Monocyte differentiation up-regulates the expression of the lysosomal sialidase, Neu1, and triggers its targeting to the plasma membrane via major histocompatibility complex class II-positive compartments*. J. Biol. Chem. 2006, 281, 27526–27538. [Google Scholar] [CrossRef] [PubMed]
  118. Abdulkhalek, S.; Hrynyk, M.; Szewczuk, M.R. A novel G-protein-coupled receptor-signaling platform and its targeted translation in human disease. Res. Rep. Biochem. 2013, 3, 17–30. [Google Scholar]
  119. Vajaria, B.N.; Patel, P.S. Glycosylation: A hallmark of cancer? Glycoconj. J. 2017, 34, 147–156. [Google Scholar] [CrossRef]
  120. Sparrow, L.G.; Lawrence, M.C.; Gorman, J.J.; Strike, P.M.; Robinson, C.P.; McKern, N.M.; Ward, C.W. N-linked glycans of the human insulin receptor and their distribution over the crystal structure. Proteins Struct. Funct. Bioinform. 2008, 71, 426–439. [Google Scholar] [CrossRef]
  121. DeFronzo, R.A.; Ferrannini, E. Insulin resistance: A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care 1991, 14, 173–194. [Google Scholar] [CrossRef]
  122. Kahn, S.E. The importance of the β-cell in the pathogenesis of type 2 diabetes mellitus. Am. J. Med. 2000, 108, 2–8. [Google Scholar] [CrossRef]
  123. Hirabara, S.M.; Gorjão, R.; Vinolo, M.A.; Rodrigues, A.C.; Nachbar, R.T.; Curi, R. Molecular targets related to inflammation and insulin resistance and potential interventions. J. Biomed. Biotechnol. 2012, 2012, 379024. [Google Scholar] [CrossRef]
  124. Arabkhari, M.; Bunda, S.; Wang, Y.; Wang, A.; Pshezhetsky, A.V.; Hinek, A. Desialylation of insulin receptors and IGF-1 receptors by neuraminidase-1 controls the net proliferative response of L6 myoblasts to insulin. Glycobiology 2010, 20, 603–616. [Google Scholar] [CrossRef]
  125. Fischoeder, A.; Meyborg, H.; Stibenz, D.; Fleck, E.; Graf, K.; Stawowy, P. Insulin augments matrix metalloproteinase-9 expression in monocytes. Cardiovasc. Res. 2007, 73, 841–848. [Google Scholar] [CrossRef]
  126. Oda, K.; Matsuoka, Y.; Funahashi, A.; Kitano, H. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol. Syst. Biol. 2005, 1, 2005-0010. [Google Scholar] [CrossRef]
  127. Normanno, N.; De Luca, A.; Bianco, C.; Strizzi, L.; Mancino, M.; Maiello, M.R.; Carotenuto, A.; De Feo, G.; Caponigro, F.; Salomon, D.S. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene 2006, 366, 2–16. [Google Scholar] [CrossRef]
  128. Yarden, Y.; Pines, G. The ERBB network: At last, cancer therapy meets systems biology. Nat. Rev. Cancer 2012, 12, 553–563. [Google Scholar] [CrossRef]
  129. Bonten, E.J.; Annunziata, I.; d’Azzo, A. Lysosomal multienzyme complex: Pros and cons of working together. Cell. Mol. Life Sci. 2014, 71, 2017–2032. [Google Scholar] [CrossRef]
  130. Ono, M.; Kuwano, M. Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs. Clin. Cancer Res. 2006, 12, 7242–7251. [Google Scholar] [CrossRef]
  131. Gazdar, A.F. Personalized medicine and inhibition of EGFR signaling in lung cancer. N. Engl. J. Med. 2009, 361, 1018. [Google Scholar] [CrossRef]
  132. Haxho, F.; Neufeld, R.J.; Szewczuk, M.R. Neuraminidase-1: A novel therapeutic target in multistage tumorigenesis. Oncotarget 2016, 7, 40860–40881. [Google Scholar] [CrossRef]
  133. O’Shea, L.K.; Abdulkhalek, S.; Allison, S.; Neufeld, R.J.; Szewczuk, M.R. Therapeutic targeting of Neu1 sialidase with oseltamivir phosphate (Tamiflu®) disables cancer cell survival in human pancreatic cancer with acquired chemoresistance. Onco Targets Ther. 2014, 7, 117–134. [Google Scholar] [CrossRef]
  134. Abdulkhalek, S.; Geen, O.D.; Brodhagen, L.; Haxho, F.; Alghamdi, F.; Allison, S.; Simmons, D.J.; O’Shea, L.K.; Neufeld, R.J.; Szewczuk, M.R. Transcriptional factor snail controls tumor neovascularization, growth and metastasis in mouse model of human ovarian carcinoma. Clin. Transl. Med. 2014, 3, 28. [Google Scholar] [CrossRef]
  135. Wong, H.H.; Lemoine, N.R. Pancreatic cancer: Molecular pathogenesis and new therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 412–422. [Google Scholar] [CrossRef] [PubMed]
  136. Sarmento-Ribeiro, A.B.; Scorilas, A.; Gonçalves, A.C.; Efferth, T.; Trougakos, I.P. The emergence of drug resistance to targeted cancer therapies: Clinical evidence. Drug Resist. Updates 2019, 47, 100646. [Google Scholar] [CrossRef] [PubMed]
  137. Sambi, M.; Szewczuk, M.R. Introduction to the Acquisition of Resistance to Targeted Therapy. In Current Applications for Overcoming Resistance to Targeted Therapies; Springer Nature Switzerland AG: Cham, Switzerland, 2019; pp. 1–33. [Google Scholar]
  138. Al-Zeheimi, N.; Adham, S.A. Therapies to Overcome Multidrug-Resistant Receptors. In Current Applications for Overcoming Resistance to Targeted Therapies; Springer Nature Switzerland AG: Cham, Switzerland, 2019; pp. 131–159. [Google Scholar]
  139. Long, S.S.; Prober, C.G.; Fischer, M. Principles and Practice of Pediatric Infectious Diseases E-Book; Elsevier Health Sciences: Philadelphia, PA, USA, 2022. [Google Scholar]
  140. Haxho, F.; Allison, S.; Alghamdi, F.; Brodhagen, L.; Kuta, V.E.L.; Abdulkhalek, S.; Neufeld, R.J.; Szewczuk, M.R. Oseltamivir phosphate monotherapy ablates tumor neovascularization, growth, and metastasis in mouse model of human triple-negative breast adenocarcinoma. Breast Cancer Targets Ther. 2014, 6, 191. [Google Scholar]
  141. Amith, S.R.; Jayanth, P.; Franchuk, S.; Siddiqui, S.; Seyrantepe, V.; Gee, K.; Basta, S.; Beyaert, R.; Pshezhetsky, A.V.; Szewczuk, M.R. Dependence of pathogen molecule-induced Toll-like receptor activation and cell function on Neu1 sialidase. Glycoconj. J. 2009, 26, 1197. [Google Scholar] [CrossRef] [PubMed]
  142. Abdulkhalek, S.; Szewczuk, M.R. Neu1 sialidase and matrix metalloproteinase-9 cross-talk regulates nucleic acid-induced endosomal TOLL-like receptor-7 and-9 activation, cellular signaling and pro-inflammatory responses. Cell. Signal. 2013, 25, 2093–2105. [Google Scholar] [CrossRef]
  143. Zhang, Y.; Liu, L.; Fan, P.; Bauer, N.; Gladkich, J.; Ryschich, E.; Bazhin, A.V.; Giese, N.A.; Strobel, O.; Hackert, T.; et al. Aspirin counteracts cancer stem cell features, desmoplasia and gemcitabine resistance in pancreatic cancer. Oncotarget 2015, 6, 9999–10015. [Google Scholar] [CrossRef]
  144. Kopp, E.; Ghosh, S. Inhibition of NF-kappa B by sodium salicylate and aspirin. Science 1994, 265, 956–959. [Google Scholar] [CrossRef]
  145. Yin, M.-J.; Yamamoto, Y.; Gaynor, R.B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of IκB kinase-β. Nature 1998, 396, 77–80. [Google Scholar] [CrossRef]
  146. Qorri, B.; Harless, W.; Szewczuk, M.R. Novel Molecular Mechanism of Aspirin and Celecoxib Targeting Mammalian Neuraminidase-1 Impedes Epidermal Growth Factor Receptor Signaling Axis and Induces Apoptosis in Pancreatic Cancer Cells. Drug Des. Devel. Ther. 2020, 14, 4149–4167. [Google Scholar] [CrossRef]
  147. Sambi, M.; Samuel, V.; Qorri, B.; Haq, S.; Burov, S.V.; Markvicheva, E.; Harless, W.; Szewczuk, M.R. A Triple Combination of Metformin, Acetylsalicylic Acid, and Oseltamivir Phosphate Impacts Tumour Spheroid Viability and Upends Chemoresistance in Triple-Negative Breast Cancer. Drug Des. Devel. Ther. 2020, 14, 1995–2019. [Google Scholar] [CrossRef]
  148. Qorri, B.; Mokhtari, R.B.; Harless, W.W.; Szewczuk, M.R. Next Generation of Cancer Drug Repurposing: Therapeutic Combination of Aspirin and Oseltamivir Phosphate Potentiates Gemcitabine to Disable Key Survival Pathways Critical for Pancreatic Cancer Progression. Cancers 2022, 14, 1374. [Google Scholar] [CrossRef]
  149. Qorri, B.; Mokhtari, R.B.; Harless, W.W.; Szewczuk, M.R. Repositioning of Old Drugs for Novel Cancer Therapies: Continuous Therapeutic Perfusion of Aspirin and Oseltamivir Phosphate with Gemcitabine Treatment Disables Tumor Progression, Chemoresistance, and Metastases. Cancers 2022, 14, 3595. [Google Scholar] [CrossRef]
  150. Blanco, M.J.; Moreno-Bueno, G.; Sarrio, D.; Locascio, A.; Cano, A.; Palacios, J.; Nieto, M.A. Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene 2002, 21, 3241–3246. [Google Scholar] [CrossRef]
  151. Jordà, M.; Olmeda, D.; Vinyals, A.; Valero, E.; Cubillo, E.; Llorens, A.; Cano, A.; Fabra, A. Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J. Cell Sci. 2005, 118, 3371–3385. [Google Scholar] [CrossRef]
  152. Greten, F.R.; Eckmann, L.; Greten, T.F.; Park, J.M.; Li, Z.-W.; Egan, L.J.; Kagnoff, M.F.; Karin, M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004, 118, 285–296. [Google Scholar] [CrossRef]
  153. Rosenberg, G.A. Matrix metalloproteinases in neuroinflammation. Glia 2002, 39, 279–291. [Google Scholar] [CrossRef]
  154. Wang, H.-H.; Hsieh, H.-L.; Wu, C.-Y.; Sun, C.-C.; Yang, C.-M. Oxidized low-density lipoprotein induces matrix metalloproteinase-9 expression via a p42/p44 and JNK-dependent AP-1 pathway in brain astrocytes. Glia 2009, 57, 24–38. [Google Scholar] [CrossRef]
  155. Speidl, W.S.; Kastl, S.P.; Hutter, R.; Katsaros, K.M.; Kaun, C.; Bauriedel, G.; Maurer, G.; Huber, K.; Badimon, J.J.; Wojta, J. The complement component C5a is present in human coronary lesions in vivo and induces the expression of MMP-1 and MMP-9 in human macrophages in vitro. FASEB J. 2011, 25, 35–44. [Google Scholar] [CrossRef]
  156. Hwang, Y.P.; Yun, H.J.; Choi, J.H.; Han, E.H.; Kim, H.G.; Song, G.Y.; Kwon, K.-I.; Jeong, T.C.; Jeong, H.G. Suppression of EGF-induced tumor cell migration and matrix metalloproteinase-9 expression by capsaicin via the inhibition of EGFR-mediated FAK/Akt, PKC/Raf/ERK, p38 MAPK, and AP-1 signaling. Mol. Nutr. Food Res. 2011, 55, 594–605. [Google Scholar] [CrossRef]
  157. Park, J.; Kwak, C.-H.; Ha, S.-H.; Kwon, K.-M.; Abekura, F.; Cho, S.-H.; Chang, Y.-C.; Lee, Y.-C.; Ha, K.-T.; Chung, T.-W.; et al. Ganglioside GM3 suppresses lipopolysaccharide-induced inflammatory responses in rAW 264.7 macrophage cells through NF-κB, AP-1, and MAPKs signaling. J. Cell Biochem. 2018, 119, 1173–1182. [Google Scholar] [CrossRef]
  158. Ryan, A.E.; Colleran, A.; O’Gorman, A.; O’Flynn, L.; Pindjacova, J.; Lohan, P.; O’Malley, G.; Nosov, M.; Mureau, C.; Egan, L.J. Targeting colon cancer cell NF-κB promotes an anti-tumour M1-like macrophage phenotype and inhibits peritoneal metastasis. Oncogene 2015, 34, 1563–1574. [Google Scholar] [CrossRef] [PubMed]
  159. Warburg, O. On respiratory impairment in cancer cells. Science 1956, 124, 269–270. [Google Scholar] [CrossRef] [PubMed]
  160. Allen, B.G.; Bhatia, S.K.; Anderson, C.M.; Eichenberger-Gilmore, J.M.; Sibenaller, Z.A.; Mapuskar, K.A.; Schoenfeld, J.D.; Buatti, J.M.; Spitz, D.R.; Fath, M.A. Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biol. 2014, 2, 963–970. [Google Scholar] [CrossRef] [PubMed]
  161. Weber, D.D.; Aminzadeh-Gohari, S.; Tulipan, J.; Catalano, L.; Feichtinger, R.G.; Kofler, B. Ketogenic diet in the treatment of cancer—Where do we stand? Mol. Metab. 2020, 33, 102–121. [Google Scholar] [CrossRef]
  162. Zhang, N.; Liu, C.; Jin, L.; Zhang, R.; Wang, T.; Wang, Q.; Chen, J.; Yang, F.; Siebert, H.-C.; Zheng, X. Ketogenic Diet Elicits Antitumor Properties through Inducing Oxidative Stress, Inhibiting MMP-9 Expression, and Rebalancing M1/M2 Tumor-Associated Macrophage Phenotype in a Mouse Model of Colon Cancer. J. Agric. Food Chem. 2020, 68, 11182–11196. [Google Scholar] [CrossRef]
  163. Park, S.K.; Lin, H.L.; Murphy, S. Nitric oxide regulates nitric oxide synthase-2 gene expression by inhibiting NF-kappaB binding to DNA. Biochem. J. 1997, 322, 609–613. [Google Scholar] [CrossRef]
  164. Eberhardt, W.; Beeg, T.; Beck, K.F.; Walpen, S.; Gauer, S.; Böhles, H.; Pfeilschifter, J. Nitric oxide modulates expression of matrix metalloproteinase-9 in rat mesangial cells. Kidney Int. 2000, 57, 59–69. [Google Scholar] [CrossRef]
  165. Chen, H.-H.; Wang, D.L. Nitric oxide inhibits matrix metalloproteinase-2 expression via the induction of activating transcription factor 3 in endothelial cells. Mol. Pharmacol. 2004, 65, 1130–1140. [Google Scholar] [CrossRef]
  166. Ridnour, L.A.; Windhausen, A.N.; Isenberg, J.S.; Yeung, N.; Thomas, D.D.; Vitek, M.P.; Roberts, D.D.; Wink, D.A. Nitric oxide regulates matrix metalloproteinase-9 activity by guanylyl-cyclase-dependent and -independent pathways. Proc. Natl. Acad. Sci. USA 2007, 104, 16898–16903. [Google Scholar] [CrossRef]
  167. O’Sullivan, S.; Medina, C.; Ledwidge, M.; Radomski, M.W.; Gilmer, J.F. Nitric oxide-matrix metaloproteinase-9 interactions: Biological and pharmacological significance: NO and MMP-9 interactions. Biochim. Biophys. Acta 2014, 1843, 603–617. [Google Scholar] [CrossRef]
  168. Koodie, L.; Ramakrishnan, S.; Roy, S. Morphine Suppresses Tumor Angiogenesis through a HIF-1α/p38MAPK Pathway. Am. J. Pathol. 2010, 177, 984–997. [Google Scholar] [CrossRef]
  169. Gach, K.; Wyrębska, A.; Fichna, J.; Janecka, A. The role of morphine in regulation of cancer cell growth. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2011, 384, 221–230. [Google Scholar] [CrossRef]
  170. Afsharimani, B.; Baran, J.; Watanabe, S.; Lindner, D.; Cabot, P.J.; Parat, M.-O. Morphine and breast tumor metastasis: The role of matrix-degrading enzymes. Clin. Exp. Metastasis 2014, 31, 149–158. [Google Scholar] [CrossRef]
  171. Koodie, L.; Yuan, H.; Pumper, J.A.; Yu, H.; Charboneau, R.; Ramkrishnan, S.; Roy, S. Morphine inhibits migration of tumor-infiltrating leukocytes and suppresses angiogenesis associated with tumor growth in mice. Am. J. Pathol. 2014, 184, 1073–1084. [Google Scholar] [CrossRef]
  172. Harimaya, Y.; Koizumi, K.; Andoh, T.; Nojima, H.; Kuraishi, Y.; Saiki, I. Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells. Cancer Lett. 2002, 187, 121–127. [Google Scholar] [CrossRef]
  173. Gach, K.; Wyrebska, A.; Szemraj, J.; Janecka, A. The influence of opioid peptides on matrix metalloproteinase-9 and urokinase plasminogen activator expression in three cancer cell lines. Mol. Biol. 2012, 46, 894–899. [Google Scholar] [CrossRef]
  174. Bimonte, S.; Barbieri, A.; Palma, G.; Arra, C. The role of morphine in animal models of human cancer: Does morphine promote or inhibit the tumor growth? Biomed. Res. Int. 2013, 2013, 258141. [Google Scholar] [CrossRef]
  175. Roy, S.; Cain, K.J.; Chapin, R.B.; Charboneau, R.G.; Barke, R.A. Morphine Modulates NFκB Activation in Macrophages. Biochem. Biophys. Res. Commun. 1998, 245, 392–396. [Google Scholar] [CrossRef]
  176. Khabbazi, S.; Hassanshahi, M.; Hassanshahi, A.; Peymanfar, Y.; Su, Y.-W.; Xian, C.J. Opioids and matrix metalloproteinases: The influence of morphine on MMP-9 production and cancer progression. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 123–133. [Google Scholar] [CrossRef]
  177. Sawada, M.; Ichinose, M.; Stefano, G.B. Nitric oxide inhibits the dopamine-induced K+ current via guanylate cyclase in Aplysia neurons. J. Neurosci. Res. 1997, 50, 450–456. [Google Scholar] [CrossRef]
  178. Fimiani, C.; Arcuri, E.; Santoni, A.; Rialas, C.M.; Bilfinger, T.V.; Peter, D.; Salzet, B.; Stefano, G.B. Mu3 opiate receptor expression in lung and lung carcinoma: Ligand binding and coupling to nitric oxide release. Cancer Lett. 1999, 146, 45–51. [Google Scholar] [CrossRef] [PubMed]
  179. Welters, I.D.; Menzebach, A.; Goumon, Y.; Cadet, P.; Menges, T.; Hughes, T.K.; Hempelmann, G.; Stefano, G.B. Morphine inhibits NF-kappaB nuclear binding in human neutrophils and monocytes by a nitric oxide-dependent mechanism. Anesthesiology 2000, 92, 1677–1684. [Google Scholar] [CrossRef] [PubMed]
  180. Stefano, G.B.; Kream, R.M.; Mantione, K.J.; Sheehan, M.; Cadet, P.; Zhu, W.; Bilfinger, T.V.; Esch, T. Endogenous morphine/nitric oxide-coupled regulation of cellular physiology and gene expression: Implications for cancer biology. Semin. Cancer Biol. 2008, 18, 199–210. [Google Scholar] [CrossRef] [PubMed]
  181. Gach, K.; Szemraj, J.; Wyrębska, A.; Janecka, A. The influence of opioids on matrix metalloproteinase-2 and-9 secretion and mRNA levels in MCF-7 breast cancer cell line. Mol. Biol. Rep. 2011, 38, 1231–1236. [Google Scholar] [CrossRef]
  182. Lee, Y.J.; Suh, S.-Y.; Song, J.; Lee, S.S.; Seo, A.-R.; Ahn, H.-Y.; Lee, M.A.; Kim, C.-M.; Klepstad, P. Serum and urine concentrations of morphine and morphine metabolites in patients with advanced cancer receiving continuous intravenous morphine: An observational study. BMC Palliat. Care 2015, 14, 53. [Google Scholar] [CrossRef]
  183. Kristensen, L.S.; Nielsen, H.M.; Hansen, L.L. Epigenetics and cancer treatment. Eur. J. Pharmacol. 2009, 625, 131–142. [Google Scholar] [CrossRef]
  184. Sung, B.; Pandey, M.K.; Ahn, K.S.; Yi, T.; Chaturvedi, M.M.; Liu, M.; Aggarwal, B.B. Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-kappaB-regulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-kappaBalpha kinase, leading to potentiation of apoptosis. Blood 2008, 111, 4880–4891. [Google Scholar] [CrossRef]
  185. Tan, J.; Chen, B.; He, L.; Tang, Y.; Jiang, Z.; Yin, G.; Wang, J.; Jiang, X. Anacardic acid (6-pentadecylsalicylic acid) induces apoptosis of prostate cancer cells through inhibition of androgen receptor and activation of p53 signaling. Chin. J. Cancer Res. 2012, 24, 275–283. [Google Scholar] [CrossRef]
  186. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  187. Ambros, V. The functions of animal microRNAs. Nature 2004, 431, 350–355. [Google Scholar] [CrossRef]
  188. Coolen, M.; Katz, S.; Bally-Cuif, L. miR-9: A versatile regulator of neurogenesis. Front. Cell. Neurosci. 2013, 7, 220. [Google Scholar] [CrossRef]
  189. Fuziwara, C.S.; Kimura, E.T. Insights into Regulation of the miR-17-92 Cluster of miRNAs in Cancer. Front. Med. 2015, 2, 64. [Google Scholar] [CrossRef]
  190. Pfeffer, S.R.; Yang, C.H.; Pfeffer, L.M. The Role of miR-21 in Cancer. Drug Dev. Res. 2015, 76, 270–277. [Google Scholar] [CrossRef]
  191. Sheedy, P.; Medarova, Z. The fundamental role of miR-10b in metastatic cancer. Am. J. Cancer Res. 2018, 8, 1674–1688. [Google Scholar]
  192. Li, X.; Gao, L.; Cui, Q.; Gary, B.D.; Dyess, D.L.; Taylor, W.; Shevde, L.A.; Samant, R.S.; Dean-Colomb, W.; Piazza, G.A.; et al. Sulindac inhibits tumor cell invasion by suppressing NF-κB-mediated transcription of microRNAs. Oncogene 2012, 31, 4979–4986. [Google Scholar] [CrossRef]
  193. van der Donk, W.A.; Tsai, A.-L.; Kulmacz, R.J. The cyclooxygenase reaction mechanism. Biochemistry 2002, 41, 15451–15458. [Google Scholar] [CrossRef]
  194. Hata, A.N.; Breyer, R.M. Pharmacology and signaling of prostaglandin receptors: Multiple roles in inflammation and immune modulation. Pharmacol. Ther. 2004, 103, 147–166. [Google Scholar] [CrossRef]
  195. Rolland, P.H.; Martin, P.M.; Jacquemier, J.; Rolland, A.M.; Toga, M. Prostaglandin in human breast cancer: Evidence suggesting that an elevated prostaglandin production is a marker of high metastatic potential for neoplastic cells. J. Natl. Cancer Inst. 1980, 64, 1061–1070. [Google Scholar]
  196. Kargman, S.L.; O’Neill, G.P.; Vickers, P.J.; Evans, J.F.; Mancini, J.A.; Jothy, S. Expression of prostaglandin G/H synthase-1 and -2 protein in human colon cancer. Cancer Res. 1995, 55, 2556–2559. [Google Scholar]
  197. Surh, Y.-J.; Chun, K.-S.; Cha, H.-H.; Han, S.S.; Keum, Y.-S.; Park, K.-K.; Lee, S.S. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: Down-regulation of COX-2 and iNOS through suppression of NF-κB activation. Mutat. Res. /Fundam. Mol. Mech. Mutagen. 2001, 480, 243–268. [Google Scholar] [CrossRef]
  198. Kolev, Y.; Uetake, H.; Iida, S.; Ishikawa, T.; Kawano, T.; Sugihara, K. Prognostic significance of VEGF expression in correlation with COX-2, microvessel density, and clinicopathological characteristics in human gastric carcinoma. Ann. Surg. Oncol. 2007, 14, 2738–2747. [Google Scholar] [CrossRef] [PubMed]
  199. Bu, X.; Zhao, C.; Dai, X. Involvement of COX-2/PGE(2) Pathway in the Upregulation of MMP-9 Expression in Pancreatic Cancer. Gastroenterol. Res. Pract. 2011, 2011, 214269. [Google Scholar] [CrossRef] [PubMed]
  200. Tsujii, M.; Kawano, S.; Tsuji, S.; Sawaoka, H.; Hori, M.; DuBois, R.N. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 1998, 93, 705–716. [Google Scholar] [CrossRef] [PubMed]
  201. Larkins, T.L.; Nowell, M.; Singh, S.; Sanford, G.L. Inhibition of cyclooxygenase-2 decreases breast cancer cell motility, invasion and matrix metalloproteinase expression. BMC Cancer 2006, 6, 181. [Google Scholar] [CrossRef] [PubMed]
  202. Koontongkaew, S. The Tumor Microenvironment Contribution to Development, Growth, Invasion and Metastasis of Head and Neck Squamous Cell Carcinomas. J. Cancer 2013, 4, 66–83. [Google Scholar] [CrossRef]
  203. Alfonso, L.; Ai, G.; Spitale, R.C.; Bhat, G.J. Molecular targets of aspirin and cancer prevention. Br. J. Cancer 2014, 111, 61–67. [Google Scholar] [CrossRef]
  204. Liao, X.; Lochhead, P.; Nishihara, R.; Morikawa, T.; Kuchiba, A.; Yamauchi, M.; Imamura, Y.; Qian, Z.R.; Baba, Y.; Shima, K. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N. Engl. J. Med. 2012, 367, 1596–1606. [Google Scholar] [CrossRef]
  205. Cheng, C.-Y.; Hsieh, H.-L.; Hsiao, L.-D.; Yang, C.-M. PI3-K/Akt/JNK/NF-κB is essential for MMP-9 expression and outgrowth in human limbal epithelial cells on intact amniotic membrane. Stem Cell Res. 2012, 9, 9–23. [Google Scholar] [CrossRef]
  206. Xue, J.; Hua, Y.N.; Xie, M.L.; Gu, Z.L. Aspirin inhibits MMP-9 mRNA expression and release via the PPARα/γ and COX-2/mPGES-1-mediated pathways in macrophages derived from THP-1 cells. Biomed. Pharmacother. 2010, 64, 118–123. [Google Scholar] [CrossRef]
  207. Wan Mohd Tajuddin, W.N.B.; Lajis, N.H.; Abas, F.; Othman, I.; Naidu, R. Mechanistic Understanding of Curcumin’s Therapeutic Effects in Lung Cancer. Nutrients 2019, 11, 2989. [Google Scholar] [CrossRef]
  208. Lev-Ari, S.; Starr, A.; Vexler, A.; Karaush, V.; Loew, V.; Greif, J.; Fenig, E.; Aderka, D.; Ben-Yosef, R. Inhibition of pancreatic and lung adenocarcinoma cell survival by curcumin is associated with increased apoptosis, down-regulation of COX-2 and EGFR and inhibition of Erk1/2 activity. Anticancer Res. 2006, 26, 4423–4430. [Google Scholar]
  209. Jobin, C.; Bradham, C.A.; Russo, M.P.; Juma, B.; Narula, A.S.; Brenner, D.A.; Sartor, R.B. Curcumin blocks cytokine-mediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity. J. Immunol. 1999, 163, 3474–3483. [Google Scholar] [CrossRef]
  210. Lin, S.-S.; Lai, K.-C.; Hsu, S.-C.; Yang, J.-S.; Kuo, C.-L.; Lin, J.-P.; Ma, Y.-S.; Wu, C.-C.; Chung, J.-G. Curcumin inhibits the migration and invasion of human A549 lung cancer cells through the inhibition of matrix metalloproteinase-2 and -9 and Vascular Endothelial Growth Factor (VEGF). Cancer Lett. 2009, 285, 127–133. [Google Scholar] [CrossRef]
  211. Lee, J.; Pilch, P.F. The insulin receptor: Structure, function, and signaling. Am. J. Physiol. 1994, 266, C319–C334. [Google Scholar] [CrossRef]
  212. Pilch, P.F.; Czech, M.P. The subunit structure of the high affinity insulin receptor. Evidence for a disulfide-linked receptor complex in fat cell and liver plasma membranes. J. Biol. Chem. 1980, 255, 1722–1731. [Google Scholar] [CrossRef]
  213. Ullrich, A.; Bell, J.R.; Chen, E.Y.; Herrera, R.; Petruzzelli, L.M.; Dull, T.J.; Gray, A.; Coussens, L.; Liao, Y.C.; Tsubokawa, M. Human insulin receptor and its relationship to the tyrosine kinase family of oncogenes. Nature 1985, 313, 756–761. [Google Scholar] [CrossRef]
  214. Ebina, Y.; Ellis, L.; Jarnagin, K.; Edery, M.; Graf, L.; Clauser, E.; Ou, J.H.; Masiarz, F.; Kan, Y.W.; Goldfine, I.D. The human insulin receptor cDNA: The structural basis for hormone-activated transmembrane signalling. Cell 1985, 40, 747–758. [Google Scholar] [CrossRef]
  215. Boucher, J.; Kleinridders, A.; Kahn, C.R. Insulin Receptor Signaling in Normal and Insulin-Resistant States. Cold Spring Harb. Perspect. Biol. 2014, 6, a009191. [Google Scholar] [CrossRef]
  216. De Meyts, P. The Insulin Receptor and Its Signal Transduction Network. In Endotext; Feingold, K.R., Anawalt, B., Boyce, A., Chrousos, G., de Herder, W.W., Dhatariya, K., Dungan, K., Hershman, J.M., Hofland, J., Kalra, S., et al., Eds.; MDText: South Dartmouth, MA, USA, 2000. [Google Scholar]
  217. Tokarz, V.L.; MacDonald, P.E.; Klip, A. The cell biology of systemic insulin function. J. Cell Biol. 2018, 217, 2273–2289. [Google Scholar] [CrossRef]
  218. Cefalu, W.T. Insulin resistance: Cellular and clinical concepts. Exp. Biol. Med. 2001, 226, 13–26. [Google Scholar] [CrossRef]
  219. Reaven, G. The metabolic syndrome or the insulin resistance syndrome? Different names, different concepts, and different goals. Endocrinol. Metab. Clin. N. Am. 2004, 33, 283–303. [Google Scholar] [CrossRef] [PubMed]
  220. Wheatcroft, S.B.; Williams, I.L.; Shah, A.M.; Kearney, M.T. Pathophysiological implications of insulin resistance on vascular endothelial function. Diabet. Med. 2003, 20, 255–268. [Google Scholar] [CrossRef] [PubMed]
  221. Blaise, S.; Romier, B.; Kawecki, C.; Ghirardi, M.; Rabenoelina, F.; Baud, S.; Duca, L.; Maurice, P.; Heinz, A.; Schmelzer, C.E.H.; et al. Elastin-derived peptides are new regulators of insulin resistance development in mice. Diabetes 2013, 62, 3807–3816. [Google Scholar] [CrossRef] [PubMed]
  222. Dridi, L.; Seyrantepe, V.; Fougerat, A.; Pan, X.; Bonneil, E.; Thibault, P.; Moreau, A.; Mitchell, G.A.; Heveker, N.; Cairo, C.W.; et al. Positive regulation of insulin signaling by neuraminidase 1. Diabetes 2013, 62, 2338–2346. [Google Scholar] [CrossRef]
  223. Schlein, M.; Havelund, S.; Kristensen, C.; Dunn, M.F.; Kaarsholm, N.C. Ligand-induced conformational change in the minimized insulin receptor. J. Mol. Biol. 2000, 303, 161–169. [Google Scholar] [CrossRef]
  224. Rozengurt, E. Mitogenic signaling pathways induced by G protein-coupled receptors. J. Cell Physiol. 2007, 213, 589–602. [Google Scholar] [CrossRef]
  225. Isami, S.; Kishikawa, H.; Araki, E.; Uehara, M.; Kaneko, K.; Shirotani, T.; Todaka, M.; Ura, S.; Motoyoshi, S.; Matsumoto, K.; et al. Bradykinin enhances GLUT4 translocation through the increase of insulin receptor tyrosine kinase in primary adipocytes: Evidence that bradykinin stimulates the insulin signalling pathway. Diabetologia 1996, 39, 412–420. [Google Scholar] [CrossRef]
  226. Pollak, M.N.; Schernhammer, E.S.; Hankinson, S.E. Insulin-like growth factors and neoplasia. Nat. Rev. Cancer 2004, 4, 505–518. [Google Scholar] [CrossRef]
  227. Pollak, M. Insulin and insulin-like growth factor signalling in neoplasia. Nat. Rev. Cancer 2008, 8, 915–928. [Google Scholar] [CrossRef]
  228. Kasuya, J.; Paz, I.B.; Maddux, B.A.; Goldfine, I.D.; Hefta, S.A.; Fujita-Yamaguchi, Y. Characterization of human placental insulin-like growth factor-I/insulin hybrid receptors by protein microsequencing and purification. Biochemistry 1993, 32, 13531–13536. [Google Scholar] [CrossRef]
  229. Belfiore, A. The role of insulin receptor isoforms and hybrid insulin/IGF-I receptors in human cancer. Curr. Pharm. Des. 2007, 13, 671–686. [Google Scholar] [CrossRef]
  230. Benyoucef, S.; Surinya, K.H.; Hadaschik, D.; Siddle, K. Characterization of insulin/IGF hybrid receptors: Contributions of the insulin receptor L2 and Fn1 domains and the alternatively spliced exon 11 sequence to ligand binding and receptor activation. Biochem. J. 2007, 403, 603–613. [Google Scholar] [CrossRef]
  231. Belfiore, A.; Frasca, F.; Pandini, G.; Sciacca, L.; Vigneri, R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr. Rev. 2009, 30, 586–623. [Google Scholar] [CrossRef]
  232. Bailyes, E.M.; Navé, B.T.; Soos, M.A.; Orr, S.R.; Hayward, A.C.; Siddle, K. Insulin receptor/IGF-I receptor hybrids are widely distributed in mammalian tissues: Quantification of individual receptor species by selective immunoprecipitation and immunoblotting. Biochem. J. 1997, 327, 209–215. [Google Scholar] [CrossRef]
  233. White, M.F. The IRS-signalling system: A network of docking proteins that mediate insulin action. Mol. Cell. Biochem. 1998, 182, 3–11. [Google Scholar] [CrossRef]
  234. Milazzo, G.; Giorgino, F.; Damante, G.; Sung, C.; Stampfer, M.R.; Vigneri, R.; Goldfine, I.D.; Belfiore, A. Insulin receptor expression and function in human breast cancer cell lines. Cancer Res. 1992, 52, 3924–3930. [Google Scholar]
  235. Rozengurt, E.; Sinnett-Smith, J.; Kisfalvi, K. Crosstalk between Insulin/IGF-1 and GPCR Signaling Systems: A Novel Target for the Anti-diabetic Drug Metformin in Pancreatic Cancer. Clin. Cancer Res. 2010, 16, 2505–2511. [Google Scholar] [CrossRef]
  236. Kisfalvi, K.; Rey, O.; Young, S.H.; Sinnett-Smith, J.; Rozengurt, E. Insulin potentiates Ca2+ signaling and phosphatidylinositol 4,5-bisphosphate hydrolysis induced by Gq protein-coupled receptor agonists through an mTOR-dependent pathway. Endocrinology 2007, 148, 3246–3257. [Google Scholar] [CrossRef]
  237. Kisfalvi, K.; Eibl, G.; Sinnett-Smith, J.; Rozengurt, E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 2009, 69, 6539–6545. [Google Scholar] [CrossRef]
  238. Mulvihill, M.J.; Cooke, A.; Rosenfeld-Franklin, M.; Buck, E.; Foreman, K.; Landfair, D.; O’Connor, M.; Pirritt, C.; Sun, Y.; Yao, Y.; et al. Discovery of OSI-906: A selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Med. Chem. 2009, 1, 1153–1171. [Google Scholar] [CrossRef]
  239. Haeusler, R.A.; McGraw, T.E.; Accili, D. Biochemical and cellular properties of insulin receptor signalling. Nat. Rev. Mol. Cell Biol. 2018, 19, 31–44. [Google Scholar] [CrossRef] [PubMed]
  240. Awasthi, N.; Zhang, C.; Ruan, W.; Schwarz, M.A.; Schwarz, R.E. BMS-754807, a small-molecule inhibitor of insulin-like growth factor-1 receptor/insulin receptor, enhances gemcitabine response in pancreatic cancer. Mol. Cancer Ther. 2012, 11, 2644–2653. [Google Scholar] [CrossRef] [PubMed]
  241. Stratikopoulos, E.E.; Dendy, M.; Szabolcs, M.; Khaykin, A.J.; Lefebvre, C.; Zhou, M.-M.; Parsons, R. Kinase and BET Inhibitors Together Clamp Inhibition of PI3K Signaling and Overcome Resistance to Therapy. Cancer Cell 2015, 27, 837–851. [Google Scholar] [CrossRef] [PubMed]
  242. Okada, T.; Kawano, Y.; Sakakibara, T.; Hazeki, O.; Ui, M. Essential role of phosphatidylinositol 3-kinase in insulin-induced glucose transport and antilipolysis in rat adipocytes. Studies with a selective inhibitor wortmannin. J. Biol. Chem. 1994, 269, 3568–3573. [Google Scholar] [CrossRef]
  243. Robey, R.B.; Hay, N. Is Akt the “Warburg kinase”?-Akt-energy metabolism interactions and oncogenesis. Semin. Cancer Biol. 2009, 19, 25–31. [Google Scholar] [CrossRef]
  244. White, M.F. IRS proteins and the common path to diabetes. Am. J. Physiol. Endocrinol. Metab. 2002, 283, E413–E422. [Google Scholar] [CrossRef]
  245. Dong, X.; Park, S.; Lin, X.; Copps, K.; Yi, X.; White, M.F. Irs1 and Irs2 signaling is essential for hepatic glucose homeostasis and systemic growth. J. Clin. Investig. 2006, 116, 101–114. [Google Scholar] [CrossRef]
  246. Shaw, L.M. The insulin receptor substrate (IRS) proteins: At the intersection of metabolism and cancer. Cell Cycle 2011, 10, 1750–1756. [Google Scholar] [CrossRef]
  247. Zhao, J.; Li, Z.; Chen, Y.; Zhang, S.; Guo, L.; Gao, B.; Jiang, Y.; Tian, W.; Hao, S.; Zhang, X. MicroRNA-766 inhibits papillary thyroid cancer progression by directly targeting insulin receptor substrate 2 and regulating the PI3K/Akt pathway. Int. J. Oncol. 2019, 54, 315–325. [Google Scholar] [CrossRef]
  248. Xu, L.; Tan, Y.; Xu, F.; Zhang, Y. Long noncoding RNA ADIRF antisense RNA 1 upregulates insulin receptor substrate 1 to decrease the aggressiveness of osteosarcoma by sponging microRNA-761. Bioengineered 2022, 13, 2028–2043. [Google Scholar] [CrossRef]
  249. Amar, L.; Benoit, C.; Beaumont, G.; Vacher, C.M.; Crepin, D.; Taouis, M.; Baroin-Tourancheau, A. MicroRNA expression profiling of hypothalamic arcuate and paraventricular nuclei from single rats using Illumina sequencing technology. J. Neurosci. Methods 2012, 209, 134–143. [Google Scholar] [CrossRef]
  250. Eguchi, T.; Watanabe, K.; Hara, E.S.; Ono, M.; Kuboki, T.; Calderwood, S.K. OstemiR: A Novel Panel of MicroRNA Biomarkers in Osteoblastic and Osteocytic Differentiation from Mesencymal Stem Cells. PLoS ONE 2013, 8, e58796. [Google Scholar] [CrossRef]
  251. Liu, M.-M.; Li, Z.; Han, X.-D.; Shi, J.-H.; Tu, D.-Y.; Song, W.; Zhang, J.; Qiu, X.-L.; Ren, Y.; Zhen, L.-L. MiR-30e inhibits tumor growth and chemoresistance via targeting IRS1 in Breast Cancer. Sci. Rep. 2017, 7, 15929. [Google Scholar] [CrossRef]
  252. Garofalo, C.; Capristo, M.; Mancarella, C.; Reunevi, H.; Picci, P.; Scotlandi, K. Preclinical Effectiveness of Selective Inhibitor of IRS-1/2 NT157 in Osteosarcoma Cell Lines. Front. Endocrinol. 2015, 6, 74. [Google Scholar] [CrossRef]
Figure 1. Bradykinin (BR2) and angiotensin II receptor type I (AT2R) are tethered within a multimeric receptor IRβ, receptor (TLR) transactivation-signaling axis, mediated by Neu1 sialidase and the glycosylation modification of TLRs. CB1 cannabinoid receptor with modeled N-terminus and the locations of orthosteric and allosteric binding sites. Cannabidiol (CBD)—An allosteric ligand, and Δ9-tetrahydrocannabinol (Δ9-THC)—an orthosteric ligand of CB1R. According to Lapraire et al., [96] and Sabatucci et al., [97], CBD ligands very likely bind to the allosteric site 1. The biased GPCR-signaling platform here potentiates Neu1 and MMP-9 cross-talk on the cell surface, which is essential for the transactivation of TLRs and subsequent NFκB cellular signaling and inducing epigenetic rewiring. Notes: TLR ligand, as well as GPCR agonists, can potentiate biased NMBR-TLR signaling and subsequently induce MMP-9 activation and Neu1 sialidase activity. Activated MMP-9 is proposed here to remove the EBP as part of the molecular multienzymatic complex that contains β-galactosidase/Neu1 and PPCA. Activated Neu1 then hydrolyzes α-2,3 sialyl residues of TLR at the ectodomain to remove steric hindrance to facilitate TLR association and subsequent recruitment of MyD88 and downstream signaling. Citation: Taken in part from Qorri et al., (2018) [98], Jakowiecki et al., (2021) [99], (The article is open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/ (accessed on 23 April 2021)) and Reber et al., (2009) [100]. (This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited).
Figure 1. Bradykinin (BR2) and angiotensin II receptor type I (AT2R) are tethered within a multimeric receptor IRβ, receptor (TLR) transactivation-signaling axis, mediated by Neu1 sialidase and the glycosylation modification of TLRs. CB1 cannabinoid receptor with modeled N-terminus and the locations of orthosteric and allosteric binding sites. Cannabidiol (CBD)—An allosteric ligand, and Δ9-tetrahydrocannabinol (Δ9-THC)—an orthosteric ligand of CB1R. According to Lapraire et al., [96] and Sabatucci et al., [97], CBD ligands very likely bind to the allosteric site 1. The biased GPCR-signaling platform here potentiates Neu1 and MMP-9 cross-talk on the cell surface, which is essential for the transactivation of TLRs and subsequent NFκB cellular signaling and inducing epigenetic rewiring. Notes: TLR ligand, as well as GPCR agonists, can potentiate biased NMBR-TLR signaling and subsequently induce MMP-9 activation and Neu1 sialidase activity. Activated MMP-9 is proposed here to remove the EBP as part of the molecular multienzymatic complex that contains β-galactosidase/Neu1 and PPCA. Activated Neu1 then hydrolyzes α-2,3 sialyl residues of TLR at the ectodomain to remove steric hindrance to facilitate TLR association and subsequent recruitment of MyD88 and downstream signaling. Citation: Taken in part from Qorri et al., (2018) [98], Jakowiecki et al., (2021) [99], (The article is open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/ (accessed on 23 April 2021)) and Reber et al., (2009) [100]. (This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited).
Cancers 15 01030 g001
Figure 2. Angiotensin II receptor type I (AT2R) forms a multimeric receptor complex with neuromedin B GPCR (NMBR), insulin receptor β subunit (IRβ), and neuraminidase-1 (Neu1) in naïve (unstimulated) and stimulated HTC-IR cells [105]. Here, the interaction and signaling mechanism(s) of these molecules uncover a biased GPCR agonist-induced IRβ trans-activation signaling axis, mediated by Neu1 sialidase with the modification of insulin receptor glycosylation. The biased G protein-coupled receptor (GPCR)-signaling platform potentiates Neu1 and matrix metalloproteinase-9 (MMP-9) cell surface cross-talk that is essential for the activation of the IRβ tyrosine kinases. Notes: Insulin-binding receptor α subunits (IRα) as well GPCR agonists potentiate a biased NMBR-IRβ signaling and MMP-9 activation to induce Neu1 sialidase. Activated MMP-9 is proposed to remove the elastin-binding protein (EBP) as part of the molecular multi-enzymatic complex that contains β-galactosidase/Neu1 and protective protein cathepsin A (PPCA). Activated Neu1 hydrolyzes α-2,3 sialyl residues of IRβ to remove steric hindrance to facilitate IRβ subunits association and tyrosine kinase activation. Activated phospho-IRβ subunits phosphorylate insulin receptor substrate-1 (pIRS1), which initiates intracellular insulin signaling via the Ras-MAPK and the PI3K-Akt pathway, among others, such as NFκB. Abbreviations: PI3K: phosphatidylinositol 3-kinase; GTP: guanine triphosphate; IRS1: insulin receptor substrate-1; p: phosphorylation. Citation: Taken in part from Haxho et al., (2018) [105], Alghamdi et al., (2014) [112], Bordoni et al., (2019) [113], 2019 Elsevier B.V. and Société Française de Biochimie et Biologie Moléculaire (SFBBM). Open access under CC BY-NC-ND license. This is an Open Access article which permits unrestricted non-commercial use, provided the original work be properly cited.
Figure 2. Angiotensin II receptor type I (AT2R) forms a multimeric receptor complex with neuromedin B GPCR (NMBR), insulin receptor β subunit (IRβ), and neuraminidase-1 (Neu1) in naïve (unstimulated) and stimulated HTC-IR cells [105]. Here, the interaction and signaling mechanism(s) of these molecules uncover a biased GPCR agonist-induced IRβ trans-activation signaling axis, mediated by Neu1 sialidase with the modification of insulin receptor glycosylation. The biased G protein-coupled receptor (GPCR)-signaling platform potentiates Neu1 and matrix metalloproteinase-9 (MMP-9) cell surface cross-talk that is essential for the activation of the IRβ tyrosine kinases. Notes: Insulin-binding receptor α subunits (IRα) as well GPCR agonists potentiate a biased NMBR-IRβ signaling and MMP-9 activation to induce Neu1 sialidase. Activated MMP-9 is proposed to remove the elastin-binding protein (EBP) as part of the molecular multi-enzymatic complex that contains β-galactosidase/Neu1 and protective protein cathepsin A (PPCA). Activated Neu1 hydrolyzes α-2,3 sialyl residues of IRβ to remove steric hindrance to facilitate IRβ subunits association and tyrosine kinase activation. Activated phospho-IRβ subunits phosphorylate insulin receptor substrate-1 (pIRS1), which initiates intracellular insulin signaling via the Ras-MAPK and the PI3K-Akt pathway, among others, such as NFκB. Abbreviations: PI3K: phosphatidylinositol 3-kinase; GTP: guanine triphosphate; IRS1: insulin receptor substrate-1; p: phosphorylation. Citation: Taken in part from Haxho et al., (2018) [105], Alghamdi et al., (2014) [112], Bordoni et al., (2019) [113], 2019 Elsevier B.V. and Société Française de Biochimie et Biologie Moléculaire (SFBBM). Open access under CC BY-NC-ND license. This is an Open Access article which permits unrestricted non-commercial use, provided the original work be properly cited.
Cancers 15 01030 g002
Figure 3. Neu1-MMP9-GPCR signaling platform in the regulation of RT K and the molecular targeting of multistage tumorigenesis. Neuraminidase-1 (Neu1) and matrix metalloproteinase-9 (MMP9) cross-talk in alliance with G protein-coupled receptor(s) (GPCR) regulates receptor tyrosine kinases (RTKs) and extracellular and intracellular Toll-like (TLR) receptors in cancer cells. This process sets the stage for multistage tumorigenesis. Citation: Taken in part from: Abdulkhalek et al., (2013) [118], Abdulkhalek et al., (2014) [134]. Publisher and licensee Dove Medical Press Ltd. These are an Open Access articles which permits unrestricted non-commercial use, provided the original work is properly cited.
Figure 3. Neu1-MMP9-GPCR signaling platform in the regulation of RT K and the molecular targeting of multistage tumorigenesis. Neuraminidase-1 (Neu1) and matrix metalloproteinase-9 (MMP9) cross-talk in alliance with G protein-coupled receptor(s) (GPCR) regulates receptor tyrosine kinases (RTKs) and extracellular and intracellular Toll-like (TLR) receptors in cancer cells. This process sets the stage for multistage tumorigenesis. Citation: Taken in part from: Abdulkhalek et al., (2013) [118], Abdulkhalek et al., (2014) [134]. Publisher and licensee Dove Medical Press Ltd. These are an Open Access articles which permits unrestricted non-commercial use, provided the original work is properly cited.
Cancers 15 01030 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bunsick, D.A.; Matsukubo, J.; Szewczuk, M.R. Cannabinoids Transmogrify Cancer Metabolic Phenotype via Epigenetic Reprogramming and a Novel CBD Biased G Protein-Coupled Receptor Signaling Platform. Cancers 2023, 15, 1030. https://doi.org/10.3390/cancers15041030

AMA Style

Bunsick DA, Matsukubo J, Szewczuk MR. Cannabinoids Transmogrify Cancer Metabolic Phenotype via Epigenetic Reprogramming and a Novel CBD Biased G Protein-Coupled Receptor Signaling Platform. Cancers. 2023; 15(4):1030. https://doi.org/10.3390/cancers15041030

Chicago/Turabian Style

Bunsick, David A., Jenna Matsukubo, and Myron R. Szewczuk. 2023. "Cannabinoids Transmogrify Cancer Metabolic Phenotype via Epigenetic Reprogramming and a Novel CBD Biased G Protein-Coupled Receptor Signaling Platform" Cancers 15, no. 4: 1030. https://doi.org/10.3390/cancers15041030

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